Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 295(25): 8401-8412, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32358066

ABSTRACT

Vesicles that are coated by coat protein complex II (COPII) are the primary mediators of vesicular traffic from the endoplasmic reticulum to the Golgi apparatus. Secretion-associated Ras-related GTPase 1 (SAR1) is a small GTPase that is part of COPII and, upon GTP binding, recruits the other COPII proteins to the endoplasmic reticulum membrane. Mammals have two SAR1 paralogs that genetic data suggest may have distinct physiological roles, e.g. in lipoprotein secretion in the case of SAR1B. Here we identified two amino acid clusters that have conserved SAR1 paralog-specific sequences. We observed that one cluster is adjacent to the SAR1 GTP-binding pocket and alters the kinetics of GTP exchange. The other cluster is adjacent to the binding site for two COPII components, SEC31 homolog A COPII coat complex component (SEC31) and SEC23. We found that the latter cluster confers to SAR1B a binding preference for SEC23A that is stronger than that of SAR1A for SEC23A. Unlike SAR1B, SAR1A was prone to oligomerize on a membrane surface. SAR1B knockdown caused loss of lipoprotein secretion, overexpression of SAR1B but not of SAR1A could restore secretion, and a divergent cluster adjacent to the SEC31/SEC23-binding site was critical for this SAR1B function. These results highlight that small primary sequence differences between the two mammalian SAR1 paralogs lead to pronounced biochemical differences that significantly affect COPII assembly and identify a specific function for SAR1B in lipoprotein secretion, providing insights into the mechanisms of large cargo secretion that may be relevant for COPII-related diseases.


Subject(s)
Monomeric GTP-Binding Proteins/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , CRISPR-Cas Systems/genetics , Cell Line , Dimerization , Gene Editing , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Molecular Dynamics Simulation , Monomeric GTP-Binding Proteins/chemistry , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Phylogeny , Protein Binding , Protein Conformation, alpha-Helical , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment , Vesicular Transport Proteins/chemistry
2.
Nat Med ; 26(1): 98-109, 2020 01.
Article in English | MEDLINE | ID: mdl-31932796

ABSTRACT

Discovery of genotype-phenotype relationships remains a major challenge in clinical medicine. Here, we combined three sources of phenotypic data to uncover a new mechanism for rare and common diseases resulting from collagen secretion deficits. Using a zebrafish genetic screen, we identified the ric1 gene as being essential for skeletal biology. Using a gene-based phenome-wide association study (PheWAS) in the EHR-linked BioVU biobank, we show that reduced genetically determined expression of RIC1 is associated with musculoskeletal and dental conditions. Whole-exome sequencing identified individuals homozygous-by-descent for a rare variant in RIC1 and, through a guided clinical re-evaluation, it was discovered that they share signs with the BioVU-associated phenome. We named this new Mendelian syndrome CATIFA (cleft lip, cataract, tooth abnormality, intellectual disability, facial dysmorphism, attention-deficit hyperactivity disorder) and revealed further disease mechanisms. This gene-based, PheWAS-guided approach can accelerate the discovery of clinically relevant disease phenome and associated biological mechanisms.


Subject(s)
Abnormalities, Multiple/pathology , Biological Specimen Banks , Guanine Nucleotide Exchange Factors/genetics , Phenomics , Zebrafish Proteins/genetics , Animals , Behavior, Animal , Chondrocytes/pathology , Chondrocytes/ultrastructure , Disease Models, Animal , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Models, Biological , Musculoskeletal System/pathology , Osteogenesis , Phenotype , Procollagen/metabolism , Protein Transport , Secretory Pathway , Syndrome , Zebrafish
3.
J Mol Med (Berl) ; 93(2): 165-76, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559265

ABSTRACT

Anderson disease (ANDD) or chylomicron retention disease (CMRD) is a rare, hereditary lipid malabsorption syndrome associated with mutations in the SAR1B gene that is characterized by failure to thrive and hypocholesterolemia. Although the SAR1B structure has been resolved and its role in formation of coat protein II (COPII)-coated carriers is well established, little is known about the requirement for SAR1B during embryogenesis. To address this question, we have developed a zebrafish model of Sar1b deficiency based on antisense oligonucleotide knockdown. We show that zebrafish sar1b is highly conserved among vertebrates; broadly expressed during development; and enriched in the digestive tract organs, brain, and craniofacial skeleton. Consistent with ANDD symptoms of chylomicron retention, we found that dietary lipids in Sar1b-deficient embryos accumulate in enterocytes. Transgenic expression analysis revealed that Sar1b is required for growth of exocrine pancreas and liver. Furthermore, we found abnormal differentiation and maturation of craniofacial cartilage associated with defects in procollagen II secretion and absence of select, neuroD-positive neurons of the midbrain and hindbrain. The model presented here will help to systematically dissect developmental roles of Sar1b and to discover molecular and cellular mechanisms leading to organ-specific ANDD pathology. Key messages: Sar1b depletion phenotype in zebrafish resembles Anderson disease deficits. Sar1b deficiency results in multi-organ developmental deficits. Sar1b is required for dietary cholesterol uptake into enterocytes.


Subject(s)
Hypobetalipoproteinemias/genetics , Hypobetalipoproteinemias/metabolism , Lipid Metabolism/genetics , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Monomeric GTP-Binding Proteins/deficiency , Animals , Animals, Genetically Modified , Body Patterning/genetics , Bone and Bones/embryology , Bone and Bones/metabolism , Brain/embryology , Brain/metabolism , Disease Models, Animal , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Immunohistochemistry , Organogenesis/genetics , Phenotype , Zebrafish
4.
J Biol Chem ; 289(17): 11971-11985, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24627485

ABSTRACT

Sec13 is a dual function protein, being a core component of both the COPII coat, which mediates protein trafficking from the endoplasmic reticulum to the Golgi apparatus, and the nuclear pore complex (NPC), which facilitates nucleo-cytoplasmic traffic. Here, we present a genetic model to differentiate the roles of these two functions of Sec13 in vivo. We report that sec13(sq198) mutant embryos develop small eyes that exhibit disrupted retinal lamination and that the mutant retina contains an excessive number of apoptotic cells. Surprisingly, we found that loss of COPII function by oligonucleotide-mediated gene knockdown of sec31a and sec31b or brefeldin A treatment did not disrupt retinal lamination, although it did result in digestive organ defects similar to those seen in sec13(sq198), suggesting that the digestive organ defects observed in sec13(sq198) are due to loss of COPII function, whereas the retinal lamination defects are due to loss of the NPC function. We showed that the retinal cells of sec13(sq198) failed to form proper nuclear pores, leading to a nuclear accumulation of total mRNA and abnormal activation of the p53-dependent apoptosis pathway, causing the retinal defect in sec13(sq198). Furthermore, we found that a mutant lacking Nup107, a key NPC-specific component, phenocopied the retinal lamination phenotype as observed in sec13(sq198). Our results demonstrate a requirement for the nuclear pore function of Sec13 in development of the retina and provide the first genetic evidence to differentiate the contributions of the NPC and the COPII functions of Sec13 during organogenesis.


Subject(s)
Nuclear Pore/physiology , Retina/embryology , Zebrafish Proteins/physiology , Animals , Base Sequence , DNA Primers , In Situ Hybridization , Microscopy, Electron, Transmission , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Zebrafish , Zebrafish Proteins/genetics
5.
Cancer Res ; 74(1): 38-43, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24247717

ABSTRACT

ENOX1 is a highly conserved NADH oxidase that helps to regulate intracellular nicotinamide adenine dinucleotide levels in many cell types, including endothelial cells. Pharmacologic and RNA interference (RNAi)-mediated suppression of ENOX1 impairs surrogate markers of tumor angiogenesis/vasculogenesis, providing support for the concept that ENOX1 represents an antiangiogenic druggable target. However, direct genetic evidence that demonstrates a role for ENOX1 in vascular development is lacking. In this study, we exploited a zebrafish embryonic model of development to address this question. Whole-mount in situ hybridization coupled with immunofluorescence performed on zebrafish embryos demonstrate that enox1 message and translated protein are expressed in most tissues, and its expression is enriched in blood vessels and heart. Morpholino-mediated suppression of Enox1 in Tg(fli1-eGFP) and Tg(flk1-eGFP) zebrafish embryos significantly impairs the development of vasculature and blood circulation. Using in vivo multiphoton microscopy, we show that morpholino-mediated knockdown of enox1 increases NADH levels, consistent with loss of enzyme. VJ115 is a small-molecule inhibitor of Enox1's oxidase activity shown to increase intracellular NADH in endothelial cells; we used VJ115 to determine if the oxidase activity was crucial for vascular development. We found that VJ115 suppressed vasculogenesis in Tg(fli1-eGFP) embryos and impaired circulation. Previously, it was shown that suppression of ENOX1 radiosensitizes proliferating tumor vasculature, a consequence of enhanced endothelial cell apoptosis. Thus, our current findings, coupled with previous research, support the hypothesis that ENOX1 represents a potential cancer therapy target, one that combines molecular targeting with cytotoxic sensitization.


Subject(s)
Endothelium, Vascular/embryology , Endothelium, Vascular/growth & development , Multienzyme Complexes/physiology , NADH, NADPH Oxidoreductases/physiology , Animals , Animals, Genetically Modified , Endothelium, Vascular/enzymology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Neovascularization, Physiologic/physiology , Zebrafish
6.
Int J Biochem Cell Biol ; 47: 57-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24333299

ABSTRACT

Cellular life depends on protein transport and membrane traffic. In multicellular organisms, membrane traffic is required for extracellular matrix deposition, cell adhesion, growth factor release, and receptor signaling, which are collectively required to integrate the development and physiology of tissues and organs. Understanding the regulatory mechanisms that govern cargo and membrane flow presents a prime challenge in cell biology. Extracellular matrix (ECM) secretion remains poorly understood, although given its essential roles in the regulation of cell migration, differentiation, and survival, ECM secretion mechanisms are likely to be tightly controlled. Recent studies in vertebrate model systems, from fishes to mammals and in human patients, have revealed complex and diverse loss-of-function phenotypes associated with mutations in components of the secretory machinery. A broad spectrum of diseases from skeletal and cardiovascular to neurological deficits have been linked to ECM trafficking. These discoveries have directly challenged the prevailing view of secretion as an essential but monolithic process. Here, we will discuss the latest findings on mechanisms of ECM trafficking in vertebrates.


Subject(s)
Extracellular Matrix/metabolism , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Disease Models, Animal , Humans
7.
Dis Model Mech ; 6(2): 332-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23223679

ABSTRACT

Atrial fibrillation (AF) is the most common cardiac arrhythmia and carries a significant risk of stroke and heart failure. The molecular etiologies of AF are poorly understood, leaving patients with limited therapeutic options. AF has been recognized as an inherited disease in almost 30% of patient cases. However, few genetic loci have been identified and the mechanisms linking genetic variants to AF susceptibility remain unclear. By sequencing 193 probands with lone AF, we identified a Q76E variant within the coding sequence of the bone morphogenetic protein (BMP) antagonist gremlin-2 (GREM2) that increases its inhibitory activity. Functional modeling in zebrafish revealed that, through regulation of BMP signaling, GREM2 is required for cardiac laterality and atrial differentiation during embryonic development. GREM2 overactivity results in slower cardiac contraction rates in zebrafish, and induction of previously identified AF candidate genes encoding connexin-40, sarcolipin and atrial natriuretic peptide in differentiated mouse embryonic stem cells. By live heart imaging in zebrafish overexpressing wild-type or variant GREM2, we found abnormal contraction velocity specifically in atrial cardiomyocytes. These results implicate, for the first time, regulators of BMP signaling in human AF, providing mechanistic insights into the pathogenesis of the disease and identifying potential new therapeutic targets.


Subject(s)
Atrial Fibrillation/genetics , Cell Differentiation/genetics , Disease Models, Animal , Heart Atria/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Myocytes, Cardiac/pathology , Zebrafish Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Atrial Fibrillation/complications , Atrial Fibrillation/physiopathology , Bone Morphogenetic Proteins/metabolism , Cytokines , Female , Gene Expression Regulation, Developmental , Heart Atria/embryology , Heart Atria/pathology , Heart Rate/physiology , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Middle Aged , Molecular Sequence Data , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Pedigree , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
8.
Dev Biol ; 360(1): 173-85, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21963426

ABSTRACT

The neural crest is a stem cell-like population exclusive to vertebrates that gives rise to many different cell types including chondrocytes, neurons and melanocytes. Arising from the neural plate border at the intersection of Wnt and Bmp signaling pathways, the complexity of neural crest gene regulatory networks has made the earliest steps of induction difficult to elucidate. Here, we report that tfap2a and foxd3 participate in neural crest induction and are necessary and sufficient for this process to proceed. Double mutant tfap2a (mont blanc, mob) and foxd3 (mother superior, mos) mob;mos zebrafish embryos completely lack all neural crest-derived tissues. Moreover, tfap2a and foxd3 are expressed during gastrulation prior to neural crest induction in distinct, complementary, domains; tfap2a is expressed in the ventral non-neural ectoderm and foxd3 in the dorsal mesendoderm and ectoderm. We further show that Bmp signaling is expanded in mob;mos embryos while expression of dkk1, a Wnt signaling inhibitor, is increased and canonical Wnt targets are suppressed. These changes in Bmp and Wnt signaling result in specific perturbations of neural crest induction rather than general defects in neural plate border or dorso-ventral patterning. foxd3 overexpression, on the other hand, enhances the ability of tfap2a to ectopically induce neural crest around the neural plate, overriding the normal neural plate border limit of the early neural crest territory. Although loss of either Tfap2a or Foxd3 alters Bmp and Wnt signaling patterns, only their combined inactivation sufficiently alters these signaling gradients to abort neural crest induction. Collectively, our results indicate that tfap2a and foxd3, in addition to their respective roles in the differentiation of neural crest derivatives, also jointly maintain the balance of Bmp and Wnt signaling in order to delineate the neural crest induction domain.


Subject(s)
Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Transcription Factor AP-2/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Base Sequence , Body Patterning , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Death , DNA Primers/genetics , Embryonic Stem Cells/cytology , Forkhead Transcription Factors/genetics , Gastrulation , Gene Expression Regulation, Developmental , Genes, p53 , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Neural Crest/cytology , Neurogenesis , Transcription Factor AP-2/genetics , Wnt Signaling Pathway , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
G3 (Bethesda) ; 1(1): 3-9, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21949597

ABSTRACT

A surprising diversity of mechanisms controls sex determination of vertebrate organisms, even among closely related species. Both genetic and temperature-dependent systems of sex determination have been described in teleost fish. In the common zebrafish model organism, heteromorphic sex chromosomes are not observed, and the potential role of a genetic component of sex determination remains largely unknown. Here we report a genome-wide linkage study of sex determination in zebrafish using a novel SNP genetic map. We identified loci on zebrafish chromosomes 5 (LOD score 7.9) and 16 (LOD score 9.3) governing sex determination as a complex trait, rather than as an XY or ZW genetic system. Each of these loci contains a prominent candidate gene with a conserved role in sex determination across additional species that suggest potential mechanisms of sex determination in zebrafish. The chromosome 5 locus harbors dmrt1, a key gene in sex determination from fruit flies to humans; mutation of the human DMRT1 ortholog is a cause of complete sex reversal of XY individuals. The chromosome 16 locus harbors cyp21a2; mutation of the human CYP21A2 ortholog is one of the more common causes of pseudohermaphroditism. Mutation detection at each of these candidate genes within the zebrafish cross identified hypomorphic variants on the female-associated allele of each locus. The two loci together accounted for 16% of variance of the trait. Interacting environmental cues are likely to be an additional important component of sex determination in zebrafish.

10.
Dis Model Mech ; 4(6): 763-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21729877

ABSTRACT

Craniofacial and skeletal dysmorphologies account for the majority of birth defects. A number of the disease phenotypes have been attributed to abnormal synthesis, maintenance and composition of extracellular matrix (ECM), yet the molecular and cellular mechanisms causing these ECM defects remain poorly understood. The zebrafish feelgood mutant manifests a severely malformed head skeleton and shortened body length due to defects in the maturation stage of chondrocyte development. In vivo analyses reveal a backlog of type II and type IV collagens in rough endoplasmic reticulum (ER) similar to those found in coat protein II complex (COPII)-deficient cells. The feelgood mutation hinders collagen deposition in the ECM, but trafficking of small cargos and other large ECM proteins such as laminin to the extracellular space is unaffected. We demonstrate that the zebrafish feelgood mutation causes a single amino acid substitution within the DNA-binding domain of transcription factor Creb3l2. We show that Creb3l2 selectively regulates the expression of genes encoding distinct COPII proteins (sec23a, sec23b and sec24d) but find no evidence for its regulation of sec24c expression. Moreover, we did not detect activation of ER stress response genes despite intracellular accumulation of collagen and prominent skeletal defects. Promoter trans-activation assays show that the Creb3l2 feelgood variant is a hypomorphic allele that retains approximately 50% of its transcriptional activity. Transgenic rescue experiments of the feelgood phenotype restore craniofacial development, illustrating that a precise level of Creb3l2 transcriptional activity is essential for skeletogenesis. Our results indicate that Creb3l2 modulates the availability of COPII machinery in a tissue- and cargo-specific manner. These findings could lead to a better understanding of the etiology of human craniofacial and skeletal birth defects as well as adult-onset diseases that are linked to dysregulated ECM deposition, such as arthritis, fibrosis or osteoporosis.


Subject(s)
Bone and Bones/metabolism , COP-Coated Vesicles/metabolism , Extracellular Matrix Proteins/metabolism , Morphogenesis , Mutation/genetics , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/metabolism , Amino Acid Sequence , Animals , Base Sequence , Bone and Bones/embryology , Bone and Bones/pathology , Branchial Region/growth & development , Branchial Region/metabolism , Branchial Region/pathology , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/ultrastructure , Collagen Type II/metabolism , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Stress , Gene Knockdown Techniques , Genetic Loci/genetics , Glycosaminoglycans/metabolism , Melanosomes/metabolism , Melanosomes/pathology , Molecular Sequence Data , Notochord/metabolism , Notochord/pathology , Protein Transport , Transcription Factors/chemistry , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
11.
Dev Dyn ; 240(6): 1613-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21523853

ABSTRACT

Initial stages of embryonic development rely on rapid, synchronized cell divisions of the fertilized egg followed by a set of morphogenetic movements collectively called epiboly and gastrulation. Lzap is a putative tumor suppressor whose expression is lost in 30% of head and neck squamous cell carcinomas. Lzap activities include regulation of cell cycle progression and response to therapeutic agents. Here, we explore developmental roles of the lzap gene during zebrafish morphogenesis. Lzap is highly conserved among vertebrates and is maternally deposited. Expression is initially ubiquitous during gastrulation, and later becomes more prominent in the pharyngeal arches, digestive tract, and brain. Antisense morpholino-mediated depletion of Lzap resulted in delayed cell divisions and apoptosis during blastomere formation, resulting in fewer, larger cells. Cell cycle analysis suggested that Lzap loss in early embryonic cells resulted in a G2/M arrest. Furthermore, the Lzap-deficient embryos failed to initiate epiboly--the earliest morphogenetic movement in animal development--which has been shown to be dependent on cell adhesion and migration of epithelial sheets. Our results strongly implicate Lzap in regulation of cell cycle progression, adhesion and migratory activity of epithelial cell sheets during early development. These functions provide further insight into Lzap activity that may contribute not only to development, but also to tumor formation.


Subject(s)
Cell Cycle/genetics , Cell Movement/genetics , Nerve Tissue Proteins/physiology , Tumor Suppressor Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Zebrafish/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Genes, Tumor Suppressor/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Morphogenesis/genetics , Morphogenesis/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Sequence Homology, Amino Acid , Time Factors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Cell Adh Migr ; 5(2): 114-8, 2011.
Article in English | MEDLINE | ID: mdl-21178403

ABSTRACT

Extracellular matrix (ECM) proteins, cell adhesion molecules, cytokines, morphogens and membrane receptors are synthesized in the ER and transported through the Golgi complex to the cell surface and the extracellular space. The first leg in this journey from the ER to Golgi is facilitated by the Coat Protein II (COPII) vesicular carriers. Genetic defects in genes encoding various COPII components cause a broad spectrum of human diseases, from anemia to skeletal deformities. Here, we summarize our findings in zebrafish and discuss how mutations in COPII elements may cause specific cellular and developmental defects.


Subject(s)
COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , COP-Coated Vesicles/genetics , Carrier Proteins/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Humans , Protein Transport , Saccharomyces cerevisiae Proteins/metabolism , Vesicular Transport Proteins/genetics , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/genetics
13.
PLoS One ; 5(4): e10367, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20442775

ABSTRACT

Protein transport from endoplasmic reticulum (ER) to Golgi is primarily conducted by coated vesicular carriers such as COPII. Here, we describe zebrafish bulldog mutations that disrupt the function of the cargo adaptor Sec24D, an integral component of the COPII complex. We show that Sec24D is essential for secretion of cartilage matrix proteins, whereas the preceding development of craniofacial primordia and pre-chondrogenic condensations does not depend on this isoform. Bulldog chondrocytes fail to secrete type II collagen and matrilin to extracellular matrix (ECM), but membrane bound receptor beta1-Integrin and Cadherins appear to leave ER in Sec24D-independent fashion. Consequently, Sec24D-deficient cells accumulate proteins in the distended ER, although a subset of ER compartments and Golgi complexes as visualized by electron microscopy and NBD C(6)-ceramide staining appear functional. Consistent with the backlog of proteins in the ER, chondrocytes activate the ER stress response machinery and significantly upregulate BiP transcription. Failure of ECM secretion hinders chondroblast intercalations thus resulting in small and malformed cartilages and severe craniofacial dysmorphology. This defect is specific to Sec24D mutants since knockdown of Sec24C, a close paralog of Sec24D, does not result in craniofacial cartilage dysmorphology. However, craniofacial development in double Sec24C/Sec24D-deficient animals is arrested earlier than in bulldog/sec24d, suggesting that Sec24C can compensate for loss of Sec24D at initial stages of chondrogenesis, but Sec24D is indispensable for chondrocyte maturation. Our study presents the first developmental perspective on Sec24D function and establishes Sec24D as a strong candidate for cartilage maintenance diseases and craniofacial birth defects.


Subject(s)
Extracellular Matrix Proteins/metabolism , Morphogenesis , Vesicular Transport Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Bone and Bones , COP-Coated Vesicles , Cartilage , Chondrocytes , Chondrogenesis , Molecular Sequence Data , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...