Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters








Publication year range
1.
J Cell Sci ; 137(5)2024 03 01.
Article in English | MEDLINE | ID: mdl-37818620

ABSTRACT

The membrane potential (MP) controls cell homeostasis by directing molecule transport and gene expression. How the MP is set upon epithelial differentiation is unknown. Given that tissue architecture also controls homeostasis, we investigated the relationship between basoapical polarity and resting MP in three-dimensional culture of the HMT-3522 breast cancer progression. A microelectrode technique to measure MP and input resistance reveals that the MP is raised by gap junction intercellular communication (GJIC), which directs tight-junction mediated apical polarity, and is decreased by the Na+/K+/2Cl- (NKCC, encoded by SLC12A1 and SLC12A2) co-transporter, active in multicellular structures displaying basal polarity. In the tumor counterpart, the MP is reduced. Cancer cells display diminished GJIC and do not respond to furosemide, implying loss of NKCC activity. Induced differentiation of cancer cells into basally polarized multicellular structures restores widespread GJIC and NKCC responses, but these structures display the lowest MP. The absence of apical polarity, necessary for cancer onset, in the non-neoplastic epithelium is also associated with the lowest MP under active Cl- transport. We propose that the loss of apical polarity in the breast epithelium destabilizes cellular homeostasis in part by lowering the MP.


Subject(s)
Mammary Glands, Human , Humans , Membrane Potentials , Epithelium/metabolism , Breast , Cell Communication/physiology , Cell Polarity/physiology , Epithelial Cells , Solute Carrier Family 12, Member 2/metabolism
2.
Front Cell Dev Biol ; 10: 887107, 2022.
Article in English | MEDLINE | ID: mdl-35800889

ABSTRACT

Epithelial tissues form continuous barriers to protect against external environments. Within these tissues, epithelial cells build environment-facing apical membranes, junction complexes that anchor neighbors together, and basolateral surfaces that face other cells. Critically, to form a continuous apical barrier, neighboring epithelial cells must align their apico-basolateral axes to create global polarity along the entire tissue. Here, we will review mechanisms of global tissue-level polarity establishment, with a focus on how neighboring epithelial cells of different origins align their apical surfaces. Epithelial cells with different developmental origins and/or that polarize at different times and places must align their respective apico-basolateral axes. Connecting different epithelial tissues into continuous sheets or tubes, termed epithelial fusion, has been most extensively studied in cases where neighboring cells initially dock at an apical-to-apical interface. However, epithelial cells can also meet basal-to-basal, posing several challenges for apical continuity. Pre-existing basement membrane between the tissues must be remodeled and/or removed, the cells involved in docking are specialized, and new cell-cell adhesions are formed. Each of these challenges can involve changes to apico-basolateral polarity of epithelial cells. This minireview highlights several in vivo examples of basal docking and how apico-basolateral polarity changes during epithelial fusion. Understanding the specific molecular mechanisms of basal docking is an area ripe for further exploration that will shed light on complex morphogenetic events that sculpt developing organisms and on the cellular mechanisms that can go awry during diseases involving the formation of cysts, fistulas, atresias, and metastases.

3.
J Genet Genomics ; 48(1): 52-62, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33771456

ABSTRACT

Although the unique organization of vertebrate cone mosaics was first described long ago, both their underlying molecular basis and physiological significance are largely unknown. Here, we demonstrate that Crumbs proteins, the key regulators of epithelial apical polarity, establish the planar cellular polarity of photoreceptors in zebrafish. Via heterophilic Crb2a-Crb2b interactions, the apicobasal polarity protein Crb2b restricts the asymmetric planar distribution of Crb2a in photoreceptors. The planar polarized Crumbs proteins thus balance intercellular adhesions and tension between photoreceptors, thereby stabilizing the geometric organization of cone mosaics. Notably, loss of Crb2b in zebrafish induces a nearsightedness-like phenotype in zebrafish accompanied by an elongated eye axis and impairs zebrafish visual perception for predation. These data reveal a detailed mechanism for cone mosaic homeostasis via previously undiscovered apical-planar polarity coordination and propose a pathogenic mechanism for nearsightedness.


Subject(s)
Membrane Proteins , Retinal Cone Photoreceptor Cells , Zebrafish Proteins , Zebrafish , Animals , Cell Polarity/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Dev Cell ; 56(3): 325-340.e8, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33561422

ABSTRACT

Primary cilia are sensory organelles that utilize the compartmentalization of membrane and cytoplasm to communicate signaling events, and yet, how the formation of a cilium is coordinated with reorganization of the cortical membrane and cytoskeleton is unclear. Using polarized epithelia, we find that cortical actin clearing and apical membrane partitioning occur where the centrosome resides at the cell surface prior to ciliation. RAB19, a previously uncharacterized RAB, associates with the RAB-GAP TBC1D4 and the HOPS-tethering complex to coordinate cortical clearing and ciliary membrane growth, which is essential for ciliogenesis. This RAB19-directed pathway is not exclusive to polarized epithelia, as RAB19 loss in nonpolarized cell types blocks ciliogenesis with a docked ciliary vesicle. Remarkably, inhibiting actomyosin contractility can substitute for the function of the RAB19 complex and restore ciliogenesis in knockout cells. Together, this work provides a mechanistic understanding behind a cytoskeletal clearing and membrane partitioning step required for ciliogenesis.


Subject(s)
Cell Membrane/metabolism , Cilia/metabolism , Organogenesis , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Cell Line , Cell Polarity , Centrosome/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , GTPase-Activating Proteins , Humans , Intracellular Space/metabolism , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport
5.
Front Cell Dev Biol ; 9: 619596, 2021.
Article in English | MEDLINE | ID: mdl-33634121

ABSTRACT

Cephalization is a major innovation of animal evolution and implies a synchronization of nervous system, mouth, and foregut polarization to align alimentary tract and sensomotoric system for effective foraging. However, the underlying integration of morphogenetic programs is poorly understood. Here, we show that invagination of neuroectoderm through de novo polarization and apical constriction creates the mouth opening in the Caenorhabditis elegans embryo. Simultaneously, all 18 juxta-oral sensory organ dendritic tips become symmetrically positioned around the mouth: While the two bilaterally symmetric amphid sensilla endings are towed to the mouth opening, labial and cephalic sensilla become positioned independently. Dendrite towing is enabled by the pre-polarized sensory amphid pores intercalating into the leading edge of the anteriorly migrating epidermal sheet, while apical constriction-mediated cell-cell re-arrangements mediate positioning of all other sensory organs. These two processes can be separated by gradual inactivation of the 26S proteasome activator, RPN-6.1. Moreover, RPN-6.1 also shows a dose-dependent requirement for maintenance of coordinated apical polarization of other organs with apical lumen, the pharynx, and the intestine. Thus, our data unveil integration of morphogenetic programs during the coordination of alimentary tract and sensory organ formation and suggest that this process requires tight control of ubiquitin-dependent protein degradation.

6.
BMC Dev Biol ; 21(1): 2, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33407086

ABSTRACT

BACKGROUND: Mammalian early development comprises the proliferation, differentiation, and self-assembly of the embryonic cells. The classic experiment undertaken by Townes and Holtfreter demonstrated the ability of dissociated embryonic cells to sort and self-organize spontaneously into the original tissue patterns. Here, we further explored the principles and mechanisms underlying the phenomenon of spontaneous tissue organization by studying aggregation and sorting of mouse embryonic stem (ES) cells with differential adhesive affinity in culture. RESULTS: As observed previously, in aggregates of wild-type and E-cadherin-deficient ES cells, the cell assemblies exhibited an initial sorting pattern showing wild-type cells engulfed by less adhesive E-cadherin-deficient ES cells, which fits the pattern predicted by the differential adhesive hypothesis proposed by Malcom Steinberg. However, in further study of more mature cell aggregates, the initial sorting pattern reversed, with the highly adhesive wild-type ES cells forming an outer shell enveloping the less adhesive E-cadherin-deficient cells, contradicting Steinberg's sorting principle. The outer wild-type cells of the more mature aggregates did not differentiate into endoderm, which is known to be able to sort to the exterior from previous studies. In contrast to the naive aggregates, the mature aggregates presented polarized, highly adhesive cells at the outer layer. The surface polarity was observed as an actin cap contiguously spanning across the apical surface of multiple adjacent cells, though independent of the formation of tight junctions. CONCLUSIONS: Our experimental findings suggest that the force of differential adhesive affinity can be overcome by even subtle polarity generated from strong bilateral ligation of highly adhesive cells in determining cell sorting patterns.


Subject(s)
Adhesives , Embryonic Stem Cells , Animals , Cell Differentiation , Cell Polarity , Endoderm , Mice
7.
Development ; 147(22)2020 11 19.
Article in English | MEDLINE | ID: mdl-33060129

ABSTRACT

Adherens junction remodeling regulated by apical polarity proteins constitutes a major driving force for tissue morphogenesis, although the precise mechanism remains inconclusive. Here, we report that, in zebrafish, the Crumbs complex component MPP5a interacts with small GTPase Rab11 in Golgi to transport cadherin and Crumbs components synergistically to the apical domain, thus establishing apical epithelial polarity and adherens junctions. In contrast, Par complex recruited by MPP5a is incapable of interacting with Rab11 but might assemble cytoskeleton to facilitate cadherin exocytosis. In accordance, dysfunction of MPP5a induces an invasive migration of epithelial cells. This adherens junction remodeling pattern is frequently observed in zebrafish lens epithelial cells and neuroepithelial cells. The data identify an unrecognized MPP5a-Rab11 complex and describe its essential role in guiding apical polarization and zonula adherens formation in epithelial cells.


Subject(s)
Adherens Junctions/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Guanylate Cyclase/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , rab GTP-Binding Proteins/metabolism , Adherens Junctions/genetics , Animals , Cadherins/genetics , Cadherins/metabolism , Epithelial Cells , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Guanylate Cyclase/genetics , Protein Transport/physiology , Zebrafish/genetics , Zebrafish Proteins/genetics , rab GTP-Binding Proteins/genetics
8.
Methods Mol Biol ; 2092: 123-145, 2020.
Article in English | MEDLINE | ID: mdl-31786786

ABSTRACT

Tissue-specific or cell-type-specific transcription of protein-coding genes is controlled by both trans-regulatory elements (TREs) and cis-regulatory elements (CREs). However, it is challenging to identify TREs and CREs, which are unknown for most genes. Here, we describe a protocol for identifying two types of transcription-activating CREs-core promoters and enhancers-of zebrafish photoreceptor type-specific genes. This protocol is composed of three phases: bioinformatic prediction, experimental validation, and characterization of the CREs. To better illustrate the principles and logic of this protocol, we exemplify it with the discovery of the core promoter and enhancer of the mpp5b apical polarity gene (also known as ponli), whose red, green, and blue (RGB) cone-specific transcription requires its enhancer, a member of the rainbow enhancer family. While exemplified with an RGB-cone-specific gene, this protocol is general and can be used to identify the core promoters and enhancers of other protein-coding genes.


Subject(s)
Regulatory Elements, Transcriptional/genetics , Retinal Cone Photoreceptor Cells/physiology , Transcription, Genetic/genetics , Zebrafish/genetics , Animals , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Zebrafish Proteins/genetics
9.
Genes (Basel) ; 10(12)2019 11 29.
Article in English | MEDLINE | ID: mdl-31795518

ABSTRACT

The Crumbs complex has prominent roles in the control of apical cell polarity, in the coupling of cell density sensing to downstream cell signaling pathways, and in regulating junctional structures and cell adhesion. The Crumbs complex acts as a conductor orchestrating multiple downstream signaling pathways in epithelial and neuronal tissue development. These pathways lead to the regulation of cell size, cell fate, cell self-renewal, proliferation, differentiation, migration, mitosis, and apoptosis. In retinogenesis, these are all pivotal processes with important roles for the Crumbs complex to maintain proper spatiotemporal cell processes. Loss of Crumbs function in the retina results in loss of the stratified appearance resulting in retinal degeneration and loss of visual function. In this review, we begin by discussing the physiology of vision. We continue by outlining the processes of retinogenesis and how well this is recapitulated between the human fetal retina and human embryonic stem cell (ESC) or induced pluripotent stem cell (iPSC)-derived retinal organoids. Additionally, we discuss the functionality of in utero and preterm human fetal retina and the current level of functionality as detected in human stem cell-derived organoids. We discuss the roles of apical-basal cell polarity in retinogenesis with a focus on Leber congenital amaurosis which leads to blindness shortly after birth. Finally, we discuss Crumbs homolog (CRB)-based gene augmentation.


Subject(s)
Cell Polarity , Human Embryonic Stem Cells/metabolism , Leber Congenital Amaurosis/embryology , Organogenesis , Retina/embryology , Signal Transduction , Human Embryonic Stem Cells/pathology , Humans , Leber Congenital Amaurosis/pathology , Retina/pathology
10.
J Cell Sci ; 132(10)2019 05 16.
Article in English | MEDLINE | ID: mdl-30992345

ABSTRACT

Cell-cell communication is essential for tissue homeostasis, but its contribution to disease prevention remains to be understood. We demonstrate the involvement of connexin 43 (Cx43, also known as GJA1) and related gap junction in epithelial homeostasis, illustrated by polarity-mediated cell cycle entry and mitotic spindle orientation (MSO). Cx43 localization is restricted to the apicolateral membrane of phenotypically normal breast luminal epithelial cells in 3D culture and in vivo Chemically induced blockade of gap junction intercellular communication (GJIC), as well as the absence of Cx43, disrupt the apicolateral distribution of polarity determinant tight junction marker ZO-1 (also known as TJP1) and lead to random MSO and cell multilayering. Induced expression of Cx43 in cells that normally lack this protein reestablishes polarity and proper MSO in 3D culture. Cx43-directed MSO implicates PI3K-aPKC signaling, and Cx43 co-precipitates with signaling node proteins ß-catenin (CTNNB1) and ZO-2 (also known as TJP2) in the polarized epithelium. The distribution of Cx43 is altered by pro-inflammatory breast cancer risk factors such as leptin and high-fat diet, as shown in cell culture and on tissue biopsy sections. The control of polarity-mediated quiescence and MSO may contribute to the tumor-suppressive role of Cx43.


Subject(s)
Breast/cytology , Breast/metabolism , Cell Polarity/physiology , Connexin 43/metabolism , Spindle Apparatus/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Cell Line , Epithelium/metabolism , Female , Gap Junctions/metabolism , Humans , Mitosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Zonula Occludens-2 Protein/metabolism , beta Catenin/metabolism
11.
Front Med (Lausanne) ; 6: 314, 2019.
Article in English | MEDLINE | ID: mdl-31998733

ABSTRACT

Preventing cancer is vastly better than treating the disease in terms of a patient's quality of life and healthcare costs. Yet, to screen for chemopreventative drugs or evaluate interventions aimed at lowering cancer risk, quantitative readouts of risk are needed. In the breast and in other organs of epithelial origin, apical-basal polarity is key to homeostasis and is one of the first tissue characteristics lost during cancer initiation. Therefore, apical-basal polarity may be leveraged as an "architectural" determinant of cancer risk. A classic approach to quantify the localization of epithelial polarity markers is visual scoring at the microscope by trained investigators. This approach is time-intensive and limited to low throughput. To increase the speed, accuracy, and scoring volume, we developed an algorithm that essentially replaces the human eye to objectively quantify epithelial polarity in microscopy images of breast glandular units (acini). Acini in culture are identified based on a nuclear stain and the corresponding masks are divided into concentric terraces of equal width. This positional information is used to calculate radial intensity profiles (RP) of polarity markers. Profiles with a steep slope represent polarized structures, whereas more horizontal curves are indicative of non-polarized acini. To compare treatment effects, RP curves are integrated into summary values of polarity. We envision applications of this method for primary cancer prevention research with acini organoids, specifically (1) to screen for chemoprevention drugs, (2) for toxicological assessment of suspected carcinogens and pharmacological hit compounds, and (3) for personalized evaluation of cancer risk and risk-reducing interventions. The RadialProfiler algorithm developed for the MATLAB computing environment and for users without prior informatics knowledge is publicly available on the Open Science Framework (OSF).

13.
Curr Biol ; 27(9): 1381-1386, 2017 May 08.
Article in English | MEDLINE | ID: mdl-28457868

ABSTRACT

Throughout animals, embryonic cells must ultimately organize into polarized epithelial layers that provide the structural basis for gastrulation or subsequent developmental events [1]. Precisely how this primary epithelium maintains continuous integrity during rapid and repeated cell divisions has never been directly addressed, particularly in cases where early cleavages are driven in synchrony. Representing the early-branching non-bilaterian phylum Cnidaria, embryos of the sea anemone Nematostella vectensis undergo rapid synchronous cell divisions and ultimately give rise to a diploblastic epithelial body plan after gastrulation [2, 3]. Here, using live imaging of apical polarity proteins in Nematostella embryos, we demonstrate that cell polarity is established by the four-cell stage and then reiteratively lost during subsequent mitoses, correlating with transient adhesion disengagement and dramatic deformations of embryonic morphology. Intriguingly, the re-establishment of polarity and adhesion during each interphase is associated with a process of whole-embryo compaction analogous to that observed in mammals [4-7]. Because similar protein dynamics are observed in dividing epithelial cells in Drosophila melanogaster, we propose that cell-cycle-coupled oscillations in apical polarity may be conserved throughout Metazoa.


Subject(s)
Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Epithelial Cells/cytology , Morphogenesis , Sea Anemones/embryology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Cell Cycle , Cell Polarity , Cells, Cultured , Drosophila melanogaster/physiology , Embryo, Nonmammalian/physiology , Epithelial Cells/physiology , Female , Sea Anemones/physiology
14.
Genes Dev ; 31(23-24): 2376-2390, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29330353

ABSTRACT

Proper lumen morphogenesis during pancreas development is critical to endocrine and exocrine cell fate. Recent studies showed that a central network of lumens (termed core), but not the surrounding terminal branches (termed periphery), produces most islet endocrine cells. To date, it remains unclear how pancreatic lumens form and remodel and which aspects of lumen morphogenesis influence cell fate. Importantly, models testing the function of the central lumen network as an endocrine niche are lacking. Here, we identify mechanisms underlying lumen formation and remodeling and show that central lumen network morphogenesis impacts pancreatic endocrine mass. We show that loss of the scaffolding protein Afadin disrupts de novo lumenogenesis and lumen continuity in the tip epithelium. Codepletion of the actomyosin regulator RhoA and Afadin results in defects in the central lumens and arrests lumen remodeling. This arrest leads to prolonged perdurance of the central lumen network over developmental time and expansion of the endocrine progenitor population and, eventually, endocrine mass. Our study uncovers essential roles of Afadin and RhoA in pancreatic central lumen morphogenesis, which subsequently determines endocrine cell mass.


Subject(s)
Microfilament Proteins/metabolism , Morphogenesis/genetics , Pancreas/embryology , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Membrane/metabolism , Endocrine Cells/cytology , Endocrine Cells/metabolism , Endocrine Cells/ultrastructure , Mice , Microfilament Proteins/genetics , Microscopy, Electron, Transmission , Mutation , Pancreas/cytology , Pancreas/ultrastructure , rhoA GTP-Binding Protein
15.
Front Physiol ; 7: 450, 2016.
Article in English | MEDLINE | ID: mdl-27774068

ABSTRACT

Na+, K+-ATPase, or the Na+ pump, is a key component in the maintenance of the epithelial phenotype. In most epithelia, the pump is located in the basolateral domain. Studies from our laboratory have shown that the ß1 subunit of Na+, K+-ATPase plays an important role in this mechanism because homotypic ß1-ß1 interactions between neighboring cells stabilize the pump in the lateral membrane. However, in the retinal pigment epithelium (RPE), the Na+ pump is located in the apical domain. The mechanism of polarization in this epithelium is unclear. We hypothesized that the apical polarization of the pump in RPE cells depends on the expression of its ß2 subunit. ARPE-19 cells cultured for up to 8 weeks on inserts did not polarize, and Na+, K+-ATPase was expressed in the basolateral membrane. In the presence of insulin, transferrin and selenic acid (ITS), ARPE-19 cells cultured for 4 weeks acquired an RPE phenotype, and the Na+ pump was visible in the apical domain. Under these conditions, Western blot analysis was employed to detect the ß2 isoform and immunofluorescence analysis revealed an apparent apical distribution of the ß2 subunit. qPCR results showed a time-dependent increase in the level of ß2 isoform mRNA, suggesting regulation at the transcriptional level. Moreover, silencing the expression of the ß2 isoform in ARPE-19 cells resulted in a decrease in the apical localization of the pump, as assessed by the mislocalization of the α2 subunit in that domain. Our results demonstrate that the apical polarization of Na+, K+-ATPase in RPE cells depends on the expression of the ß2 subunit.

16.
Development ; 142(14): 2392-6, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26199408

ABSTRACT

The retromer complex is a multimeric protein complex involved in recycling proteins from endosomes to the trans-Golgi network or plasma membrane. It thus regulates the abundance and subcellular distribution of its cargo within cells. Studies using model organisms show that the retromer complex is involved in specific developmental processes. Moreover, a number of recent studies implicate aberrant retromer function in photoreceptor degeneration, Alzheimer's disease and Parkinson's disease. Here, and in the accompanying poster, we provide an overview of the molecular and cellular mechanisms of retromer-mediated protein trafficking, highlighting key examples of retromer function in vivo.


Subject(s)
Gene Expression Regulation, Developmental , Multiprotein Complexes/physiology , Alzheimer Disease/metabolism , Animals , Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Endosomes/metabolism , Humans , Neurons/metabolism , Parkinson Disease/metabolism , Protein Transport/physiology , Signal Transduction , Vesicular Transport Proteins/metabolism , Wnt Proteins/metabolism , trans-Golgi Network/metabolism
17.
Development ; 142(11): 2048-57, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25977367

ABSTRACT

Tubes are essential for nutrient transport and gas exchange in multicellular eukaryotes, but how connections between different tube types are maintained over time is unknown. In the Drosophila tracheal system, mutations in oak gall (okg) and conjoined (cnj) confer identical defects, including late onset blockage near the terminal cell-stalk cell junction and the ectopic extension of autocellular, seamed tubes into the terminal cell. We determined that okg and cnj encode the E and G subunits of the vacuolar ATPase (vATPase) and showed that both the V0 and V1 domains are required for terminal cell morphogenesis. Remarkably, the ectopic seamed tubes running along vATPase-deficient terminal cells belonged to the neighboring stalk cells. All vATPase-deficient tracheal cells had reduced apical domains and terminal cells displayed mislocalized apical proteins. Consistent with recent reports that the mTOR and vATPase pathways intersect, we found that mTOR pathway mutants phenocopied okg and cnj. Furthermore, terminal cells depleted for the apical determinants Par6 or aPKC had identical ectopic seamed tube defects. We thus identify a novel mechanism of compensatory branching in which stalk cells extend autocellular tubes into neighboring terminal cells with undersized apical domains. This compensatory branching also occurs in response to injury, with damaged terminal cells being rapidly invaded by their stalk cell neighbor.


Subject(s)
Drosophila melanogaster/cytology , Morphogenesis , Trachea/cytology , Adherens Junctions/metabolism , Animals , Cell Polarity , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Holoenzymes/metabolism , Intracellular Space/metabolism , Mutation/genetics , Protein Subunits/metabolism , Proton Pumps , Proton-Translocating ATPases/metabolism , Signal Transduction , Trachea/growth & development , Vacuoles/enzymology
18.
Development ; 141(7): 1492-502, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24574009

ABSTRACT

Hox genes encode a conserved family of homeodomain transcription factors regulating development along the major body axis. During embryogenesis, Hox proteins are expressed in segment-specific patterns and control numerous different segment-specific cell fates. It has been unclear, however, whether Hox proteins drive the epithelial cell segregation mechanism that is thought to initiate the segmentation process. Here, we investigate the role of vertebrate Hox proteins during the partitioning of the developing hindbrain into lineage-restricted units called rhombomeres. Loss-of-function mutants and ectopic expression assays reveal that Hoxb4 and its paralogue Hoxd4 are necessary and sufficient for cell segregation, and for the most caudal rhombomere boundary (r6/r7). Hox4 proteins regulate Eph/ephrins and other cell-surface proteins, and can function in a non-cell-autonomous manner to induce apical cell enlargement on both sides of their expression border. Similarly, other Hox proteins expressed at more rostral rhombomere interfaces can also regulate Eph/ephrins, induce apical remodelling and drive cell segregation in ectopic expression assays. However, Krox20, a key segmentation factor expressed in odd rhombomeres (r3 and r5), can largely override Hox proteins at the level of regulation of a cell surface target, Epha4. This study suggests that most, if not all, Hox proteins share a common potential to induce cell segregation but in some contexts this is masked or modulated by other transcription factors.


Subject(s)
Body Patterning/genetics , Cell Movement/genetics , Homeodomain Proteins/physiology , Rhombencephalon/embryology , Animals , Animals, Genetically Modified , Chick Embryo , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Genes, Homeobox/physiology , Membrane Proteins/genetics , Mice , Rhombencephalon/metabolism , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL