Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Annu Rev Cell Dev Biol ; 38: 447-466, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35767871

ABSTRACT

Organoids are miniaturized and simplified versions of an organ produced in vitro from stem or progenitor cells. They are used as a model system consisting of multiple cell types forming an architecture relevant to the organ and carrying out the function of the organ. They are a useful tool to study development, homeostasis, regeneration, and disease. The imaging of organoids has become a pivotal method to visualize and understand their self-organization, symmetry breaking, growth, differentiation, and function. In this review, we discuss imaging methods, how to analyze these images, and challenges in organoid research.


Subject(s)
Organoids , Stem Cells , Cell Differentiation
2.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-36314540

ABSTRACT

Organoids have become one of the fastest progressing and applied models in biological and medical research, and various organoids have now been developed for most of the organs of the body. Here, we review the methods developed to generate pancreas organoids in vitro from embryonic, fetal and adult cells, as well as pluripotent stem cells. We discuss how these systems have been used to learn new aspects of pancreas development, regeneration and disease, as well as their limitations and potential for future discoveries.


Subject(s)
Biomedical Research , Pluripotent Stem Cells , Organoids , Organogenesis , Pancreas
3.
Development ; 142(5): 858-70, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25715394

ABSTRACT

In human, mutations in bicaudal C1 (BICC1), an RNA binding protein, have been identified in patients with kidney dysplasia. Deletion of Bicc1 in mouse leads to left-right asymmetry randomization and renal cysts. Here, we show that BICC1 is also expressed in both the pancreatic progenitor cells that line the ducts during development, and in the ducts after birth, but not in differentiated endocrine or acinar cells. Genetic inactivation of Bicc1 leads to ductal cell over-proliferation and cyst formation. Transcriptome comparison between WT and Bicc1 KO pancreata, before the phenotype onset, reveals that PKD2 functions downstream of BICC1 in preventing cyst formation in the pancreas. Moreover, the analysis highlights immune cell infiltration and stromal reaction developing early in the pancreas of Bicc1 knockout mice. In addition to these functions in duct morphogenesis, BICC1 regulates NEUROG3(+) endocrine progenitor production. Its deletion leads to a late but sustained endocrine progenitor decrease, resulting in a 50% reduction of endocrine cells. We show that BICC1 functions downstream of ONECUT1 in the pathway controlling both NEUROG3(+) endocrine cell production and ductal morphogenesis, and suggest a new candidate gene for syndromes associating kidney dysplasia with pancreatic disorders, including diabetes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Fluorescent Antibody Technique , Genotype , Hepatocyte Nuclear Factor 6/genetics , In Situ Nick-End Labeling , Mice , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Stem Cells/cytology , Stem Cells/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
4.
PLoS Biol ; 13(3): e1002111, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25786211

ABSTRACT

Organogenesis relies on the spatiotemporal balancing of differentiation and proliferation driven by an expanding pool of progenitor cells. In the mouse pancreas, lineage tracing at the population level has shown that the expanding pancreas progenitors can initially give rise to all endocrine, ductal, and acinar cells but become bipotent by embryonic day 13.5, giving rise to endocrine cells and ductal cells. However, the dynamics of individual progenitors balancing self-renewal and lineage-specific differentiation has never been described. Using three-dimensional live imaging and in vivo clonal analysis, we reveal the contribution of individual cells to the global behaviour and demonstrate three modes of progenitor divisions: symmetric renewing, symmetric endocrinogenic, and asymmetric generating a progenitor and an endocrine progenitor. Quantitative analysis shows that the endocrine differentiation process is consistent with a simple model of cell cycle-dependent stochastic priming of progenitors to endocrine fate. The findings provide insights to define control parameters to optimize the generation of ß-cells in vitro.


Subject(s)
Acinar Cells/cytology , Cell Cycle/genetics , Cell Lineage/genetics , Insulin-Secreting Cells/cytology , Stem Cells/cytology , Acinar Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Cell Tracking , Embryo, Mammalian , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Image Processing, Computer-Assisted , Insulin-Secreting Cells/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organogenesis/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Tissue Culture Techniques , Trans-Activators/genetics , Trans-Activators/metabolism , Red Fluorescent Protein
5.
Development ; 141(5): 1120-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24550118

ABSTRACT

Coordinated arterial-venous differentiation is crucial for vascular development and function. The origin of the cardinal vein (CV) in mammals is unknown, while conflicting theories have been reported in chick and zebrafish. Here, we provide the first molecular characterization of endothelial cells (ECs) expressing venous molecular markers, or venous-fated ECs, within the emergent dorsal aorta (DA). These ECs, expressing the venous molecular markers Coup-TFII and EphB4, cohabited the early DA with ECs expressing the arterial molecular markers ephrin B2, Notch and connexin 40. These mixed ECs in the early DA expressed either the arterial or venous molecular marker, but rarely both. Subsequently, the DA exhibited uniform arterial markers. Real-time imaging of mouse embryos revealed EC movement from the DA to the CV during the stage when venous-fated ECs occupied the DA. We analyzed mutants for EphB4, which encodes a receptor tyrosine kinase for the ephrin B2 ligand, as we hypothesized that ephrin B2/EphB4 signaling may mediate the repulsion of venous-fated ECs from the DA to the CV. Using an EC quantification approach, we discovered that venous-fated ECs increased in the DA and decreased in the CV in the mutants, whereas the rest of the ECs in each vessel were unaffected. This result suggests that the venous-fated ECs were retained in the DA and missing in the CV in the EphB4 mutant, and thus that ephrin B2/EphB4 signaling normally functions to clear venous-fated ECs from the DA to the CV by cell repulsion. Therefore, our cellular and molecular evidence suggests that the DA harbors venous progenitors that move to participate in CV formation, and that ephrin B2/EphB4 signaling regulates this aortic contribution to the mammalian CV.


Subject(s)
Aorta/cytology , Stem Cells/cytology , Veins/cytology , Animals , Endothelial Cells/cytology , Endothelial Cells/metabolism , Mice , Mice, Transgenic , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Stem Cells/metabolism
6.
Dev Biol ; 405(2): 316-27, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26156633

ABSTRACT

To contribute to devise successful beta-cell differentiation strategies for the cure of Type 1 diabetes we sought to uncover barriers that restrict endocrine fate acquisition by studying the role of the transcriptional repressor REST in the developing pancreas. Rest expression is prevented in neurons and in endocrine cells, which is necessary for their normal function. During development, REST represses a subset of genes in the neuronal differentiation program and Rest is down-regulated as neurons differentiate. Here, we investigate the role of REST in the differentiation of pancreatic endocrine cells, which are molecularly close to neurons. We show that Rest is widely expressed in pancreas progenitors and that it is down-regulated in differentiated endocrine cells. Sustained expression of REST in Pdx1(+) progenitors impairs the differentiation of endocrine-committed Neurog3(+) progenitors, decreases beta and alpha cell mass by E18.5, and triggers diabetes in adulthood. Conditional inactivation of Rest in Pdx1(+) progenitors is not sufficient to trigger endocrine differentiation but up-regulates a subset of differentiation genes. Our results show that the transcriptional repressor REST is active in pancreas progenitors where it gates the activation of part of the beta cell differentiation program.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Pancreas/metabolism , Repressor Proteins/physiology , Animals , Blood Glucose/metabolism , Down-Regulation , Endocrine Cells/cytology , Endocrine Cells/metabolism , Endocrine System/metabolism , Gene Deletion , Homeodomain Proteins/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Pancreas/embryology , Stem Cells/cytology , Trans-Activators/metabolism , Transgenes
8.
Dev Cell ; 58(21): 2292-2308.e6, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37591246

ABSTRACT

Basic helix-loop-helix genes, particularly proneural genes, are well-described triggers of cell differentiation, yet information on their dynamics is limited, notably in human development. Here, we focus on Neurogenin 3 (NEUROG3), which is crucial for pancreatic endocrine lineage initiation. By monitoring both NEUROG3 gene expression and protein in single cells using a knockin dual reporter in 2D and 3D models of human pancreas development, we show an approximately 2-fold slower expression of human NEUROG3 than that of the mouse. We observe heterogeneous peak levels of NEUROG3 expression and reveal through long-term live imaging that both low and high NEUROG3 peak levels can trigger differentiation into hormone-expressing cells. Based on fluorescence intensity, we statistically integrate single-cell transcriptome with dynamic behaviors of live cells and propose a data-mapping methodology applicable to other contexts. Using this methodology, we identify a role for KLK12 in motility at the onset of NEUROG3 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Nerve Tissue Proteins , Humans , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreas/metabolism , Cell Differentiation/genetics , Endocrine System/metabolism
9.
Dev Dyn ; 240(3): 589-604, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21287656

ABSTRACT

During development, pancreatic endocrine cells are specified within the pancreatic epithelium. They subsequently delaminate out of the epithelium and cluster in the mesenchyme to form the islets of Langerhans. Neurogenin3 (Ngn3) is a transcription factor required for the differentiation of all endocrine cells and we investigated its role in their delamination. We observed in the mouse pancreas that most Ngn3-positive cells have lost contact with the lumen of the epithelium, showing that the delamination from the progenitor layer is initiated in endocrine progenitors. Subsequently, in both mouse and chick newly born endocrine cells at the periphery of the epithelium strongly decrease E-cadherin, break-down the basal lamina and cluster into islets of Langerhans. Repression of E-cadherin is sufficient to promote delamination from the epithelium. We further demonstrate that Ngn3 indirectly controls Snail2 protein expression post-transcriptionally to repress E-cadherin. In the chick embryo, Ngn3 independently controls epithelium delamination and differentiation programs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Insulin-Secreting Cells/cytology , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cadherins/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Polarity/genetics , Cell Polarity/physiology , Chick Embryo , Chickens , Electroporation , Female , Fluorescent Antibody Technique , In Situ Hybridization , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Islets of Langerhans/metabolism , Mice , Nerve Tissue Proteins/genetics , Pancreas/metabolism , Pregnancy
10.
Stem Cell Reports ; 17(5): 1215-1228, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35452596

ABSTRACT

With the aim of producing ß cells for replacement therapies to treat diabetes, several protocols have been developed to differentiate human pluripotent stem cells to ß cells via pancreatic progenitors. While in vivo pancreatic progenitors expand throughout development, the in vitro protocols have been designed to make these cells progress as fast as possible to ß cells. Here, we report on a protocol enabling a long-term expansion of human pancreatic progenitors in a defined medium on fibronectin, in the absence of feeder layers. Moreover, through a screening of a polymer library we identify a polymer that can replace fibronectin. Our experiments, comparing expanded progenitors to directly differentiated progenitors, show that the expanded progenitors differentiate more efficiently into glucose-responsive ß cells and produce fewer glucagon-expressing cells. The ability to expand progenitors under defined conditions and cryopreserve them will provide flexibility in research and therapeutic production.


Subject(s)
Insulin-Secreting Cells , Pluripotent Stem Cells , Cell Differentiation , Fibronectins/pharmacology , Humans , Pancreas , Polymers
11.
J Cell Biol ; 172(1): 151-62, 2006 Jan 02.
Article in English | MEDLINE | ID: mdl-16391003

ABSTRACT

Morphogenesis of a vascular network requires dynamic vessel growth and regression. To investigate the cellular mechanism underlying this process, we deleted focal adhesion kinase (FAK), a key signaling mediator, in endothelial cells (ECs) using Tie2-Cre mice. Targeted FAK depletion occurred efficiently early in development, where mutants exhibited a distinctive and irregular vasculature, resulting in hemorrhage and lethality between embryonic day (e) 10.5 and 11.5. Capillaries and intercapillary spaces in yolk sacs were dilated before any other detectable abnormalities at e9.5, and explants demonstrate that the defects resulted from the loss of FAK and not from organ failure. Time-lapse microscopy monitoring EC behavior during vascular formation in explants revealed no apparent decrease in proliferation or migration but revealed increases in cell retraction and death leading to reduced vessel growth and increased vessel regression. Consistent with this phenotype, ECs derived from mutant embryos exhibited aberrant lamellipodial extensions, altered actin cytoskeleton, and nonpolarized cell movement. This study reveals that FAK is crucial for vascular morphogenesis and the regulation of EC survival and morphology.


Subject(s)
Blood Vessels/abnormalities , Cardiovascular Abnormalities/enzymology , Cardiovascular Abnormalities/genetics , Endothelium, Vascular/enzymology , Focal Adhesion Kinase 1/deficiency , Pseudopodia/genetics , Animals , Blood Vessels/pathology , Capillaries/abnormalities , Capillaries/pathology , Cardiovascular Abnormalities/pathology , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Embryo, Mammalian/blood supply , Embryo, Mammalian/pathology , Endothelium, Vascular/pathology , Focal Adhesion Kinase 1/genetics , Integrases/genetics , Mice , Mice, Transgenic , Mutation , Neovascularization, Physiologic/genetics , Pseudopodia/pathology , Receptor, TIE-2/genetics
12.
J Mol Med (Berl) ; 99(4): 449-462, 2021 04.
Article in English | MEDLINE | ID: mdl-33221939

ABSTRACT

Organoids constitute biological systems which are used to model organ development, homeostasis, regeneration, and disease in vitro and hold promise for use in therapy. Reflecting in vivo development, organoids form from tissue cells or pluripotent stem cells. Cues provided from the media and individual cells promote self-organization of these uniform starting cells into a structure, with emergent differentiated cells, morphology, and often functionality that resemble the tissue of origin. Therefore, organoids provide a complement to two-dimensional in vitro culture and in vivo animal models of development, providing the experimental control and flexibility of in vitro methods with the three-dimensional context of in vivo models, with fewer ethical restraints than human or animal work. However, using organoids, we are only just beginning to understand on the cellular level how the external conditions and signaling between individual cells promote the emergence of cells and structures. In this review, we focus specifically on organoids derived from endodermal tissues: the starting conditions of the cells, signaling mechanisms, and external media that allow the emergence of higher order self-organization.


Subject(s)
Endoderm/cytology , Organoids/cytology , Adult Stem Cells/cytology , Animals , Cell Communication , Cell Culture Techniques/methods , Cell Differentiation , Feedback, Physiological , Humans , Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Mice , Morphogenesis , Organ Specificity , Organogenesis , Organoids/ultrastructure , Signal Transduction
13.
Nat Commun ; 12(1): 3144, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035279

ABSTRACT

Human organogenesis remains relatively unexplored for ethical and practical reasons. Here, we report the establishment of a single-cell transcriptome atlas of the human fetal pancreas between 7 and 10 post-conceptional weeks of development. To interrogate cell-cell interactions, we describe InterCom, an R-Package we developed for identifying receptor-ligand pairs and their downstream effects. We further report the establishment of a human pancreas culture system starting from fetal tissue or human pluripotent stem cells, enabling the long-term maintenance of pancreas progenitors in a minimal, defined medium in three-dimensions. Benchmarking the cells produced in 2-dimensions and those expanded in 3-dimensions to fetal tissue identifies that progenitors expanded in 3-dimensions are transcriptionally closer to the fetal pancreas. We further demonstrate the potential of this system as a screening platform and identify the importance of the EGF and FGF pathways controlling human pancreas progenitor expansion.


Subject(s)
Cell Culture Techniques/methods , Organogenesis , Pancreas/embryology , Pluripotent Stem Cells/physiology , Tissue Culture Techniques/methods , Aborted Fetus , Animals , Cell Communication , Cell Differentiation , Cell Line , Datasets as Topic , Embryo, Mammalian , Epidermal Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Pancreas/cytology , RNA-Seq , Signal Transduction/physiology , Single-Cell Analysis , Spheroids, Cellular , Transcriptome
14.
Curr Top Dev Biol ; 129: 143-190, 2018.
Article in English | MEDLINE | ID: mdl-29801529

ABSTRACT

Here, we review how human pluripotent stem cell models of pancreas development have emerged and became an important tool to study human development and disease. Initially developed toward the production of ß cells for diabetes therapy, the protocols have been refined based on knowledge of pancreas development in model organisms. While the cells produced are closer and closer to the end goal of a functional ß cell, these models have also been used to carry out functional experiments addressing gene function and expression as well as regulatory and epigenetic landscape changes during human pancreas development. They thereby complement model organisms and reports from human genetic variants predisposing to different forms of diabetes, as well as observations on human fetal tissue. In this review, we therefore compare these different sources of information and discuss how human stem cell models are evolving to inform us on pancreatic diseases and possible treatments.


Subject(s)
Pancreas/embryology , Pluripotent Stem Cells/cytology , Body Patterning , Cell Differentiation , Cell Lineage , Endoderm/embryology , Humans , Pancreas/cytology
15.
Nat Commun ; 8(1): 605, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928395

ABSTRACT

Spatiotemporal balancing of cellular proliferation and differentiation is crucial for postnatal tissue homoeostasis and organogenesis. During embryonic development, pancreatic progenitors simultaneously proliferate and differentiate into the endocrine, ductal and acinar lineages. Using in vivo clonal analysis in the founder population of the pancreas here we reveal highly heterogeneous contribution of single progenitors to organ formation. While some progenitors are bona fide multipotent and contribute progeny to all major pancreatic cell lineages, we also identify numerous unipotent endocrine and ducto-endocrine bipotent clones. Single-cell transcriptional profiling at E9.5 reveals that endocrine-committed cells are molecularly distinct, whereas multipotent and bipotent progenitors do not exhibit different expression profiles. Clone size and composition support a probabilistic model of cell fate allocation and in silico simulations predict a transient wave of acinar differentiation around E11.5, while endocrine differentiation is proportionally decreased. Increased proliferative capacity of outer progenitors is further proposed to impact clonal expansion.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Organogenesis , Pancreas/embryology , Acinar Cells , Animals , Computer Simulation , Gene Expression Profiling , Mice , Single-Cell Analysis
16.
Development ; 135(12): 2193-202, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18480158

ABSTRACT

beta1 integrin (encoded by Itgb1) is established as a regulator of angiogenesis based upon the phenotypes of complete knockouts of beta1 heterodimer partners or ligands and upon antibody inhibition studies in mice. Its direct function in endothelial cells (ECs) in vivo has not been determined because Itgb1(-/-) embryos die before vascular development. Excision of Itgb1 from ECs and a subset of hematopoietic cells, using Tie2-Cre, resulted in abnormal vascular development by embryonic day (e) 8.5 and lethality by e10.5. Tie1-Cre mediated a more restricted excision of Itgb1 from ECs and hematopoietic cells and resulted in embryonic lethal vascular defects by e11.5. Capillaries of the yolk sacs were disorganized, and the endothelium of major blood vessels and of the heart was frequently discontinuous in mutant embryos. We also found similar vascular morphogenesis defects characterized by EC disorganization in embryonic explants and isolated ECs. Itgb1-null ECs were deficient in adhesion and migration in a ligand-specific fashion, with impaired responses to laminin and collagens, but not to fibronectin. Deletion of Itgb1 reduced EC survival, but did not affect proliferation. Our findings demonstrate that beta1 integrin is essential for EC adhesion, migration and survival during angiogenesis, and further validate that therapies targeting beta1 integrins may effectively impair neovascularization.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Endothelial Cells/physiology , Integrin beta1/physiology , Neovascularization, Physiologic/physiology , Animals , Cell Survival , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Immunomagnetic Separation , Integrin beta1/genetics , Mice , Mice, Transgenic , Models, Cardiovascular
17.
Development ; 135(22): 3755-64, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18952909

ABSTRACT

A mutual coordination of size between developing arteries and veins is essential for establishing proper connections between these vessels and, ultimately, a functional vasculature; however, the cellular and molecular regulation of this parity is not understood. Here, we demonstrate that the size of the developing dorsal aorta and cardinal vein is reciprocally balanced. Mouse embryos carrying gain-of-function Notch alleles show enlarged aortae and underdeveloped cardinal veins, whereas those with loss-of-function mutations show small aortae and large cardinal veins. Notch does not affect the overall number of endothelial cells but balances the proportion of arterial to venous endothelial cells, thereby modulating the relative sizes of both vessel types. Loss of ephrin B2 or its receptor EphB4 also leads to enlarged aortae and underdeveloped cardinal veins; however, endothelial cells with venous identity are mislocalized in the aorta, suggesting that ephrin B2/EphB4 signaling functions distinctly from Notch by sorting arterial and venous endothelial cells into their respective vessels. Our findings provide mechanistic insight into the processes underlying artery and vein size equilibration during angiogenesis.


Subject(s)
Arteries/metabolism , Ephrin-B2/metabolism , Neovascularization, Physiologic , Proto-Oncogene Proteins/metabolism , Receptor, EphB4/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Veins/metabolism , Animals , Arteries/anatomy & histology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Endothelial Cells/metabolism , Ephrin-B2/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Receptor, EphB4/genetics , Receptor, Notch1/deficiency , Receptor, Notch1/genetics , Receptor, Notch4 , Receptors, Notch/genetics , Veins/anatomy & histology
18.
Biochemistry ; 41(47): 14076-84, 2002 Nov 26.
Article in English | MEDLINE | ID: mdl-12437365

ABSTRACT

Crystallins, the major structural proteins in the lens of the eye, are maintained with little turnover throughout the lifetime of the host. With time, lens crystallins undergo post-translational modifications that may play an important role in loss of vision during aging and cataract formation. Specific modifications include deamidation and truncation. Urea-induced denaturation was studied for recombinantly expressed wild-type betaB1 (WT), the deamidated mutant (Q204E), an N-terminally truncated mutant (betaB1(DeltaN41)), and other truncated versions of these proteins generated by calpain II digestion. Tryptophan fluorescence was used to monitor loss of global tertiary structure. Loss of secondary structure was followed by circular dichroism, and electron paramagnetic resonance site-directed spin labeling was used to monitor loss of tertiary structure selectively in the N-terminal domain. Our results indicated that the deamidated mutant was significantly destabilized relative to WT. Q204E showed a two-step denaturation curve with transitions at 4.1 and 7.2 M urea, whereas denaturation of WT occurred in a cooperative single step with a transition midpoint of 5.9 M urea. Unfolding of WT was completely reversible, whereas Q204E failed to fully refold. Prolonged incubation under denaturing conditions led to aggregation, which was also more pronounced for Q204E dimers than for WT. Truncation of 41 residues from the N-terminus or 47 and 5 residues from the N- and C-termini did not affect stability. These studies indicated that a single-site deamidation could significantly diminish the stability of lens betaB1-crystallin, supporting the idea that such modifications may play an important role in age-related cataract formation.


Subject(s)
Crystallins/chemistry , Amino Acid Substitution , Circular Dichroism , Cloning, Molecular , Crystallins/genetics , Crystallins/metabolism , Drug Stability , Electron Spin Resonance Spectroscopy , Escherichia coli/genetics , Lens, Crystalline/chemistry , Lens, Crystalline/physiology , Mutagenesis, Site-Directed , Protein Conformation , Protein Denaturation , Protein Structure, Secondary , Sequence Deletion , Urea
SELECTION OF CITATIONS
SEARCH DETAIL