Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 40(3): 364-373, 2022 03.
Article in English | MEDLINE | ID: mdl-34608326

ABSTRACT

Mapping the localization of multiple proteins in their native three-dimensional (3D) context would be useful across many areas of biomedicine, but multiplexed fluorescence imaging has limited intrinsic multiplexing capability, and most methods for increasing multiplexity can only be applied to thin samples (<100 µm). Here, we harness the narrow spectrum of Raman spectroscopy and introduce Raman dye imaging and tissue clearing (RADIANT), an optical method that is capable of imaging multiple targets in thick samples in one shot. We expanded the range of suitable bioorthogonal Raman dyes and developed a tissue-clearing strategy for them (Raman 3D imaging of solvent-cleared organs (rDISCO)). We applied RADIANT to image up to 11 targets in millimeter-thick brain slices, extending the imaging depth 10- to 100-fold compared to prior multiplexed protein imaging methods. We showcased the utility of RADIANT in extracting systems information, including region-specific correlation networks and their topology in cerebellum development. RADIANT will facilitate the exploration of the intricate 3D protein interactions in complex systems.


Subject(s)
Coloring Agents , Optical Imaging , Brain/diagnostic imaging , Imaging, Three-Dimensional/methods , Optical Imaging/methods
2.
Development ; 148(6)2021 03 24.
Article in English | MEDLINE | ID: mdl-33658226

ABSTRACT

Groucho-related genes (GRGs) are transcriptional co-repressors that are crucial for many developmental processes. Several essential pancreatic transcription factors are capable of interacting with GRGs; however, the in vivo role of GRG-mediated transcriptional repression in pancreas development is still not well understood. In this study, we used complex mouse genetics and transcriptomic analyses to determine that GRG3 is essential for ß cell development, and in the absence of Grg3 there is compensatory upregulation of Grg4Grg3/4 double mutant mice have severe dysregulation of the pancreas gene program with ectopic expression of canonical liver genes and Foxa1, a master regulator of the liver program. Neurod1, an essential ß cell transcription factor and predicted target of Foxa1, becomes downregulated in Grg3/4 mutants, resulting in reduced ß cell proliferation, hyperglycemia, and early lethality. These findings uncover novel functions of GRG-mediated repression during pancreas development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Co-Repressor Proteins/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Pancreas/growth & development , Repressor Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental/genetics , Insulin-Secreting Cells/metabolism , Liver/growth & development , Liver/metabolism , Mice , Mutation/genetics , Organogenesis/genetics , Pancreas/metabolism
3.
Cell Metab ; 30(6): 1091-1106.e8, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31607563

ABSTRACT

Many studies have highlighted the role of dysregulated glucagon secretion in the etiology of hyperglycemia and diabetes. Accordingly, understanding the mechanisms underlying pancreatic islet α cell development and function has important implications for the discovery of new therapies for diabetes. In this study, comparative transcriptome analyses between embryonic mouse pancreas and adult mouse islets identified several pancreatic lncRNAs that lie in close proximity to essential pancreatic transcription factors, including the Pax6-associated lncRNA Paupar. We demonstrate that Paupar is enriched in glucagon-producing α cells where it promotes the alternative splicing of Pax6 to an isoform required for activation of essential α cell genes. Consistently, deletion of Paupar in mice resulted in dysregulation of PAX6 α cell target genes and corresponding α cell dysfunction, including blunted glucagon secretion. These findings illustrate a distinct mechanism by which a pancreatic lncRNA can coordinate glucose homeostasis by cell-specific regulation of a broadly expressed transcription factor.


Subject(s)
Diabetes Mellitus/metabolism , Glucagon-Secreting Cells/metabolism , PAX6 Transcription Factor/metabolism , RNA, Long Noncoding/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Gene Expression Profiling , Glucagon/metabolism , Glucose/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Diabetes ; 68(12): 2259-2271, 2019 12.
Article in English | MEDLINE | ID: mdl-31519700

ABSTRACT

Inactivation of the ß-cell transcription factor NEUROD1 causes diabetes in mice and humans. In this study, we uncovered novel functions of NEUROD1 during murine islet cell development and during the differentiation of human embryonic stem cells (HESCs) into insulin-producing cells. In mice, we determined that Neurod1 is required for perinatal proliferation of α- and ß-cells. Surprisingly, apoptosis only makes a minor contribution to ß-cell loss when Neurod1 is deleted. Inactivation of NEUROD1 in HESCs severely impaired their differentiation from pancreatic progenitors into insulin-expressing (HESC-ß) cells; however, survival or proliferation was not affected at the time points analyzed. NEUROD1 was also required in HESC-ß cells for the full activation of an essential ß-cell transcription factor network. These data reveal conserved and distinct functions of NEUROD1 during mouse and human ß-cell development and maturation, with important implications about the function of NEUROD1 in diabetes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Human Embryonic Stem Cells/metabolism , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cell Survival/physiology , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/cytology , Humans , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics
5.
Diabetes ; 67(8): 1461-1470, 2018 08.
Article in English | MEDLINE | ID: mdl-29937433

ABSTRACT

Diabetes is a complex group of metabolic disorders that can be accompanied by several comorbidities, including increased risk of early death. Decades of diabetes research have elucidated many genetic drivers of normal islet function and dysfunction; however, a lack of suitable treatment options suggests our knowledge about the disease remains incomplete. The establishment of long noncoding RNAs (lncRNAs), once dismissed as "junk" DNA, as essential gene regulators in many biological processes has redefined the central role for RNA in cells. Studies showing that misregulation of lncRNAs can lead to disease have contributed to the emergence of lncRNAs as attractive candidates for drug targeting. These findings underscore the need to reexamine islet biology in the context of a regulatory role for RNA. This review will 1) highlight what is known about lncRNAs in the context of diabetes, 2) summarize the strategies used in lncRNA discovery pipelines, and 3) discuss future directions and the potential impact of studying the role of lncRNAs in diabetes.


Subject(s)
Gene Expression Regulation , Islets of Langerhans/metabolism , Models, Biological , RNA, Long Noncoding/metabolism , Animals , Biomedical Research/methods , Biomedical Research/trends , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/therapy , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/pathology , Therapies, Investigational/trends
6.
Elife ; 62017 01 10.
Article in English | MEDLINE | ID: mdl-28071588

ABSTRACT

Many pancreatic transcription factors that are essential for islet cell differentiation have been well characterized; however, because they are often expressed in several different cell populations, their functional hierarchy remains unclear. To parse out the spatiotemporal regulation of islet cell differentiation, we used a Neurog3-Cre allele to ablate Nkx2.2, one of the earliest and most broadly expressed islet transcription factors, specifically in the Neurog3+ endocrine progenitor lineage (Nkx2.2△endo). Remarkably, many essential components of the ß cell transcriptional network that were down-regulated in the Nkx2.2KO mice, were maintained in the Nkx2.2△endo mice - yet the Nkx2.2△endo mice displayed defective ß cell differentiation and recapitulated the Nkx2.2KO phenotype. This suggests that Nkx2.2 is not only required in the early pancreatic progenitors, but has additional essential activities within the endocrine progenitor population. Consistently, we demonstrate Nkx2.2 functions as an integral component of a modular regulatory program to correctly specify pancreatic islet cell fates.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/physiology , Islets of Langerhans/cytology , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Animals , Homeobox Protein Nkx-2.2 , Mice , Mice, Knockout , Zebrafish Proteins
7.
Curr Opin Endocrinol Diabetes Obes ; 22(2): 77-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25692923

ABSTRACT

PURPOSE OF REVIEW: The identification and characterization of essential islet transcription factors have improved our understanding of ß cell development, provided insights into many of the cellular dysfunctions related to diabetes, and facilitated the successful generation of ß cells from alternative cell sources. Recently, noncoding RNAs have emerged as a novel set of molecules that may represent missing components of the known islet regulatory pathways. The purpose of this article is to highlight studies that have implicated noncoding RNAs as important regulators of pancreas cell development and ß cell function. RECENT FINDINGS: Disruption of essential components of the microRNA processing machinery, in addition to misregulation of individual microRNAs, has revealed the importance of microRNAs in pancreas development and ß cell function. Furthermore, over 1000 islet-specific long noncoding RNAs have been identified in mouse and human islets, suggesting that this class of noncoding molecules will also play important functional roles in the ß cell. SUMMARY: The analysis of noncoding RNAs in the pancreas will provide important new insights into pancreatic regulatory processes that will improve our ability to understand and treat diabetes, and may facilitate the generation of replacement ß cells from alternative cell sources.


Subject(s)
Diabetes Mellitus/metabolism , Insulin-Secreting Cells/metabolism , RNA, Untranslated/metabolism , Animals , Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Diabetes Mellitus/therapy , Gene Expression Regulation , Humans , Prognosis , RNA, Untranslated/genetics , Signal Transduction
8.
PLoS Genet ; 9(2): e1003206, 2013.
Article in English | MEDLINE | ID: mdl-23437001

ABSTRACT

Danforth's short tail mutant (Sd) mouse, first described in 1930, is a classic spontaneous mutant exhibiting defects of the axial skeleton, hindgut, and urogenital system. We used meiotic mapping in 1,497 segregants to localize the mutation to a 42.8-kb intergenic segment on chromosome 2. Resequencing of this region identified an 8.5-kb early retrotransposon (ETn) insertion within the highly conserved regulatory sequences upstream of Pancreas Specific Transcription Factor, 1a (Ptf1a). This mutation resulted in up to tenfold increased expression of Ptf1a as compared to wild-type embryos at E9.5 but no detectable changes in the expression levels of other neighboring genes. At E9.5, Sd mutants exhibit ectopic Ptf1a expression in embryonic progenitors of every organ that will manifest a developmental defect: the notochord, the hindgut, and the mesonephric ducts. Moreover, at E 8.5, Sd mutant mice exhibit ectopic Ptf1a expression in the lateral plate mesoderm, tail bud mesenchyme, and in the notochord, preceding the onset of visible defects such as notochord degeneration. The Sd heterozygote phenotype was not ameliorated by Ptf1a haploinsufficiency, further suggesting that the developmental defects result from ectopic expression of Ptf1a. These data identify disruption of the spatio-temporal pattern of Ptf1a expression as the unifying mechanism underlying the multiple congenital defects in Danforth's short tail mouse. This striking example of an enhancer mutation resulting in profound developmental defects suggests that disruption of conserved regulatory elements may also contribute to human malformation syndromes.


Subject(s)
Embryonic Development/genetics , Mutagenesis, Insertional/genetics , Retroelements/genetics , Transcription Factors , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Mesoderm/abnormalities , Mesoderm/growth & development , Mice , Pancreas/abnormalities , Pancreas/growth & development , Spinal Cord/abnormalities , Spinal Cord/growth & development , Tail/anatomy & histology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Genes Dev ; 25(21): 2291-305, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22056672

ABSTRACT

Regulation of cell differentiation programs requires complex interactions between transcriptional and epigenetic networks. Elucidating the principal molecular events responsible for the establishment and maintenance of cell fate identities will provide important insights into how cell lineages are specified and maintained and will improve our ability to recapitulate cell differentiation events in vitro. In this study, we demonstrate that Nkx2.2 is part of a large repression complex in pancreatic ß cells that includes DNMT3a, Grg3, and HDAC1. Mutation of the endogenous Nkx2.2 tinman (TN) domain in mice abolishes the interaction between Nkx2.2 and Grg3 and disrupts ß-cell specification. Furthermore, we demonstrate that Nkx2.2 preferentially recruits Grg3 and HDAC1 to the methylated Aristaless homeobox gene (Arx) promoter in ß cells. The Nkx2.2 TN mutation results in ectopic expression of Arx in ß cells, causing ß-to-α-cell transdifferentiation. A corresponding ß-cell-specific deletion of DNMT3a is also sufficient to cause Arx-dependent ß-to-α-cell reprogramming. Notably, subsequent removal of Arx in the ß cells of Nkx2.2(TNmut/TNmut) mutant mice reverts the ß-to-α-cell conversion, indicating that the repressor activities of Nkx2.2 on the methylated Arx promoter in ß cells are the primary regulatory events required for maintaining ß-cell identity.


Subject(s)
Glucagon-Secreting Cells/cytology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Co-Repressor Proteins , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Diabetes Mellitus/physiopathology , Gene Expression Regulation , Ghrelin/metabolism , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Insulin/metabolism , Mice , Mutation , Nuclear Proteins , Organ Specificity/genetics , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Transcription Factors/genetics , Zebrafish Proteins
10.
Development ; 138(15): 3213-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21750032

ABSTRACT

The homeodomain transcription factor Nkx2.2 is essential for pancreatic development and islet cell type differentiation. We have identified Tm4sf4, an L6 domain tetraspanin family member, as a transcriptional target of Nkx2.2 that is greatly upregulated during pancreas development in Nkx2.2(-/-) mice. Tetraspanins and L6 domain proteins recruit other membrane receptors to form active signaling centers that coordinate processes such as cell adhesion, migration and differentiation. In this study, we determined that Tm4sf4 is localized to the ductal epithelial compartment and is prominent in the Ngn3(+) islet progenitor cells. We also established that pancreatic tm4sf4 expression and regulation by Nkx2.2 is conserved during zebrafish development. Loss-of-function studies in zebrafish revealed that tm4sf4 inhibits α and ß cell specification, but is necessary for ε cell fates. Thus, Tm4sf4 functional output opposes that of Nkx2.2. Further investigation of how Tm4sf4 functions at the cellular level in vitro showed that Tm4sf4 inhibits Rho-activated cell migration and actin organization in a ROCK-independent fashion. We propose that the primary role of Nkx2.2 is to inhibit Tm4sf4 in endocrine progenitor cells, allowing for delamination, migration and/or appropriate cell fate decisions. Identification of a role for Tm4sf4 during endocrine differentiation provides insight into islet progenitor cell behaviors and potential targetable regenerative mechanisms.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Islets of Langerhans/embryology , Islets of Langerhans/growth & development , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Ghrelin/genetics , Ghrelin/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins , Sequence Alignment , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/genetics , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...