Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
FEBS Open Bio ; 11(6): 1731-1738, 2021 06.
Article in English | MEDLINE | ID: mdl-33932144

ABSTRACT

Nonreceptor tyrosine kinase c-Abl participates in several cellular processes by phosphorylating transcription factors or cofactors. c-Abl binds and phosphorylates four-and-a-half-LIM-only protein 2 (FHL2), but the identity of the phosphorylation sites and their contribution to cell cycle regulation is unclear. In this study, we demonstrate that c-Abl highly phosphorylates FHL2 at Y97, Y176, Y217, and Y236 through mass spectrometry and tyrosine-to-phenylalanine (Y â†’ F) mutant analysis. Proliferation was inhibited in cells expressing wild-type (WT) FHL2 but not cells expressing the phosphorylation-defective mutant FHL2(4YF). Moreover, FHL2 contributed to cell cycle arrest at G2/M induced by ionizing radiation (IR). FHL2 WT but not FHL2(4YF) rescued FHL2 function in FHL2-depleted cells by causing IR-induced G2/M arrest. These results demonstrate that c-Abl regulates cell cycle progression by phosphorylating FHL2.


Subject(s)
LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Transcription Factors/metabolism , Cell Proliferation , Cells, Cultured , G2 Phase Cell Cycle Checkpoints , Humans , LIM-Homeodomain Proteins/deficiency , Muscle Proteins/deficiency , Phosphorylation , Radiation, Ionizing , Transcription Factors/deficiency
2.
Biol Open ; 10(3)2021 03 08.
Article in English | MEDLINE | ID: mdl-33579692

ABSTRACT

LIM homeobox 9 (Lhx9) is a member of the LIM homeodomain transcription factor family, which expresses and functions in various vertebrate tissues, such as the gonads and pineal gland. Previous studies on lhx9 in zebrafish have mainly focused on the brain. However, little is known about the expression pattern of lhx9 during embryogenesis. Here, we detected lhx9 expression in zebrafish embryos using whole-mount in situ hybridization and found lhx9 expressed in heart, pectoral fin, and retina during their development in zebrafish. We then detailed the expression of lhx9 in retinal development. To further investigate the function of Lhx9 in retinogenesis, we performed morpholino (MO) knockdown experiments and found that upon lhx9 knockdown by MO, larvae presented normal eye development, retinal neural development, differentiation, proliferation, apoptosis, and responses to light stimulus. We not only elaborated the expression pattern of lhx9 in zebrafish embryogenesis, but we also demonstrated that lhx9 knockdown by morpholino does not affect the zebrafish retinal development, and our study provides data for further understanding of the role of Lhx9 in zebrafish retinal development.


Subject(s)
LIM-Homeodomain Proteins/deficiency , Morpholinos/genetics , Organogenesis/genetics , Retina/embryology , Retina/metabolism , Animals , Apoptosis/genetics , Cell Differentiation , Cell Proliferation , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques/methods , Zebrafish
3.
Cell Rep ; 32(11): 108144, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32937137

ABSTRACT

Retinal bipolar cells (BCs) connect with photoreceptors and relay visual information to retinal ganglion cells (RGCs). Retina-specific deletion of Lhx4 in mice results in a visual defect resembling human congenital stationary night blindness. This visual dysfunction results from the absence of rod bipolar cells (RBCs) and the loss of selective rod-connecting cone bipolar cell (CBC) subtypes and AII amacrine cells (ACs). Inactivation of Lhx4 causes the apoptosis of BCs and cell fate switch from some BCs to ACs, whereas Lhx4 overexpression promotes BC genesis. Moreover, Lhx4 positively regulates Lhx3 expression to drive the fate choice of type 2 BCs over the GABAergic ACs. Lhx4 inactivation ablates Bhlhe23 expression, whereas overexpression of Bhlhe23 partially rescues RBC development in the absence of Lhx4. Thus, by acting upstream of Bhlhe23, Prdm8, Fezf2, Lhx3, and other BC genes, Lhx4, together with Isl1, could play essential roles in regulating the subtype-specific development of RBCs and CBCs.


Subject(s)
Cell Differentiation , LIM-Homeodomain Proteins/metabolism , Retinal Bipolar Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Transcription Factors/metabolism , Aging/pathology , Amacrine Cells/metabolism , Amacrine Cells/pathology , Animals , Apoptosis , Electroretinography , LIM-Homeodomain Proteins/deficiency , Mice , Night Vision , Retinal Bipolar Cells/pathology , Retinal Cone Photoreceptor Cells/pathology , Transcription Factors/deficiency , Transcriptome/genetics
4.
J Clin Invest ; 130(8): 4501-4515, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32453714

ABSTRACT

The transcription factor ISL1 is expressed in pituitary gland stem cells and the thyrotrope and gonadotrope lineages. Pituitary-specific Isl1 deletion causes hypopituitarism with increased stem cell apoptosis, reduced differentiation of thyrotropes and gonadotropes, and reduced body size. Conditional Isl1 deletion causes development of multiple Rathke's cleft-like cysts, with 100% penetrance. Foxa1 and Foxj1 are abnormally expressed in the pituitary gland and associated with a ciliogenic gene-expression program in the cysts. We confirmed expression of FOXA1, FOXJ1, and stem cell markers in human Rathke's cleft cyst tissue, but not craniopharyngiomas, which suggests these transcription factors are useful, pathological markers for diagnosis of Rathke's cleft cysts. These studies support a model whereby expression of ISL1 in pituitary progenitors drives differentiation into thyrotropes and gonadotropes and without it, activation of FOXA1 and FOXJ1 permits development of an oral epithelial cell fate with mucinous cysts. This pituitary-specific Isl1 mouse knockout sheds light on the etiology of Rathke's cleft cysts and the role of ISL1 in normal pituitary development.


Subject(s)
Central Nervous System Cysts/metabolism , Gene Deletion , LIM-Homeodomain Proteins/deficiency , Neoplasm Proteins/deficiency , Pituitary Gland/metabolism , Stem Cells/metabolism , Transcription Factors/deficiency , Animals , Central Nervous System Cysts/genetics , Central Nervous System Cysts/pathology , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Pituitary Gland/pathology , Stem Cells/pathology , Transcription Factors/metabolism
5.
Cells ; 9(1)2020 01 19.
Article in English | MEDLINE | ID: mdl-31963815

ABSTRACT

Cholestasis occurs in different clinical circumstances and leads to severe hepatic disorders. The four-and-a-half LIM-domain protein 2 (FHL2) is a scaffolding protein that modulates multiple signal transduction pathways in a tissue- and cell context-specific manner. In this study, we aimed to gain insight into the function of FHL2 in cholestatic liver injury. FHL2 expression was significantly increased in the bile duct ligation (BDL) model in mice. In Fhl2-deficient (Fhl2-ko) mice, BDL caused a more severe portal and parenchymal inflammation, extended portal fibrosis, higher serum transaminase levels, and higher pro-inflammatory and pro-fibrogenic gene expression compared to wild type (wt) mice. FHL2 depletion in HepG2 cells with siRNA resulted in a higher expression of the bile acid transporter Na+-taurocholate cotransporting polypeptide (NTCP) gene. Furthermore, FHL2-depleted HepG2 cells showed higher expression of markers for oxidative stress, lower B-cell lymphoma 2 (Bcl2) expression, and higher Bcl2-associated X protein (BAX) expression after stimulation with deoxycholic acid (DCA). In hepatic stellate cells (HSCs), FHL2 depletion caused an increased expression of TGF-ß and several pro-fibrogenic matrix metalloproteinases. In summary, our study shows that deficiency in FHL2 aggravates cholestatic liver injury and suggests FHL2-mediated effects on bile acid metabolisms and HSCs as potential mechanisms for pronounced hepatocellular injury and fibrosis.


Subject(s)
Cholestasis/metabolism , Cholestasis/pathology , LIM-Homeodomain Proteins/deficiency , Liver/injuries , Muscle Proteins/deficiency , Transcription Factors/deficiency , Animals , Bile Acids and Salts/metabolism , Bile Ducts/pathology , Disease Models, Animal , Gene Expression Regulation , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Inflammation/pathology , LIM-Homeodomain Proteins/metabolism , Ligation , Liver/pathology , Liver Cirrhosis/pathology , Male , Mice, Knockout , Muscle Proteins/metabolism , Transcription Factors/metabolism
6.
Vascul Pharmacol ; 125-126: 106634, 2020.
Article in English | MEDLINE | ID: mdl-31866461

ABSTRACT

Despite the advent of new-generation drug-eluting stents, in-stent restenosis remains a significant problem in patients with coronary artery disease. In- stent restenosis is defined as the gradual re-narrowing of a stented coronary artery lesion due to arterial damage with subsequent local inflammation of the vessel wall and excessive growth of the vascular smooth muscle cells (vSMCs). Four-and-a-half LIM-domain protein 2 (FHL2) is a scaffold protein involved in regulating vSMC function and inflammation. Previously we have demonstrated that FHL2 prevents vSMC proliferation in a murine carotid artery ligation model. However, the effect of FHL2 on the inflammatory response of the vSMCs is not investigated. Therefore, we studied the inflammatory response in the vessel wall of FHL2-deficient (-KO) mice after carotid artery ligation. We found that circulating cytokines and local macrophage infiltration in the ligated carotid vessels were increased in FHL2-KO mice after carotid artery ligation. Moreover, FHL2-KO vSMCs showed increased secretion of cytokines such as SDF-1α and RANTES, and enhanced activation of the NFκB pathway. Finally, we found that blocking the NFκB signalling pathway abrogated this pro-inflammatory state in FHL2-KO vSMCs. Taken together, our results demonstrate that FHL2 decreases the inflammatory response of vSMCs through inhibition of the NFkB-signalling pathway.


Subject(s)
Carotid Artery Diseases/metabolism , Inflammation/metabolism , LIM-Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Transcription Factors/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Artery Diseases/prevention & control , Cells, Cultured , Cytokines/blood , Disease Models, Animal , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics
7.
Elife ; 82019 07 29.
Article in English | MEDLINE | ID: mdl-31355748

ABSTRACT

Formation of long-range axons occurs over multiple stages of morphological maturation. However, the intrinsic transcriptional mechanisms that temporally control different stages of axon projection development are unknown. Here, we addressed this question by studying the formation of mouse serotonin (5-HT) axons, the exemplar of long-range profusely arborized axon architectures. We report that LIM homeodomain factor 1b (Lmx1b)-deficient 5-HT neurons fail to generate axonal projections to the forebrain and spinal cord. Stage-specific targeting demonstrates that Lmx1b is required at successive stages to control 5-HT axon primary outgrowth, selective routing, and terminal arborization. We show a Lmx1b→Pet1 regulatory cascade is temporally required for 5-HT arborization and upregulation of the 5-HT axon arborization gene, Protocadherin-alphac2, during postnatal development of forebrain 5-HT axons. Our findings identify a temporal regulatory mechanism in which a single continuously expressed transcription factor functions at successive stages to orchestrate the progressive development of long-range axon architectures enabling expansive neuromodulation.


Subject(s)
Axons/physiology , LIM-Homeodomain Proteins/metabolism , Serotonergic Neurons/physiology , Transcription Factors/metabolism , Animals , Gene Expression Profiling , LIM-Homeodomain Proteins/deficiency , Mice , Prosencephalon/cytology , Spinal Cord/cytology , Transcription Factors/deficiency
8.
Mech Ageing Dev ; 181: 29-41, 2019 07.
Article in English | MEDLINE | ID: mdl-31158363

ABSTRACT

Understanding the molecular mechanisms underlying variation in lifespan is central to ensure long life. Lim3 encoding a homolog of the vertebrate Lhx3/4 transcription factors plays a key role in Drosophila neuron development. Here, we demonstrated that Lim3 knockdown early in life decreased survival of adult flies. To study the mechanisms underlying this effect, we identified embryonic Lim3 targets using combined RNA-seq and RT-qPCR analyses complemented by in silico analysis of Lim3 binding sites. Though genes with neuronal functions were revealed as Lim3 targets, the characteristics of neurons were not affected by Lim3 depletion. Many of the direct and indirect Lim3 target genes were associated with mitochondrial function, ATP-related activity, redox processes and antioxidant defense. Consistent with the observed changes in the embryonic transcription of these genes, ROS levels were increased in embryos, which could cause changes in the transcription of indirect Lim3 targets known to affect lifespan. We hypothesize that altered mitochondrial activity is crucial for the decrease of adult lifespan caused by Lim3 knockdown early in life. In adults that encountered Lim3 depletion early in life, the transcription of several genes remained altered, and mitochondrial membrane potential, ATP level and locomotion were increased, confirming the existence of carry-over effects.


Subject(s)
Drosophila Proteins/deficiency , Gene Knockdown Techniques , LIM-Homeodomain Proteins/deficiency , Longevity , Mitochondria/metabolism , Transcription Factors/deficiency , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster , LIM-Homeodomain Proteins/metabolism , Larva/genetics , Larva/metabolism , Mitochondria/genetics , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism
9.
Dev Growth Differ ; 61(5): 327-336, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31111476

ABSTRACT

Development of an embryo is driven by a series of molecular instructions that control the differentiation of tissue precursor cells and shape the tissues into major body parts. LIM homeobox 1 (LHX1) is a transcription factor that plays a major role in the development of the embryonic head of the mouse. Loss of LHX1 function disrupts the morphogenetic movement of head tissue precursors and impacts on the function of molecular factors in modulating the activity of the WNT signaling pathway. LHX1 acts with a transcription factor complex to regulate the transcription of target genes in multiple phases of development and in a range of embryonic tissues of the mouse and Xenopus. Determining the interacting factors and transcriptional targets of LHX1 will be key to unraveling the ensemble of factors involved in head development and building a head gene regulatory network.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Regulatory Networks , Head/embryology , LIM-Homeodomain Proteins/metabolism , Animals , Gene Regulatory Networks/genetics , Humans , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
10.
FASEB J ; 33(7): 7799-7809, 2019 07.
Article in English | MEDLINE | ID: mdl-30939249

ABSTRACT

The 4-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein that can interact with cell surface receptors, cytosolic adaptor and structural proteins, kinases, and nuclear transcription factors. It is involved in numerous functional activities, including the epithelial-mesenchymal transition, cell proliferation, apoptosis, adhesion, migration, structural stability, and gene expression. Despite this, FHL2-knockout (KO) mice are viable and fertile with no obvious abnormalities, rather suggesting a high capacity for fine-tuning adjustment and functional redundancy of FHL2. Indeed, challenging FHL2-KO cells or mice provided numerous evidences for the great functional significance of FHL2. In recent years, several reviews have been published describing the high capacity of FHL2 to bind diverse proteins as well as the versatile functions of FHL2, emphasizing in particular its role in cardiovascular diseases and carcinogenesis. Here, we view the function of FHL2 from a different perspective. We summarize the published data demonstrating the impact of FHL2 on wound healing and inflammation. FHL2 seems to be involved in numerous steps of these extremely complex and multidirectional but tightly regulated tissue remodeling processes, supporting tissue repair and coordinating inflammation. Deficiency of FHL2 not only slows down ongoing wound healing but also often turns it into a chronic condition.-Wixler, V. The role of FHL2 in wound healing and inflammation.


Subject(s)
Inflammation/physiopathology , LIM-Homeodomain Proteins/physiology , Muscle Proteins/physiology , Transcription Factors/physiology , Wound Healing/physiology , Animals , Chemotaxis, Leukocyte/physiology , Cytokines/physiology , Epithelial-Mesenchymal Transition/physiology , Inflammation/immunology , LIM-Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Muscle Proteins/biosynthesis , Muscle Proteins/deficiency , Muscle Proteins/genetics , Myofibroblasts/physiology , Regeneration/physiology , Signal Transduction/physiology , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic/physiology , Up-Regulation
11.
Cell Res ; 29(6): 486-501, 2019 06.
Article in English | MEDLINE | ID: mdl-31024170

ABSTRACT

Generation of widely differing and specialized cell types from a single totipotent zygote involves large-scale transcriptional changes and chromatin reorganization. Pioneer transcription factors play key roles in programming the epigenome and facilitating recruitment of additional regulatory factors during successive cell lineage specification and differentiation steps. Here we show that Isl1 acts as a pioneer factor driving cardiomyocyte lineage commitment by shaping the chromatin landscape of cardiac progenitor cells. Using an Isl1 hypomorphic mouse line which shows congenital heart defects, genome-wide profiling of Isl1 binding together with RNA- and ATAC-sequencing of cardiac progenitor cells and their derivatives, we uncover a regulatory network downstream of Isl1 that orchestrates cardiogenesis. Mechanistically, we show that Isl1 binds to compacted chromatin and works in concert with the Brg1-Baf60c-based SWI/SNF complex to promote permissive cardiac lineage-specific alterations in the chromatin landscape not only of genes with critical functions in cardiac progenitor cells, but also of cardiomyocyte structural genes that are highly expressed when Isl1 itself is no longer present. Thus, the Isl1/Brg1-Baf60c complex plays a crucial role in orchestrating proper cardiogenesis and in establishing epigenetic memory of cardiomyocyte fate commitment.


Subject(s)
Epigenesis, Genetic/genetics , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Myocytes, Cardiac/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation , Cells, Cultured , HEK293 Cells , Humans , LIM-Homeodomain Proteins/deficiency , Magnetic Resonance Imaging , Mice , Mice, Knockout , Mice, Transgenic , Transcription Factors/deficiency
12.
Cell Rep ; 22(7): 1710-1721, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29444425

ABSTRACT

Cortical networks are composed of excitatory projection neurons and inhibitory interneurons. Finding the right balance between the two is important for controlling overall cortical excitation and network dynamics. However, it is unclear how the correct number of cortical interneurons (CIs) is established in the mammalian forebrain. CIs are generated in excess from basal forebrain progenitors, and their final numbers are adjusted via an intrinsically determined program of apoptosis that takes place during an early postnatal window. Here, we provide evidence that the extent of CI apoptosis during this critical period is plastic and cell-type specific and can be reduced in a cell-autonomous manner by acute increases in neuronal activity. We propose that the physiological state of the emerging neural network controls the activity levels of local CIs to modulate their numbers in a homeostatic manner.


Subject(s)
Apoptosis , Cerebral Cortex/cytology , Interneurons/cytology , Neural Inhibition , Animals , Cell Count , Cell Lineage , Cell Survival , Cellular Microenvironment , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Median Eminence/cytology , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Up-Regulation/genetics
13.
Neural Dev ; 12(1): 19, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141678

ABSTRACT

Patterning of the telencephalic neuroepithelium is a tightly regulated process controlled by transcription factors and signalling molecules. The cortical primordium is flanked by two signalling centres, the hem medially, and the antihem laterally. The hem induces the formation of the hippocampus in adjacent neuroepithelium. Therefore, the position of the hem defines the position of the hippocampus in the brain. The antihem is positioned at the boundary between the dorsal and ventral telencephalon and proposed to provide patterning cues during development. LIM-homeodomain (LIM-HD) transcription factor LHX2 suppresses both hem and antihem fate in the cortical neuroepithelium. Upon loss of Lhx2, medial cortical neuroepithelium is transformed into hem, whereas lateral cortical neuroepithelium is transformed into antihem. Here, we show that transcription factor PAX6, known to regulate patterning of the lateral telencephalon, restricts this tissue from transforming into hem upon loss of Lhx2. When Lhx2 and Pax6 are both deleted, the cortical hem expands to occupy almost the complete extent of the cortical primordium, indicating that both factors act to suppress hem fate in the lateral telencephalon. Furthermore, the shift in the pallial-subpallial boundary and absence of the antihem, observed in the Pax6 mutant, are both restored in the Lhx2; Pax6 double mutant. Together, these results not only reveal a novel function for LHX2 in regulating dorsoventral patterning in the telencephalon, but also identify PAX6 as a fundamental regulator of where the hem can form, and therefore implicate this molecule as a determinant of hippocampal positioning.


Subject(s)
LIM-Homeodomain Proteins/deficiency , Neurogenesis/physiology , PAX6 Transcription Factor/deficiency , Telencephalon/embryology , Transcription Factors/deficiency , Animals , Mice , Mice, Knockout
14.
Nature ; 548(7669): 582-587, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28847002

ABSTRACT

Multiple populations of wake-promoting neurons have been characterized in mammals, but few sleep-promoting neurons have been identified. Wake-promoting cell types include hypocretin and GABA (γ-aminobutyric-acid)-releasing neurons of the lateral hypothalamus, which promote the transition to wakefulness from non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. Here we show that a subset of GABAergic neurons in the mouse ventral zona incerta, which express the LIM homeodomain factor Lhx6 and are activated by sleep pressure, both directly inhibit wake-active hypocretin and GABAergic cells in the lateral hypothalamus and receive inputs from multiple sleep-wake-regulating neurons. Conditional deletion of Lhx6 from the developing diencephalon leads to decreases in both NREM and REM sleep. Furthermore, selective activation and inhibition of Lhx6-positive neurons in the ventral zona incerta bidirectionally regulate sleep time in adult mice, in part through hypocretin-dependent mechanisms. These studies identify a GABAergic subpopulation of neurons in the ventral zona incerta that promote sleep.


Subject(s)
GABAergic Neurons/metabolism , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Sleep/physiology , Transcription Factors/metabolism , Zona Incerta/cytology , gamma-Aminobutyric Acid/metabolism , Animals , Cell Lineage , GABAergic Neurons/drug effects , Gene Deletion , Hippocampus/cytology , Hippocampus/physiology , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/drug effects , LIM-Homeodomain Proteins/genetics , Male , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Orexins/metabolism , Presynaptic Terminals/metabolism , Sleep/drug effects , Sleep/genetics , Sleep, REM/drug effects , Sleep, REM/genetics , Sleep, REM/physiology , Time Factors , Transcription Factors/deficiency , Transcription Factors/drug effects , Transcription Factors/genetics , Wakefulness/drug effects , Wakefulness/genetics , Wakefulness/physiology , Zona Incerta/drug effects , Zona Incerta/physiology
15.
Proc Natl Acad Sci U S A ; 113(30): E4387-96, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27407143

ABSTRACT

The LIM-homeodomain transcription factors Lmx1a and Lmx1b play critical roles during the development of midbrain dopaminergic progenitors, but their functions in the adult brain remain poorly understood. We show here that sustained expression of Lmx1a and Lmx1b is required for the survival of adult midbrain dopaminergic neurons. Strikingly, inactivation of Lmx1a and Lmx1b recreates cellular features observed in Parkinson's disease. We found that Lmx1a/b control the expression of key genes involved in mitochondrial functions, and their ablation results in impaired respiratory chain activity, increased oxidative stress, and mitochondrial DNA damage. Lmx1a/b deficiency caused axonal pathology characterized by α-synuclein(+) inclusions, followed by a progressive loss of dopaminergic neurons. These results reveal the key role of these transcription factors beyond the early developmental stages and provide mechanistic links between mitochondrial dysfunctions, α-synuclein aggregation, and the survival of dopaminergic neurons.


Subject(s)
Dopaminergic Neurons/metabolism , LIM-Homeodomain Proteins/genetics , Mesencephalon/metabolism , Mitochondria/metabolism , Transcription Factors/genetics , Animals , Cell Survival/genetics , DNA Damage , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , LIM-Homeodomain Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Oxidative Stress , Protein Aggregation, Pathological , Transcription Factors/deficiency , alpha-Synuclein/metabolism
16.
Dev Biol ; 414(2): 181-92, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27126199

ABSTRACT

Angioblasts of the developing vascular system require many signaling inputs to initiate their migration, proliferation and differentiation into endothelial cells. What is less studied is which intrinsic cell factors interpret these extrinsic signals. Here, we show the Lim homeodomain transcription factor islet2a (isl2a) is expressed in the lateral posterior mesoderm prior to angioblast migration. isl2a deficient angioblasts show disorganized migration to the midline to form axial vessels and fail to spread around the tailbud of the embryo. Isl2a morphants have fewer vein cells and decreased vein marker expression. We demonstrate that isl2a is required cell autonomously in angioblasts to promote their incorporation into the vein, and is permissive for vein identity. Knockout of isl2a results in decreased migration and proliferation of angioblasts during intersegmental artery growth. Since Notch signaling controls both artery-vein identity and tip-stalk cell formation, we explored the interaction of isl2a and Notch. We find that isl2a expression is negatively regulated by Notch activity, and that isl2a positively regulates flt4, a VEGF-C receptor repressed by Notch during angiogenesis. Thus Isl2a may act as an intermediate between Notch signaling and genetic programs controlling angioblast number and migration, placing it as a novel transcriptional regulator of early angiogenesis.


Subject(s)
Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/physiology , Neovascularization, Physiologic/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Arteries/embryology , Cell Movement , Gene Knockout Techniques , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Mesoderm , Morpholinos/genetics , Morpholinos/toxicity , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , RNA, Messenger/genetics , Receptors, Notch/physiology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-3/physiology , Veins/embryology , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
17.
Life Sci ; 151: 348-358, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26921632

ABSTRACT

AIMS: The pathogenesis of myocardial ischemia-reperfusion injury (MI/R) involves an inflammatory response. Since the four-and-a-half LIM domain-containing protein 2 (Fhl2) has been observed to modulate immune cell migration, we aimed to study the consequences of Fhl2(-/-) under MI/R with respect to immune reaction, scar formation, and hemodynamic performance. MATERIAL AND METHODS: In a closed chest model of 1h MI/R, immune cell invasion of phagocytic monocytes was characterized by flow cytometry and immunohistochemistry. In addition, infarct size was assessed by triphenyltetrazolium chloride/Masson trichrome staining 24h/21days after reperfusion and a set of hemodynamic parameters was recorded by catheterisation in Fhl2(-/-) mice and controls. KEY FINDINGS: While flow cytometry did not reveal differences in myocardial CD45(high) immune cell infiltrate, histological analysis showed that infiltrating immune cells in Fhl2(-/-) animals were preferentially located in the perivascular area, whereas in wild type, immune cells were well dispersed within the area at risk. After 24h and 21days of reperfusion, infarct size was significantly reduced in Fhl2(-/-) compared to WT animals. In addition, hemodynamic performance was better in Fhl2(-/-) mice, compared to WT mice up to day 21 of reperfusion. The loss of Fhl2 leads to an altered immune response to myocardial ischemia, which results in smaller infarcts and better hemodynamic performance up to 21days after myocardial ischemia reperfusion. SIGNIFICANCE: Immune cell invasion plays a pivotal role in the context of MI/R. Fhl2 significantly influences immune cell function and immune cell interaction with injured cardiac tissue leading to altered scar composition.


Subject(s)
Cicatrix/physiopathology , Hemodynamics/physiology , Inflammation/pathology , LIM-Homeodomain Proteins/deficiency , Muscle Proteins/deficiency , Myocardial Ischemia/pathology , Reperfusion Injury/physiopathology , Transcription Factors/deficiency , Animals , Cell Movement/immunology , Cicatrix/pathology , LIM-Homeodomain Proteins/genetics , Leukocyte Common Antigens/immunology , Male , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/physiology , Muscle Proteins/genetics , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Ischemia/immunology , Myocardial Ischemia/physiopathology , Reperfusion Injury/pathology , Transcription Factors/genetics
18.
Sci Rep ; 6: 19274, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26786896

ABSTRACT

Allen Brain Atlas (ABA) provides a valuable resource of spatial/temporal gene expressions in mammalian brains. Despite rich information extracted from this database, current analyses suffer from several limitations. First, most studies are either gene-centric or region-centric, thus are inadequate to capture the superposition of multiple spatial-temporal patterns. Second, standard tools of expression analysis such as matrix factorization can capture those patterns but do not explicitly incorporate spatial dependency. To overcome those limitations, we proposed a computational method to detect recurrent patterns in the spatial-temporal gene expression data of developing mouse brains. We demonstrated that regional distinction in brain development could be revealed by localized gene expression patterns. The patterns expressed in the forebrain, medullary and pontomedullary, and basal ganglia are enriched with genes involved in forebrain development, locomotory behavior, and dopamine metabolism respectively. In addition, the timing of global gene expression patterns reflects the general trends of molecular events in mouse brain development. Furthermore, we validated functional implications of the inferred patterns by showing genes sharing similar spatial-temporal expression patterns with Lhx2 exhibited differential expression in the embryonic forebrains of Lhx2 mutant mice. These analysis outcomes confirm the utility of recurrent expression patterns in studying brain development.


Subject(s)
Brain/metabolism , Gene Expression Profiling , Gene Expression Regulation , Transcriptome , Animals , Cluster Analysis , Computational Biology , Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/deficiency , Mice , Mice, Knockout , Transcription Factors/deficiency
19.
Proc Natl Acad Sci U S A ; 112(21): 6736-41, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25971728

ABSTRACT

Current knowledge suggests that cortical sensory area identity is controlled by transcription factors (TFs) that specify area features in progenitor cells and subsequently their progeny in a one-step process. However, how neurons acquire and maintain these features is unclear. We have used conditional inactivation restricted to postmitotic cortical neurons in mice to investigate the role of the TF LIM homeobox 2 (Lhx2) in this process and report that in conditional mutant cortices area patterning is normal in progenitors but strongly affected in cortical plate (CP) neurons. We show that Lhx2 controls neocortical area patterning by regulating downstream genetic and epigenetic regulators that drive the acquisition of molecular properties in CP neurons. Our results question a strict hierarchy in which progenitors dominate area identity, suggesting a novel and more comprehensive two-step model of area patterning: In progenitors, patterning TFs prespecify sensory area blueprints. Sequentially, sustained function of alignment TFs, including Lhx2, is essential to maintain and to translate the blueprints into functional sensory area properties in cortical neurons postmitotically. Our results reemphasize critical roles for Lhx2 that acts as one of the terminal selector genes in controlling principal properties of neurons.


Subject(s)
LIM-Homeodomain Proteins/physiology , Models, Neurological , Neocortex/growth & development , Neocortex/physiology , Transcription Factors/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , Epigenesis, Genetic , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mitosis , Neocortex/cytology , Neural Pathways/cytology , Neural Pathways/growth & development , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Transcription Factors/deficiency , Transcription Factors/genetics
20.
J Neurosci ; 35(13): 5233-46, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834049

ABSTRACT

Spinal cord neurons respond to peripheral noxious stimuli and relay this information to higher brain centers, but the molecules controlling the assembly of such pathways are poorly known. In this study, we use the intersection of Lmx1b and Hoxb8::Cre expression in the spinal cord to genetically define nociceptive circuits. Specifically, we show that Lmx1b, previously shown to be expressed in glutamatergic dorsal horn neurons and critical for dorsal horn development, is expressed in nociceptive dorsal horn neurons and that its deletion results in the specific loss of excitatory dorsal horn neurons by apoptosis, without any effect on inhibitory neuron numbers. To assess the behavioral consequences of Lmx1b deletion in the spinal cord, we used the brain-sparing driver Hoxb8::Cre. We show that such a deletion of Lmxb1 leads to a robust reduction in sensitivity to mechanical and thermal noxious stimulation. Furthermore, such conditional mutant mice show a loss of a subpopulation of glutamatergic dorsal horn neurons, abnormal sensory afferent innervations, and reduced spinofugal innervation of the parabrachial nucleus and the periaqueductal gray, important nociceptive structures. Together, our results demonstrate an important role for the intersection of Lmx1b and Hoxb8::cre expression in the development of nociceptive dorsal horn circuits critical for mechanical and thermal pain processing.


Subject(s)
Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/physiology , Nociception/physiology , Posterior Horn Cells/physiology , Spinal Cord Dorsal Horn/cytology , Spinal Cord Dorsal Horn/metabolism , Transcription Factors/physiology , Animals , Apoptosis , Gene Deletion , Gene Expression Regulation , Glutamic Acid/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Mice , Neural Pathways , Neurons, Afferent , Parabrachial Nucleus/physiology , Periaqueductal Gray/physiology , Posterior Horn Cells/cytology , Posterior Horn Cells/pathology , Spinal Cord Dorsal Horn/growth & development , Spinal Cord Dorsal Horn/pathology , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...