Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 871
Filter
1.
Cell ; 176(1-2): 198-212.e15, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30503211

ABSTRACT

Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2ß forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2ß for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.


Subject(s)
DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Zinc Finger Protein GLI1/metabolism , 3T3 Cells , Animals , Carcinoma, Basal Cell/metabolism , Cell Line , Chromatin , DNA-Binding Proteins/physiology , HEK293 Cells , Hedgehog Proteins/metabolism , Hedgehog Proteins/physiology , Histone Deacetylase 1/metabolism , Humans , Membrane Proteins/physiology , Mice , Molecular Chaperones/metabolism , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1/physiology , Zinc Fingers
2.
Development ; 149(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36355083

ABSTRACT

Morphogens of the Hh family trigger gene expression changes in receiving cells in a concentration-dependent manner to regulate their identity, proliferation, death or metabolism, depending on the tissue or organ. This variety of responses relies on a conserved signaling pathway. Its logic includes a negative-feedback loop involving the Hh receptor Ptc. Here, using experiments and computational models we study and compare the different spatial signaling profiles downstream of Hh in several developing Drosophila organs. We show that the spatial distributions of Ptc and the activator transcription factor CiA in wing, antenna and ocellus show similar features, but are markedly different from that in the compound eye. We propose that these two profile types represent two time points along the signaling dynamics, and that the interplay between the spatial displacement of the Hh source in the compound eye and the negative-feedback loop maintains the receiving cells effectively in an earlier stage of signaling. These results show how the interaction between spatial and temporal dynamics of signaling and differentiation processes may contribute to the informational versatility of the conserved Hh signaling pathway.


Subject(s)
Drosophila , Hedgehog Proteins , Signal Transduction , Drosophila/embryology , Animals , Hedgehog Proteins/physiology , Wings, Animal/embryology , Compound Eye, Arthropod/embryology
3.
Nat Rev Mol Cell Biol ; 14(7): 416-29, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23719536

ABSTRACT

The cloning of the founding member of the Hedgehog (HH) family of secreted proteins two decades ago inaugurated a field that has diversified to encompass embryonic development, stem cell biology and tissue homeostasis. Interest in HH signalling increased when the pathway was implicated in several cancers and congenital syndromes. The mechanism of HH signalling is complex and remains incompletely understood. Nevertheless, studies have revealed novel biological insights into this system, including the function of HH lipidation in the secretion and transport of this ligand and details of the signal transduction pathway, which involves Patched 1, Smoothened and GLI proteins (Cubitus interruptus in Drosophila melanogaster), as well as, in vertebrates, primary cilia.


Subject(s)
Body Patterning , Hedgehog Proteins/physiology , Neoplasms/metabolism , Signal Transduction , Animals , Cilia/metabolism , Humans , Protein Processing, Post-Translational , Receptors, Cell Surface/metabolism , Secretory Pathway
4.
PLoS Biol ; 19(8): e3001367, 2021 08.
Article in English | MEDLINE | ID: mdl-34379617

ABSTRACT

Damage in the nervous system induces a stereotypical response that is mediated by glial cells. Here, we use the eye disc of Drosophila melanogaster as a model to explore the mechanisms involved in promoting glial cell response after neuronal cell death induction. We demonstrate that these cells rapidly respond to neuronal apoptosis by increasing in number and undergoing morphological changes, which will ultimately grant them phagocytic abilities. We found that this glial response is controlled by the activity of Decapentaplegic (Dpp) and Hedgehog (Hh) signalling pathways. These pathways are activated after cell death induction, and their functions are necessary to induce glial cell proliferation and migration to the eye discs. The latter of these 2 processes depend on the function of the c-Jun N-terminal kinase (JNK) pathway, which is activated by Dpp signalling. We also present evidence that a similar mechanism controls glial response upon apoptosis induction in the leg discs, suggesting that our results uncover a mechanism that might be involved in controlling glial cells response to neuronal cell death in different regions of the peripheral nervous system (PNS).


Subject(s)
Compound Eye, Arthropod/growth & development , Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Hedgehog Proteins/physiology , Neuroglia/physiology , Animals , Apoptosis , Cell Movement , Compound Eye, Arthropod/cytology , Drosophila melanogaster/cytology , MAP Kinase Signaling System
5.
PLoS Biol ; 18(11): e3000946, 2020 11.
Article in English | MEDLINE | ID: mdl-33253145

ABSTRACT

Inflammation of the central nervous system (CNS) induces endothelial blood-brain barrier (BBB) opening as well as the formation of a tight junction barrier between reactive astrocytes at the Glia Limitans. We hypothesized that the CNS parenchyma may acquire protection from the reactive astrocytic Glia Limitans not only during neuroinflammation but also when BBB integrity is compromised in the resting state. Previous studies found that astrocyte-derived Sonic hedgehog (SHH) stabilizes the BBB during CNS inflammatory disease, while endothelial-derived desert hedgehog (DHH) is expressed at the BBB under resting conditions. Here, we investigated the effects of endothelial Dhh on the integrity of the BBB and Glia Limitans. We first characterized DHH expression within endothelial cells at the BBB, then demonstrated that DHH is down-regulated during experimental autoimmune encephalomyelitis (EAE). Using a mouse model in which endothelial Dhh is inducibly deleted, we found that endothelial Dhh both opens the BBB via the modulation of forkhead box O1 (FoxO1) transcriptional activity and induces a tight junctional barrier at the Glia Limitans. We confirmed the relevance of this glial barrier system in human multiple sclerosis active lesions. These results provide evidence for the novel concept of "chronic neuroinflammatory tolerance" in which BBB opening in the resting state is sufficient to stimulate a protective barrier at the Glia Limitans that limits the severity of subsequent neuroinflammatory disease. In summary, genetic disruption of the BBB generates endothelial signals that drive the formation under resting conditions of a secondary barrier at the Glia Limitans with protective effects against subsequent CNS inflammation. The concept of a reciprocally regulated CNS double barrier system has implications for treatment strategies in both the acute and chronic phases of multiple sclerosis pathophysiology.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Brain Barrier/physiopathology , Adherens Junctions/pathology , Adherens Junctions/physiology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Astrocytes/pathology , Astrocytes/physiology , Cadherins/genetics , Cadherins/physiology , Capillary Permeability/genetics , Capillary Permeability/physiology , Claudin-5/genetics , Claudin-5/physiology , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Tight Junctions/pathology , Tight Junctions/physiology
6.
Dev Biol ; 477: 177-190, 2021 09.
Article in English | MEDLINE | ID: mdl-34038742

ABSTRACT

Teleost fish fins, like all vertebrate limbs, comprise a series of bones laid out in characteristic pattern. Each fin's distal bony rays typically branch to elaborate skeletal networks providing form and function. Zebrafish caudal fin regeneration studies suggest basal epidermal-expressed Sonic hedgehog (Shh) promotes ray branching by partitioning pools of adjacent pre-osteoblasts. This Shh role is distinct from its well-studied Zone of Polarizing Activity role establishing paired limb positional information. Therefore, we investigated if and how Shh signaling similarly functions during developmental ray branching of both paired and unpaired fins while resolving cellular dynamics of branching by live imaging. We found shha is expressed uniquely by basal epidermal cells overlying pre-osteoblast pools at the distal aspect of outgrowing juvenile fins. Lateral splitting of each shha-expressing epidermal domain followed by the pre-osteoblast pools precedes overt ray branching. We use ptch2:Kaede fish and Kaede photoconversion to identify short stretches of shha+basal epidermis and juxtaposed pre-osteoblasts as the Shh/Smoothened (Smo) active zone. Basal epidermal distal collective movements continuously replenish each shha+domain with individual cells transiently expressing and responding to Shh. In contrast, pre-osteoblasts maintain Shh/Smo activity until differentiating. The Smo inhibitor BMS-833923 prevents branching in all fins, paired and unpaired, with surprisingly minimal effects on caudal fin initial skeletal patterning, ray outgrowth or bone differentiation. Staggered BMS-833923 addition indicates Shh/Smo signaling acts throughout the branching process. We use live cell tracking to find Shh/Smo restrains the distal movement of basal epidermal cells by apparent 'tethering' to pre-osteoblasts. We propose short-range Shh/Smo signaling promotes these heterotypic associations to couple instructive basal epidermal collective movements to pre-osteoblast repositioning as a unique mode of branching morphogenesis.


Subject(s)
Animal Fins/embryology , Epidermal Cells/physiology , Epidermis/embryology , Hedgehog Proteins/physiology , Morphogenesis , Zebrafish Proteins/physiology , Animal Fins/cytology , Animal Fins/metabolism , Animals , Benzamides/pharmacology , Cell Movement , Epidermis/metabolism , Patched-2 Receptor/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Smoothened Receptor/physiology , Zebrafish
7.
Development ; 146(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31676552

ABSTRACT

During cochlear development, hair cells (HCs) and supporting cells differentiate in the prosensory domain to form the organ of Corti, but how one row of inner HCs (IHCs) and three rows of outer HCs (OHCs) are organized is not well understood. Here, we investigated the process of HC induction by monitoring Atoh1 expression in cochlear explants of Atoh1-EGFP knock-in mouse embryos and showed that only the cells that express Atoh1 over a certain threshold are selected for HC fate determination. HC induction initially occurs at the medial edge of the prosensory domain to form IHCs and subsequently at the lateral edge to form OHCs, while Hedgehog signaling maintains a space between IHCs and OHCs, leading to formation of the tunnel of Corti. These results reveal dynamic Atoh1 expression in HC fate control and suggest that multi-directional signals regulate OHC induction, thereby organizing the prototype of the organ of Corti.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Cochlea/embryology , Hair Cells, Auditory/cytology , Animals , Body Patterning , Bone Morphogenetic Protein 4/physiology , Cell Differentiation , Cell Lineage , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/physiology , Hedgehog Proteins/physiology , Imaging, Three-Dimensional , Mice , Microscopy, Fluorescence , Microscopy, Video , Organ of Corti/embryology , Receptors, Notch/physiology , Signal Transduction
8.
Cereb Cortex ; 31(10): 4730-4741, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34002221

ABSTRACT

The neocortex, the center for higher brain function, emerged in mammals and expanded in the course of evolution. The expansion of outer radial glia (oRGs) and intermediate progenitor cells (IPCs) plays key roles in the expansion and consequential folding of the neocortex. Therefore, understanding the mechanisms of oRG and IPC expansion is important for understanding neocortical development and evolution. By using mice and human cerebral organoids, we previously revealed that hedgehog (HH) signaling expands oRGs and IPCs. Nevertheless, it remained to be determined whether HH signaling expanded oRGs and IPCs in vivo in gyrencephalic species, in which oRGs and IPCs are naturally expanded. Here, we show that HH signaling is necessary and sufficient to expand oRGs and IPCs in ferrets, a gyrencephalic species, through conserved cellular mechanisms. HH signaling increases oRG-producing division modes of ventricular radial glia (vRGs), oRG self-renewal, and IPC proliferation. Notably, HH signaling affects vRG division modes only in an early restricted phase before superficial-layer neuron production peaks. Beyond this restricted phase, HH signaling promotes oRG self-renewal. Thus, HH signaling expands oRGs and IPCs in two distinct but continuous phases during cortical development.


Subject(s)
Cerebral Cortex/physiology , Ependymoglial Cells/physiology , Ferrets/physiology , Hedgehog Proteins/physiology , Signal Transduction/physiology , Animals , Cerebral Cortex/cytology , Excitatory Postsynaptic Potentials/physiology , Female , Neocortex/growth & development , Neocortex/physiology , Neural Stem Cells/physiology , Neurons/physiology , Organ Culture Techniques , Pregnancy
9.
Proc Natl Acad Sci U S A ; 116(47): 23636-23642, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31685615

ABSTRACT

Sonic hedgehog (SHH) signaling plays a pivotal role in 2 different phases during brain development. Early SHH signaling derived from the prechordal plate (PrCP) triggers secondary Shh induction in the forebrain, which overlies the PrCP, and the induced SHH signaling, in turn, directs late neuronal differentiation of the forebrain. Consequently, Shh regulation in the PrCP is crucial for initiation of forebrain development. However, no enhancer that regulates prechordal Shh expression has yet been found. Here, we identified a prechordal enhancer, named SBE7, in the vicinity of a cluster of known forebrain enhancers for Shh This enhancer also directs Shh expression in the ventral midline of the forebrain, which receives the prechordal SHH signal. Thus, the identified enhancer acts not only for the initiation of Shh regulation in the PrCP but also for subsequent Shh induction in the forebrain. Indeed, removal of the enhancer from the mouse genome markedly down-regulated the expression of Shh in the rostral domains of the axial mesoderm and in the ventral midline of the forebrain and hypothalamus in the mouse embryo, and caused a craniofacial abnormality similar to human holoprosencephaly (HPE). These findings demonstrate that SHH signaling mediated by the newly identified enhancer is essential for development and growth of the ventral midline of the forebrain and hypothalamus. Understanding of the Shh regulation governed by this prechordal and brain enhancer provides an insight into the mechanism underlying craniofacial morphogenesis and the etiology of HPE.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Nerve Tissue Proteins/physiology , Prosencephalon/embryology , Animals , CRISPR-Cas Systems , Eye Proteins/physiology , Gene Knockout Techniques , Genes, Reporter , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Homeodomain Proteins/physiology , Hypothalamus/abnormalities , Hypothalamus/embryology , Hypothalamus/metabolism , Lac Operon , Mesencephalon/embryology , Mesencephalon/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Prosencephalon/abnormalities , Prosencephalon/metabolism , Signal Transduction , Transgenes , Homeobox Protein SIX3
10.
Genes Dev ; 28(22): 2547-63, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25403183

ABSTRACT

Control of Gli function by Suppressor of Fused (Sufu), a major negative regulator, is a key step in mammalian Hedgehog (Hh) signaling, but how this is achieved in the nucleus is unknown. We found that Hh signaling results in reduced Sufu protein levels and Sufu dissociation from Gli proteins in the nucleus, highlighting critical functions of Sufu in the nucleus. Through a proteomic approach, we identified several Sufu-interacting proteins, including p66ß (a member of the NuRD [nucleosome remodeling and histone deacetylase] repressor complex) and Mycbp (a Myc-binding protein). p66ß negatively and Mycbp positively regulate Hh signaling in cell-based assays and zebrafish. They function downstream from the membrane receptors, Patched and Smoothened, and the primary cilium. Sufu, p66ß, Mycbp, and Gli are also detected on the promoters of Hh targets in a dynamic manner. Our results support a new model of Hh signaling in the nucleus. Sufu recruits p66ß to block Gli-mediated Hh target gene expression. Meanwhile, Mycbp forms a complex with Gli and Sufu without Hh stimulation but remains inactive. Hh pathway activation leads to dissociation of Sufu/p66ß from Gli, enabling Mycbp to promote Gli protein activity and Hh target gene expression. These studies provide novel insight into how Sufu controls Hh signaling in the nucleus.


Subject(s)
Gene Expression Regulation , Hedgehog Proteins/physiology , Repressor Proteins/metabolism , Salivary alpha-Amylases/metabolism , Signal Transduction , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Knockdown Techniques , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mutation , NIH 3T3 Cells , Protein Binding , Proteomics , Repressor Proteins/genetics , Salivary alpha-Amylases/genetics , Zebrafish/genetics , Zinc Finger Protein GLI1
11.
Dev Biol ; 463(1): 26-38, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32151560

ABSTRACT

Non-syndromic mitral valve prolapse (MVP) is the most common heart valve disease affecting 2.4% of the population. Recent studies have identified genetic defects in primary cilia as causative to MVP, although the mechanism of their action is currently unknown. Using a series of gene inactivation approaches, we define a paracrine mechanism by which endocardially-expressed Desert Hedgehog (DHH) activates primary cilia signaling on neighboring valve interstitial cells. High-resolution imaging and functional assays show that DHH de-represses smoothened at the primary cilia, resulting in kinase activation of RAC1 through the RAC1-GEF, TIAM1. Activation of this non-canonical hedgehog pathway stimulates α-smooth actin organization and ECM remodeling. Genetic or pharmacological perturbation of this pathway results in enlarged valves that progress to a myxomatous phenotype, similar to valves seen in MVP patients. These data identify a potential molecular origin for MVP as well as establish a paracrine DHH-primary cilium cross-talk mechanism that is likely applicable across developmental tissue types.


Subject(s)
Cilia/metabolism , Hedgehog Proteins/metabolism , Mitral Valve/embryology , Actins/metabolism , Animals , Extracellular Matrix/metabolism , Heart Valve Diseases , Hedgehog Proteins/physiology , Mice , Mitral Valve Prolapse/genetics , Mitral Valve Prolapse/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Myocytes, Smooth Muscle/metabolism , Neuropeptides/metabolism , Phenotype , Signal Transduction , Transcription Factors/metabolism , rac1 GTP-Binding Protein/metabolism
12.
Neurobiol Dis ; 150: 105236, 2021 03.
Article in English | MEDLINE | ID: mdl-33383187

ABSTRACT

Development of the forebrain critically depends on the Sonic Hedgehog (Shh) signaling pathway, as illustrated in humans by the frequent perturbation of this pathway in holoprosencephaly, a condition defined as a defect in the formation of midline structures of the forebrain and face. The Shh pathway requires functional primary cilia, microtubule-based organelles present on virtually every cell and acting as cellular antennae to receive and transduce diverse chemical, mechanical or light signals. The dysfunction of cilia in humans leads to inherited diseases called ciliopathies, which often affect many organs and show diverse manifestations including forebrain malformations for the most severe forms. The purpose of this review is to provide the reader with a framework to understand the developmental origin of the forebrain defects observed in severe ciliopathies with respect to perturbations of the Shh pathway. We propose that many of these defects can be interpreted as an imbalance in the ratio of activator to repressor forms of the Gli transcription factors, which are effectors of the Shh pathway. We also discuss the complexity of ciliopathies and their relationships with forebrain disorders such as holoprosencephaly or malformations of cortical development, and emphasize the need for a closer examination of forebrain defects in ciliopathies, not only through the lens of animal models but also taking advantage of the increasing potential of the research on human tissues and organoids.


Subject(s)
Brain/abnormalities , Cilia/genetics , Ciliopathies/embryology , Craniofacial Abnormalities/embryology , Hedgehog Proteins/physiology , Prosencephalon/embryology , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Brain/embryology , Cerebellum/abnormalities , Cerebellum/embryology , Ciliary Motility Disorders/embryology , Ciliary Motility Disorders/genetics , Ciliopathies/genetics , Craniofacial Abnormalities/genetics , Developmental Disabilities/genetics , Encephalocele/embryology , Encephalocele/genetics , Eye Abnormalities/embryology , Eye Abnormalities/genetics , Gene Expression Regulation, Developmental , Holoprosencephaly/embryology , Holoprosencephaly/genetics , Humans , Kidney Diseases, Cystic/embryology , Kidney Diseases, Cystic/genetics , Polycystic Kidney Diseases/embryology , Polycystic Kidney Diseases/genetics , Retina/abnormalities , Retina/embryology , Retinitis Pigmentosa/embryology , Retinitis Pigmentosa/genetics , Signal Transduction , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli3/genetics
13.
FASEB J ; 34(5): 6791-6807, 2020 05.
Article in English | MEDLINE | ID: mdl-32223017

ABSTRACT

The frontal craniofacial skeleton derived from neural crest cells is vital for facial structure and masticatory functions. The exact role of Indian hedgehog (Ihh) in facial and masticatory development has not been fully explored. In this study, we generated craniofacial neural crest cells-specific Ihh deletion mice (Wnt1-Cre;Ihhfl/fl ;Tomatofl/+ ) and found the gradual dwarfism without perinatal lethality. Morphological and histological analyses revealed unambiguous craniofacial phenotypes in mutants, where we observed skeletal malocclusion accompanied by markedly hypoplastic nasomaxillary complex and reversed incisor occlusion. Both the replacement of nasal concha cartilage by turbinate bones and the endochondral ossification of nasal septum ethmoid bone were substantially delayed. We also observed hypoplastic mandibles in mutants where the mandibular ramus was unexpectedly the most affected. Both the condylar process and mandibular angle cartilages were distorted. However, dental examination showed no significant changes in teeth and dentition. Finally, a comprehensive RNA sequence analysis utilizing condylar cartilage identified Ihh-associated gene network including several cell cycle genes and 16 genes related to the extracellular matrix, sulfate transporters, transcription factors, receptors, a ciliogenesis factor, and an adhesion molecule. Our data provide direct in vivo evidence that Ihh plays crucial roles in midface and masticatory system formation, likely by activating key genes.


Subject(s)
Bone and Bones/pathology , Cartilage/pathology , Gene Expression Regulation, Developmental , Hedgehog Proteins/physiology , Malocclusion/pathology , Neural Crest/pathology , Wnt1 Protein/physiology , Animals , Bone and Bones/metabolism , Cartilage/metabolism , Chondrogenesis , Craniofacial Abnormalities , Female , Male , Malocclusion/genetics , Malocclusion/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Crest/metabolism , Phenotype
14.
Pharmacol Res ; 168: 105595, 2021 06.
Article in English | MEDLINE | ID: mdl-33823219

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant lethal disease due to its asymptomatic at its early lesion of the disease and drug resistance. Target therapy associated with molecular pathways so far seems not to produce reasonable outcomes. Understanding of the molecular mechanisms underlying inflammation-initiated tumorigenesis may be helpful for development of an effective therapy of the disease. A line of studies showed that pancreatic tumorigenesis was resulted from pancreatitis, which was caused synergistically by various pancreatic cells. This review focuses on those players and their possible clinic implications, such as exocrine acinar cells, ductal cells, and various stromal cells, including pancreatic stellate cells (PSCs), macrophages, lymphocytes, neutrophils, mast cells, adipocytes and endothelial cells, working together with each other in an inflammation-mediated microenvironment governed by a myriad of cellular signaling networks towards PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , Pancreatic Neoplasms/etiology , Pancreatitis/complications , Acinar Cells/physiology , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/drug therapy , Hedgehog Proteins/physiology , Humans , MAP Kinase Signaling System/physiology , Macrophages/physiology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/physiology , Signal Transduction , Tumor Microenvironment
15.
Future Oncol ; 17(31): 4185-4206, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34342489

ABSTRACT

Triple-negative breast cancer (TNBC) is the most complex, aggressive and fatal subtype of breast cancer. Owing to the lack of targeted therapy and heterogenic nature of TNBC, chemotherapy remains the sole treatment option for TNBC, with taxanes and anthracyclines representing the general chemotherapeutic regimen in TNBC therapy. But unfortunately, patients develop resistance to the existing chemotherapeutic regimen, resulting in approximately 90% treatment failure. Breast cancer stem cells (BCSCs) are one of the major causes for the development of chemoresistance in TNBC patients. After surviving the chemotherapy damage, the presence of BCSCs results in relapse and recurrence of TNBC. Several pathways are known to regulate BCSCs' survival, such as the Wnt/ß-catenin, Hedgehog, JAK/STAT and HIPPO pathways. Therefore it is imperative to target these pathways in the context of eliminating chemoresistance. In this review we will discuss the novel strategies and various preclinical and clinical studies to give an insight into overcoming TNBC chemoresistance. We present a detailed account of recent studies carried out that open an exciting perspective in relation to the mechanisms of chemoresistance.


Subject(s)
Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/drug therapy , ATP-Binding Cassette Transporters/physiology , Cell Survival , Drug Resistance, Neoplasm , Female , Hedgehog Proteins/physiology , Hippo Signaling Pathway , Humans , NF-kappa B/physiology , Receptors, Notch/physiology , Triple Negative Breast Neoplasms/pathology , Wnt Signaling Pathway
16.
Can J Physiol Pharmacol ; 99(9): 910-920, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33617370

ABSTRACT

The present study evaluated whether epigallocatechin-3-gallate (EGCG) effectively attenuates tumor growth in colon cancer cells and in the xenografts of nude mice and investigated the underlying mechanisms by focusing on the sonic hedgehog (Shh) and phosphoinositide 3-kinase (PI3K) pathways. Three kinds of colon cancer cells and BALB/c nude mice were used to evaluate the antiproliferative effect of EGCG. The apoptosis, migration, and invasion of colon cancer cells were analyzed to explore the toxicity effect of EGCG on colon cancer cells. Western blotting was used to demonstrate the expression levels of related proteins. The results showed that EGCG exhibited an antiproliferative effect against colon cancer cells in a dose-dependent manner with low toxicity against normal colon epithelial cells. Administration of EGCG caused significant apoptosis and inhibited the migration and invasion of colon cancer cells. The toxic effect of EGCG on colon cancer cells was accompanied by downregulation of the Shh and PI3K/Akt pathways. In addition, EGCG reduced tumor volume and weight without affecting the body weight of nude mice and inhibited the activation of the Shh and PI3K/AKT pathways in tumor tissue. Further study showed that purmorphamine (smoothened (Smo) agonist) or insulin like growth factor-1 (IGF-1, PI3K agonist) partly abolished the effect of EGCG on cell proliferation, migration, and apoptosis. Cyclopamine (Smo inhibitor) and LY294002 (PI3K inhibitor) showed the similar toxic effects as EGCG on colon cancer cells. In conclusion, EGCG inhibited colon tumor growth via downregulation of the Shh and PI3K pathways and may be a potential chemotherapeutic agent against colon cancer.


Subject(s)
Catechin/analogs & derivatives , Colonic Neoplasms/drug therapy , Hedgehog Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Apoptosis/drug effects , Catechin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects
17.
Neural Plast ; 2021: 8706400, 2021.
Article in English | MEDLINE | ID: mdl-34221003

ABSTRACT

Neuroinflammation plays important roles in the pathogenesis and progression of altered neurodevelopment, sensorineural hearing loss, and certain neurodegenerative diseases. Hyperoside (quercetin-3-O-ß-D-galactoside) is an active compound isolated from Hypericum plants. In this study, we investigate the protective effect of hyperoside on neuroinflammation and its possible molecular mechanism. Lipopolysaccharide (LPS) and hyperoside were used to treat HT22 cells. The cell viability was measured by MTT assay. The cell apoptosis rate was measured by flow cytometry assay. The mRNA expression levels of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α) were determined by quantitative reverse transcription polymerase chain reaction. The levels of oxidative stress indices superoxide dismutase (SOD), reactive oxygen species (ROS), catalase (CAT), glutathione (GSH), and malondialdehyde (MDA) were measured by the kits. The expression of neurotrophic factor and the relationship among hyperoside, silent mating type information regulation 2 homolog-1 (SIRT1) and Wnt/ß-catenin, and sonic hedgehog was examined by western blotting. In the LPS-induced HT22 cells, hyperoside promotes cell survival; alleviates the level of IL-1ß, IL-6, IL-8, TNF-α, ROS, MDA, Bax, and caspase-3; and increases the expression of CAT, SOD, GSH, Bcl-2, BDNF, TrkB, and NGF. In addition, hyperoside upregulated the expression of SIRT1. Further mechanistic investigation showed that hyperoside alleviated LPS-induced inflammation, oxidative stress, and apoptosis by upregulating SIRT1 to activate Wnt/ß-catenin and sonic hedgehog pathways. Taken together, our data suggested that hyperoside acts as a protector in neuroinflammation.


Subject(s)
Neurons/drug effects , Quercetin/analogs & derivatives , Signal Transduction/drug effects , Sirtuin 1/biosynthesis , Animals , Apoptosis/drug effects , Cell Line , Cytokines/blood , Drug Evaluation, Preclinical , Hedgehog Proteins/physiology , Inflammation , Lipopolysaccharides/pharmacology , Mice , Nerve Growth Factors/physiology , Neurons/metabolism , Oxidative Stress/drug effects , Quercetin/pharmacology , Sirtuin 1/genetics , Up-Regulation/drug effects , Wnt Signaling Pathway/drug effects
18.
Circulation ; 140(6): 500-513, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31163988

ABSTRACT

BACKGROUND: Genome-wide association studies have identified chromosome 14q32 as a locus for coronary artery disease. The disease-associated variants fall in a hitherto uncharacterized gene called HHIPL1 (hedgehog interacting protein-like 1), which encodes a sequence homolog of an antagonist of hedgehog signaling. The function of HHIPL1 and its role in atherosclerosis are unknown. METHODS: HHIPL1 cellular localization, interaction with sonic hedgehog (SHH), and influence on hedgehog signaling were tested. HHIPL1 expression was measured in coronary artery disease-relevant human cells, and protein localization was assessed in wild-type and Apoe-/- (apolipoprotein E deficient) mice. Human aortic smooth muscle cell phenotypes and hedgehog signaling were investigated after gene knockdown. Hhipl1-/- mice were generated and aortic smooth muscle cells collected for phenotypic analysis and assessment of hedgehog signaling activity. Hhipl1-/- mice were bred onto both the Apoe-/- and Ldlr-/- (low-density lipoprotein receptor deficient) knockout strains, and the extent of atherosclerosis was quantified after 12 weeks of high-fat diet. Cellular composition and collagen content of aortic plaques were assessed by immunohistochemistry. RESULTS: In vitro analyses revealed that HHIPL1 is a secreted protein that interacts with SHH and increases hedgehog signaling activity. HHIPL1 expression was detected in human smooth muscle cells and in smooth muscle within atherosclerotic plaques of Apoe-/- mice. The expression of Hhipl1 increased with disease progression in aortic roots of Apoe-/- mice. Proliferation and migration were reduced in Hhipl1 knockout mouse and HHIPL1 knockdown aortic smooth muscle cells, and hedgehog signaling was decreased in HHIPL1-deficient cells. Hhipl1 knockout caused a reduction of >50% in atherosclerosis burden on both Apoe-/- and Ldlr-/- knockout backgrounds, and lesions were characterized by reduced smooth muscle cell content. CONCLUSIONS: HHIPL1 is a secreted proatherogenic protein that enhances hedgehog signaling and regulates smooth muscle cell proliferation and migration. Inhibition of HHIPL1 protein function might offer a novel therapeutic strategy for coronary artery disease.


Subject(s)
Atherosclerosis/genetics , Chromosomes, Human, Pair 14/genetics , Coronary Disease/genetics , Hedgehog Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Atherosclerosis/pathology , Cell Division , Cell Movement , Cells, Cultured , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout, ApoE , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/pathology , Receptors, LDL/deficiency , Signal Transduction
19.
Hum Mol Genet ; 27(R2): R119-R129, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29718288

ABSTRACT

There is growing evidence that the complex clinical manifestations of lysosomal storage diseases (LSDs) are not fully explained by the engorgement of the endosomal-autophagic-lysosomal system. In this review, we explore current knowledge of common pathogenetic mechanisms responsible for the early onset of tissue abnormalities of two LSDs, Mucopolysaccharidosis type II (MPSII) and Niemann-Pick type C (NPC) diseases. In particular, perturbations of the homeostasis of glycosaminoglycans (GAGs) and cholesterol (Chol) in MPSII and NPC diseases, respectively, affect key biological processes, including morphogen signaling. Both GAGs and Chol finely regulate the release, reception and tissue distribution of Shh. Hence, not surprisingly, developmental processes depending on correct Shh signaling have been found altered in both diseases. Besides abnormal signaling, exaggerated activation of microglia and impairment of autophagy and mitophagy occur in both diseases, largely before the appearance of typical pathological signs.


Subject(s)
Lysosomal Storage Diseases/physiopathology , Lysosomes/pathology , Animals , Autophagy , Cholesterol/metabolism , Endocytosis , Endosomes/pathology , Glycosaminoglycans/metabolism , Hedgehog Proteins/physiology , Homeostasis , Humans , Lysosomal Storage Diseases/metabolism , Lysosomes/physiology , Mitophagy , Mucopolysaccharidosis II/pathology , Neuroimmunomodulation/immunology , Neuroimmunomodulation/physiology , Niemann-Pick Disease, Type C/pathology , Wnt Signaling Pathway/physiology
20.
Development ; 144(10): 1906-1917, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28432216

ABSTRACT

The gallbladder excretes cytotoxic bile acids into the duodenum through the cystic duct and common bile duct system. Sox17 haploinsufficiency causes biliary atresia-like phenotypes and hepatitis in late organogenesis mouse embryos, but the molecular and cellular mechanisms underlying this remain unclear. In this study, transcriptomic analyses revealed the early onset of cholecystitis in Sox17+/- embryos, together with the appearance of ectopic cystic duct-like epithelia in their gallbladders. The embryonic hepatitis showed positive correlations with the severity of cholecystitis in individual Sox17+/- embryos. Embryonic hepatitis could be induced by conditional deletion of Sox17 in the primordial gallbladder epithelia but not in fetal liver hepatoblasts. The Sox17+/- gallbladder also showed a drastic reduction in sonic hedgehog expression, leading to aberrant smooth muscle formation and defective contraction of the fetal gallbladder. The defective gallbladder contraction positively correlated with the severity of embryonic hepatitis in Sox17+/- embryos, suggesting a potential contribution of embryonic cholecystitis and fetal gallbladder contraction in the early pathogenesis of congenital biliary atresia.


Subject(s)
Biliary Atresia , Cholecystitis/embryology , Gallbladder/embryology , HMGB Proteins/genetics , Muscle Contraction/genetics , Muscle, Smooth/embryology , SOXF Transcription Factors/genetics , Animals , Biliary Atresia/embryology , Biliary Atresia/genetics , Biliary Atresia/pathology , Cells, Cultured , Cholecystitis/genetics , Disease Models, Animal , Embryo, Mammalian , Female , Gallbladder/metabolism , Gallbladder/physiology , Haploinsufficiency , Hedgehog Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth/physiology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL