Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
PLoS One ; 18(10): e0291512, 2023.
Article in English | MEDLINE | ID: mdl-37796967

ABSTRACT

Proper maintenance of mature cellular phenotypes is essential for stable physiology, suppression of disease states, and resistance to oncogenic transformation. We describe the transcriptional regulatory roles of four key DNA-binding transcription factors (Ptf1a, Nr5a2, Foxa2 and Gata4) that sit at the top of a regulatory hierarchy controlling all aspects of a highly differentiated cell-type-the mature pancreatic acinar cell (PAC). Selective inactivation of Ptf1a, Nr5a2, Foxa2 and Gata4 individually in mouse adult PACs rapidly altered the transcriptome and differentiation status of PACs. The changes most emphatically included transcription of the genes for the secretory digestive enzymes (which conscript more than 90% of acinar cell protein synthesis), a potent anabolic metabolism that provides the energy and materials for protein synthesis, suppressed and properly balanced cellular replication, and susceptibility to transformation by oncogenic KrasG12D. The simultaneous inactivation of Foxa2 and Gata4 caused a greater-than-additive disruption of gene expression and uncovered their collaboration to maintain Ptf1a expression and control PAC replication. A measure of PAC dedifferentiation ranked the effects of the conditional knockouts as Foxa2+Gata4 > Ptf1a > Nr5a2 > Foxa2 > Gata4. Whereas the loss of Ptf1a or Nr5a2 greatly accelerated Kras-mediated transformation of mature acinar cells in vivo, the absence of Foxa2, Gata4, or Foxa2+Gata4 together blocked transformation completely, despite extensive dedifferentiation. A lack of correlation between PAC dedifferentiation and sensitivity to oncogenic KrasG12D negates the simple proposition that the level of differentiation determines acinar cell resistance to transformation.


Subject(s)
Pancreas, Exocrine , Pancreatic Neoplasms , Mice , Animals , Acinar Cells/metabolism , Epithelium/metabolism , Transcription Factors/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Phenotype , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
2.
Cell Stem Cell ; 30(4): 488-497.e3, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37028408

ABSTRACT

Understanding the origin of pancreatic ß cells has profound implications for regenerative therapies in diabetes. For over a century, it was widely held that adult pancreatic duct cells act as endocrine progenitors, but lineage-tracing experiments challenged this dogma. Gribben et al. recently used two existing lineage-tracing models and single-cell RNA sequencing to conclude that adult pancreatic ducts contain endocrine progenitors that differentiate to insulin-expressing ß cells at a physiologically important rate. We now offer an alternative interpretation of these experiments. Our data indicate that the two Cre lines that were used directly label adult islet somatostatin-producing ∂ cells, which precludes their use to assess whether ß cells originate from duct cells. Furthermore, many labeled ∂ cells, which have an elongated neuron-like shape, were likely misclassified as ß cells because insulin-somatostatin coimmunolocalizations were not used. We conclude that most evidence so far indicates that endocrine and exocrine lineage borders are rarely crossed in the adult pancreas.


Subject(s)
Insulin-Secreting Cells , Evidence Gaps , Cell Differentiation , Pancreas/physiology , Pancreatic Ducts , Insulin , Somatostatin
3.
Cell Metab ; 34(11): 1779-1791.e9, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36240759

ABSTRACT

Microbiome dysbiosis is a feature of diabetes, but how microbial products influence insulin production is poorly understood. We report the mechanism of BefA, a microbiome-derived protein that increases proliferation of insulin-producing ß cells during development in gnotobiotic zebrafish and mice. BefA disseminates systemically by multiple anatomic routes to act directly on pancreatic islets. We detail BefA's atomic structure, containing a lipid-binding SYLF domain, and demonstrate that it permeabilizes synthetic liposomes and bacterial membranes. A BefA mutant impaired in membrane disruption fails to expand ß cells, whereas the pore-forming host defense protein, Reg3, stimulates ß cell proliferation. Our work demonstrates that membrane permeabilization by microbiome-derived and host defense proteins is necessary and sufficient for ß cell expansion during pancreas development, potentially connecting microbiome composition with diabetes risk.


Subject(s)
Diabetes Mellitus , Microbiota , Mice , Animals , Zebrafish , Pancreas/metabolism , Insulin/metabolism , Diabetes Mellitus/metabolism , Proteins/metabolism
4.
Neurology ; 2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34135078

ABSTRACT

OBJECTIVE: To identify novel disease associated loci for amyotrophic lateral sclerosis (ALS), we utilized sequencing data and performed in vitro and in vivo experiments to demonstrate pathogenicity of mutations identified in TP73. METHODS: We analyzed exome sequences of 87 sporadic ALS patients and 324 controls, with confirmatory sequencing in independent ALS cohorts of >2,800 patients. For the top hit, TP73, a regulator of apoptosis, differentiation, and a binding partner as well as homolog of the tumor suppressor gene TP53, we assayed mutation effects using in vitro and in vivo experiments. C2C12 myoblast differentiation assays, characterization of myotube appearance, and immunoprecipitation of p53-p73 complexes were perform in vitro. In vivo, we used CRISPR/Cas9 targeting of zebrafish tp73 to assay motor neuron number and axon morphology. RESULTS: Five heterozygous rare, nonsynonymous mutations in TP73 were identified in our sporadic ALS cohort. In independent ALS cohorts, we identified an additional 19 rare, deleterious variants in TP73. Patient TP73 mutations caused abnormal differentiation and increased apoptosis in the myoblast differentiation assay, with abnormal myotube appearance. Immunoprecipitation of mutant ΔN-p73 demonstrated that patient mutations hinder ΔN-p73's ability to bind p53. CRISPR/Cas9 knockout of tp73 in zebrafish led to impaired motor neuron development and abnormal axonal morphology, concordant with ALS pathology. CONCLUSION: Together, these results strongly suggest that variants in TP73 correlate with risk for ALS and indicate a novel role for apoptosis in ALS disease pathology.

5.
Cell Metab ; 31(2): 284-300.e7, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31813825

ABSTRACT

Although metabolic adaptations have been demonstrated to be essential for tumor cell proliferation, the metabolic underpinnings of tumor initiation are poorly understood. We found that the earliest stages of colorectal cancer (CRC) initiation are marked by a glycolytic metabolic signature, including downregulation of the mitochondrial pyruvate carrier (MPC), which couples glycolysis and glucose oxidation through mitochondrial pyruvate import. Genetic studies in Drosophila suggest that this downregulation is required because hyperplasia caused by loss of the Apc or Notch tumor suppressors in intestinal stem cells can be completely blocked by MPC overexpression. Moreover, in two distinct CRC mouse models, loss of Mpc1 prior to a tumorigenic stimulus doubled the frequency of adenoma formation and produced higher grade tumors. MPC loss was associated with a glycolytic metabolic phenotype and increased expression of stem cell markers. These data suggest that changes in cellular pyruvate metabolism are necessary and sufficient to promote cancer initiation.


Subject(s)
Adenoma/metabolism , Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Pyruvic Acid/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Drosophila , Female , Male , Mice , Mice, Inbred C57BL
6.
Dev Cell ; 50(6): 744-754.e4, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31422917

ABSTRACT

Activating mutations in Kras are nearly ubiquitous in human pancreatic cancer and initiate precancerous pancreatic intraepithelial neoplasia (PanINs) when induced in mouse acinar cells. PanINs normally take months to form but are accelerated by deletion of acinar cell differentiation factors such as Ptf1a, suggesting that loss of cell identity is rate limiting for pancreatic tumor initiation. Using a genetic mouse model that allows for independent control of oncogenic Kras and Ptf1a expression, we demonstrate that sustained Ptf1a is sufficient to prevent Kras-driven tumorigenesis, even in the presence of tumor-promoting inflammation. Furthermore, reintroducing Ptf1a into established PanINs reverts them to quiescent acinar cells in vivo. Similarly, Ptf1a re-expression in human pancreatic cancer cells inhibits their growth and colony-forming ability. Our results suggest that reactivation of an endogenous differentiation program can prevent and reverse oncogene-driven transformation in cells harboring tumor-driving mutations, introducing a potential paradigm for solid tumor prevention and treatment.


Subject(s)
Carcinogenesis/pathology , Cell Differentiation , Pancreatic Neoplasms/pathology , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Clone Cells , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Inflammation/pathology , Mice , Pancreatic Neoplasms/genetics , Pancreatitis/pathology , Phenotype , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Transcription Factors/metabolism
7.
Nature ; 554(7693): 470-472, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29469130
8.
Nature ; 554(7693): 470-472, 2018 Feb.
Article in English | MEDLINE | ID: mdl-32094929
9.
Nature ; 531(7592): 47-52, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26909576

ABSTRACT

Integrated genomic analysis of 456 pancreatic ductal adenocarcinomas identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-ß, WNT, NOTCH, ROBO/SLIT signalling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing. Expression analysis defined 4 subtypes: (1) squamous; (2) pancreatic progenitor; (3) immunogenic; and (4) aberrantly differentiated endocrine exocrine (ADEX) that correlate with histopathological characteristics. Squamous tumours are enriched for TP53 and KDM6A mutations, upregulation of the TP63∆N transcriptional network, hypermethylation of pancreatic endodermal cell-fate determining genes and have a poor prognosis. Pancreatic progenitor tumours preferentially express genes involved in early pancreatic development (FOXA2/3, PDX1 and MNX1). ADEX tumours displayed upregulation of genes that regulate networks involved in KRAS activation, exocrine (NR5A2 and RBPJL), and endocrine differentiation (NEUROD1 and NKX2-2). Immunogenic tumours contained upregulated immune networks including pathways involved in acquired immune suppression. These data infer differences in the molecular evolution of pancreatic cancer subtypes and identify opportunities for therapeutic development.


Subject(s)
Genes, Neoplasm/genetics , Genome, Human/genetics , Genomics , Mutation/genetics , Pancreatic Neoplasms/classification , Pancreatic Neoplasms/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Pancreatic Ductal/classification , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Histone Demethylases/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Receptors, Cytoplasmic and Nuclear/genetics , Survival Analysis , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Zebrafish Proteins
10.
Trends Cancer ; 2(12): 723-735, 2016 12.
Article in English | MEDLINE | ID: mdl-28630946

ABSTRACT

While recent studies demonstrate that cancer can arise from mutant stem cells, this hypothesis does not explain why tissues without defined stem cell populations are susceptible to inflammation-driven tumorigenesis. We propose that chronic inflammatory diseases, such as colitis and pancreatitis, predispose to gastrointestinal (GI) adenocarcinoma by reprogramming differentiated cells. Focusing on colon and pancreas, we discuss recently discovered connections between inflammation and loss of cell differentiation, and propose that dysregulation of cell fate may be a novel rate-limiting step of tumorigenesis. We review studies identifying differentiation mechanisms that limit tumor initiation and that, upon reactivation, can prevent or revert the cancer cell transformed phenotype. Together, these findings suggest that differentiation-targeted treatments hold promise as a therapeutic strategy in GI cancer.


Subject(s)
Gastrointestinal Neoplasms , Animals , Carcinogenesis , Cell Differentiation , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/pathology , Humans , Inflammation/complications , Stem Cells/pathology
11.
Elife ; 42015 Jul 07.
Article in English | MEDLINE | ID: mdl-26151762

ABSTRACT

Understanding the initiation and progression of pancreatic ductal adenocarcinoma (PDAC) may provide therapeutic strategies for this deadly disease. Recently, we and others made the surprising finding that PDAC and its preinvasive precursors, pancreatic intraepithelial neoplasia (PanIN), arise via reprogramming of mature acinar cells. We therefore hypothesized that the master regulator of acinar differentiation, PTF1A, could play a central role in suppressing PDAC initiation. In this study, we demonstrate that PTF1A expression is lost in both mouse and human PanINs, and that this downregulation is functionally imperative in mice for acinar reprogramming by oncogenic KRAS. Loss of Ptf1a alone is sufficient to induce acinar-to-ductal metaplasia, potentiate inflammation, and induce a KRAS-permissive, PDAC-like gene expression profile. As a result, Ptf1a-deficient acinar cells are dramatically sensitized to KRAS transformation, and reduced Ptf1a greatly accelerates development of invasive PDAC. Together, these data indicate that cell differentiation regulators constitute a new tumor suppressive mechanism in the pancreas.


Subject(s)
Acinar Cells/physiology , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Transdifferentiation , Transcription Factors/analysis , Animals , Carcinoma in Situ/pathology , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Transcription Factors/genetics
12.
Am J Pathol ; 185(1): 197-213, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25451153

ABSTRACT

Wnt glycoproteins control key processes during development and disease by activating various downstream pathways. Wnt secretion requires post-translational modification mediated by the O-acyltransferase encoded by the Drosophila porcupine homolog gene (PORCN). In humans, PORCN mutations cause focal dermal hypoplasia (FDH, or Goltz syndrome), an X-linked dominant multisystem birth defect that is frequently accompanied by ocular abnormalities such as coloboma, microphthalmia, or even anophthalmia. Although genetic ablation of Porcn in mouse has provided insight into the etiology of defects caused by ectomesodermal dysplasia in FDH, the requirement for Porcn and the actual Wnt ligands during eye development have been unknown. In this study, Porcn hemizygosity occasionally caused ocular defects reminiscent of FDH. Conditional inactivation of Porcn in periocular mesenchyme led to defects in mid- and hindbrain and in craniofacial development, but was insufficient to cause ocular abnormalities. However, a combination of conditional Porcn depletion in optic vesicle neuroectoderm, lens, and neural crest-derived periocular mesenchyme induced severe eye abnormalities with high penetrance. In particular, we observed coloboma, transdifferentiation of the dorsal and ventral retinal pigment epithelium, defective optic cup periphery, and closure defects of the eyelid, as well as defective corneal morphogenesis. Thus, Porcn is required in both extraocular and neuroectodermal tissues to regulate distinct Wnt-dependent processes during morphogenesis of the posterior and anterior segments of the eye.


Subject(s)
Eye/embryology , Focal Dermal Hypoplasia/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Acyltransferases , Alleles , Animals , Disease Models, Animal , Eye/metabolism , Female , Genotype , Glycoproteins/metabolism , Hemizygote , In Situ Hybridization , Ligands , Male , Mice , Mice, Inbred C57BL , Mutation , Recombination, Genetic , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/metabolism , Wnt Proteins/metabolism
13.
Annu Rev Physiol ; 77: 229-49, 2015.
Article in English | MEDLINE | ID: mdl-25386992

ABSTRACT

Pancreatitis is caused by inflammatory injury to the exocrine pancreas, from which both humans and animal models appear to recover via regeneration of digestive enzyme-producing acinar cells. This regenerative process involves transient phases of inflammation, metaplasia, and redifferentiation, driven by cell-cell interactions between acinar cells, leukocytes, and resident fibroblasts. The NFκB signaling pathway is a critical determinant of pancreatic inflammation and metaplasia, whereas a number of developmental signals and transcription factors are devoted to promoting acinar redifferentiation after injury. Imbalances between these proinflammatory and prodifferentiation pathways contribute to chronic pancreatitis, characterized by persistent inflammation, fibrosis, and acinar dedifferentiation. Loss of acinar cell differentiation also drives pancreatic cancer initiation, providing a mechanistic link between pancreatitis and cancer risk. Unraveling the molecular bases of exocrine regeneration may identify new therapeutic targets for treatment and prevention of both of these deadly diseases.


Subject(s)
Acinar Cells/cytology , Acinar Cells/physiology , Pancreas, Exocrine/physiology , Regeneration/physiology , Animals , Cell Differentiation/physiology , Disease Models, Animal , Humans , Pancreas, Exocrine/cytology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Pancreatitis/pathology , Pancreatitis/physiopathology , Signal Transduction/physiology
14.
Dev Biol ; 391(1): 89-98, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24721715

ABSTRACT

Pancreatic exocrine and endocrine lineages arise from multipotent pancreatic progenitor cells (MPCs). Exploiting the mechanisms that govern expansion and differentiation of these cells could enhance efforts to generate ß-cells from stem cells. Although our prior work indicates that the canonical Wnt signaling component ß-catenin is required qualitatively for exocrine acinar but not endocrine development, precisely how this requirement plays out at the level of MPCs and their lineage-restricted progeny is unknown. In addition, the contribution of ß-catenin function to ß-cell development remains controversial. To resolve the potential roles of ß-catenin in development of MPCs and ß-cells, we generated pancreas- and pre-endocrine-specific ß-catenin knockout mice. Pancreas-specific loss of ß-catenin produced not only a dramatic reduction in acinar cell numbers, but also a significant reduction in ß-cell mass. The loss of ß-cells is due not to a defect in the differentiation of endocrine precursors, but instead correlates with an early and specific loss of MPCs. In turn, this reflects a novel role for ß-catenin in maintaining proximal-distal patterning of the early epithelium, such that distal MPCs resort to a proximal, endocrine-competent "trunk" fate when ß-catenin is deleted. Moreover, ß-catenin maintains proximal-distal patterning, in part, by inhibiting Notch signaling. Subsequently, ß-catenin is required for proliferation of both distal and proximal cells, driving overall organ growth. In distinguishing two distinct roles for ß-catenin along the route of ß-cell development, we suggest that temporally appropriate positive and negative manipulation of this molecule could enhance expansion and differentiation of stem cell-derived MPCs.


Subject(s)
Epithelial Cells/cytology , Gene Expression Regulation, Developmental , Pancreas/embryology , Pancreas/metabolism , beta Catenin/genetics , beta Catenin/physiology , Animals , Body Patterning , Cell Differentiation , Cell Proliferation , Epithelium/metabolism , Genotype , Insulin-Secreting Cells/cytology , Islets of Langerhans/cytology , Mice , Mice, Knockout , Organ Size , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology
15.
Stem Cell Reports ; 2(2): 127-34, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24527386

ABSTRACT

Wnt signaling is a crucial aspect of the intestinal stem cell niche required for crypt cell proliferation and differentiation. Paneth cells or subepithelial myofibroblasts are leading candidate sources of the required Wnt ligands, but this has not been tested in vivo. To abolish Wnt-ligand secretion, we used Porcupine (Porcn) conditional-null mice crossed to strains expressing inducible Cre recombinase in the epithelium, including Paneth cells (Villin-Cre (ERT2) ); in smooth muscle, including subepithelial myofibroblasts (Myh11-Cre (ERT2) ); and simultaneously in both compartments. Elimination of Wnt secretion from any of these compartments did not disrupt tissue morphology, cell proliferation, differentiation, or Wnt pathway activity. Thus, Wnt-ligand secretion from these cell populations is dispensable for intestinal homeostasis, revealing that a minor cell type or significant and unexpected redundancy is responsible for physiologic Wnt signaling in vivo.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestines/cytology , Myofibroblasts/metabolism , Stem Cell Niche , Wnt Proteins/metabolism , Acyltransferases , Animals , Cell Proliferation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Deletion , Gene Expression , Gene Targeting , Immunohistochemistry , Intestinal Mucosa/pathology , Intestines/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Wnt Signaling Pathway
17.
Toxicol Pathol ; 42(1): 217-28, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24178582

ABSTRACT

The past several decades have seen great effort devoted to mimicking the key features of pancreatic ductal adenocarcinoma (PDAC) in animals and have produced 2 robust models of this deadly cancer. Carcinogen-treated Syrian hamsters develop PDAC with genetic lesions, which reproduce those of human, including activation of the Kras oncogene, and early studies in this species validated nongenetic risk factors for PDAC including pancreatitis, obesity, and diabetes. More recently, PDAC research has been invigorated by the development of genetically engineered mouse models based on tissue-specific Kras activation and deletion of tumor suppressor genes. Surprisingly, mouse PDAC appears to arise from exocrine acinar rather than ductal cells, via a process of phenotypic reprogramming that is accelerated by inflammation. Studies in both models have uncovered molecular mechanisms by which inflammation promotes and sustains PDAC and identified targets for chemoprevention to suppress PDAC in high-risk individuals. The mouse model, in particular, has also been instrumental in developing new approaches to early detection as well as treatment of advanced disease. Together, animal models enable diverse approaches to basic and preclinical research on pancreatic cancer, the results of which will accelerate progress against this currently intractable cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics , Cricetinae , Humans , Mice , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Risk Factors
18.
Development ; 140(24): 4870-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24198274

ABSTRACT

The formation of epithelial tubes underlies the development of diverse organs. In the skin, hair follicles resemble tube-like structures with lumens that are generated through poorly understood cellular rearrangements. Here, we show that creation of the hair follicle lumen is mediated by early outward movement of keratinocytes from within the cores of developing hair buds. These migratory keratinocytes express keratin 79 (K79) and stream out of the hair germ and into the epidermis prior to lumen formation in the embryo. Remarkably, this process is recapitulated during hair regeneration in the adult mouse, when K79(+) cells migrate out of the reactivated secondary hair germ prior to formation of a new hair canal. During homeostasis, K79(+) cells line the hair follicle infundibulum, a domain we show to be multilayered, biochemically distinct and maintained by Lrig1(+) stem cell-derived progeny. Upward movement of these cells sustains the infundibulum, while perturbation of this domain during acne progression is often accompanied by loss of K79. Our findings uncover previously unappreciated long-distance cell movements throughout the life cycle of the hair follicle, and suggest a novel mechanism by which the follicle generates its hollow core through outward cell migration.


Subject(s)
Acne Vulgaris/metabolism , Hair Follicle/embryology , Keratinocytes/metabolism , Keratins/metabolism , Regeneration , Animals , Cell Line , Cell Movement , HEK293 Cells , Hair/embryology , Hair Follicle/metabolism , Humans , Keratins/genetics , Keratins, Hair-Specific , Keratins, Type II , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Morphogenesis , Nerve Tissue Proteins/metabolism
19.
J Neurosci ; 33(30): 12197-207, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884928

ABSTRACT

The LIM-Homeodomain transcription factor Lhx2 is an essential organizer of early eye development and is subsequently expressed in retinal progenitor cells (RPCs). To determine its requirement in RPCs, we performed a temporal series of conditional inactivations in mice with the early RPC driver Pax6 α-Cre and the tamoxifen-inducible Hes1(CreERT2) driver. Deletion of Lhx2 caused a significant reduction of the progenitor population and a corresponding increase in neurogenesis. Precursor fate choice correlated with the time of inactivation; early and late inactivation led to the overproduction of retinal ganglion cells (RGCs) and rod photoreceptors, respectively. In each case, however, the overproduction was selective, occurring at the expense of other cell types and indicating a role for Lhx2 in generating cell type diversity. RPCs that persisted in the absence of Lhx2 continued to generate RGC precursors beyond their normal production window, suggesting that Lhx2 facilitates a transition in competence state. These results identify Lhx2 as a key regulator of RPC properties that contribute to the ordered production of multiple cell types during retinal tissue formation.


Subject(s)
Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Retina/embryology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Female , Gene Knock-In Techniques , Male , Mice , Mice, Mutant Strains , Neural Stem Cells/cytology , Pregnancy , Retina/cytology , Retina/growth & development , Retina/physiology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/physiology
20.
Dev Cell ; 23(3): 624-36, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975330

ABSTRACT

Previous studies have raised the possibility that Wnt signaling may regulate both neural progenitor maintenance and neuronal differentiation within a single population. Here we investigate the role of Wnt/ß-catenin activity in the zebrafish hypothalamus and find that the pathway is first required for the proliferation of unspecified hypothalamic progenitors in the embryo. At later stages, including adulthood, sequential activation and inhibition of Wnt activity is required for the differentiation of neural progenitors and negatively regulates radial glia differentiation. The presence of Wnt activity is conserved in hypothalamic progenitors of the adult mouse, where it plays a conserved role in inhibiting the differentiation of radial glia. This study establishes the vertebrate hypothalamus as a model for Wnt-regulated postembryonic neural progenitor differentiation and defines specific roles for Wnt signaling in neurogenesis.


Subject(s)
Hypothalamus/cytology , Neurogenesis , Stem Cells/cytology , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish/growth & development , Animals , Hypothalamus/metabolism , Mice , Neuroglia/cytology , Neuroglia/metabolism , Stem Cells/metabolism , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...