Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38645025

ABSTRACT

The plasticity and diversity of cell types with specialized functions likely defines the capacity of multicellular organisms to adapt to physiologic stressors. The kidney collecting ducts contribute to water, electrolyte, and pH homeostasis and are composed of mature intermingled epithelial cell types that are susceptible to transdifferentiate. The conversion of kidney collecting duct principal cells to intercalated cells is actively inhibited by Notch signaling to ensure urine concentrating capability. Here we identify Hes1, a target of Notch signaling, allows for maintenance of functionally distinct epithelial cell types within the same microenvironment by regulating mechanistic target of rapamycin complex 1 (mTORC1) activity. Hes1 directly represses the expression of insulin receptor substrate 1 ( Irs1 ), an upstream component of mTOR pathway and suppresses mTORC1 activity in principal cells. Genetic inactivation of tuberous sclerosis complex 2 ( Tsc2 ) to increase mTORC1 activity in mature principal cells is sufficient to promote acquisition of intercalated cell properties, while inhibition of mTORC1 in adult kidney epithelia suppresses intercalated cell properties. Considering that mTORC1 integrates environmental cues, the linkage of functionally distinct epithelial cell types to mTORC1 activity levels likely allows for cell plasticity to be regulated by physiologic and metabolic signals and the ability to sense/transduce these signals.

2.
Cell Death Differ ; 29(8): 1596-1610, 2022 08.
Article in English | MEDLINE | ID: mdl-35322202

ABSTRACT

Multiciliated cells (MCCs) in the brain reside in the ependyma and the choroid plexus (CP) epithelia. The CP secretes cerebrospinal fluid that circulates within the ventricular system, driven by ependymal cilia movement. Tumors of the CP are rare primary brain neoplasms mostly found in children. CP tumors exist in three forms: CP papilloma (CPP), atypical CPP, and CP carcinoma (CPC). Though CPP and atypical CPP are generally benign and can be resolved by surgery, CPC is a particularly aggressive and little understood cancer with a poor survival rate and a tendency for recurrence and metastasis. In contrast to MCCs in the CP epithelia, CPCs in humans are characterized by solitary cilia, frequent TP53 mutations, and disturbances to multiciliogenesis program directed by the GMNC-MCIDAS transcriptional network. GMNC and MCIDAS are early transcriptional regulators of MCC fate differentiation in diverse tissues. Consistently, components of the GMNC-MCIDAS transcriptional program are expressed during CP development and required for multiciliation in the CP, while CPC driven by deletion of Trp53 and Rb1 in mice exhibits multiciliation defects consequent to deficiencies in the GMNC-MCIDAS program. Previous studies revealed that abnormal NOTCH pathway activation leads to CPP. Here we show that combined defects in NOTCH and Sonic Hedgehog signaling in mice generates tumors that are similar to CPC in humans. NOTCH-driven CP tumors are monociliated, and disruption of the NOTCH complex restores multiciliation and decreases tumor growth. NOTCH suppresses multiciliation in tumor cells by inhibiting the expression of GMNC and MCIDAS, while Gmnc-Mcidas overexpression rescues multiciliation defects and suppresses tumor cell proliferation. Taken together, these findings indicate that reactivation of the GMNC-MCIDAS multiciliogenesis program is critical for inhibiting tumorigenesis in the CP, and it may have therapeutic implications for the treatment of CPC.


Subject(s)
Carcinoma , Cell Cycle Proteins , Choroid Plexus Neoplasms , Nuclear Proteins , Animals , Carcinoma/genetics , Cell Cycle Proteins/genetics , Choroid Plexus Neoplasms/genetics , Choroid Plexus Neoplasms/pathology , Hedgehog Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics
3.
JCI Insight ; 5(21)2020 11 05.
Article in English | MEDLINE | ID: mdl-33001861

ABSTRACT

Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type-specific expression and unique subcellular localization of the NM2 proteins, encoded by the Myh9 and Myh10 genes, in the mouse kidney tubules led us to hypothesize that these proteins have specialized functional roles within the renal epithelium. Inducible conditional knockout (cKO) of Myh9 and Myh10 in the renal tubules of adult mice resulted in progressive kidney disease. Prior to overt renal tubular injury, we observed intracellular accumulation of the glycosylphosphatidylinositol-anchored protein uromodulin (UMOD) and gradual loss of Na+ K+ 2Cl- cotransporter from the apical membrane of the thick ascending limb epithelia. The UMOD accumulation coincided with expansion of endoplasmic reticulum (ER) tubules and activation of ER stress and unfolded protein response pathways in Myh9&10-cKO kidneys. We conclude that NM2 proteins are required for localization and transport of UMOD and loss of function results in accumulation of UMOD and ER stress-mediated progressive renal tubulointerstitial disease. These observations establish cell type-specific role(s) for NM2 proteins in regulation of specialized renal epithelial transport pathways and reveal the possibility that human kidney disease associated with MYH9 mutations could be of renal epithelial origin.


Subject(s)
Endoplasmic Reticulum Stress , Epithelium/pathology , Kidney Diseases/pathology , Kidney Tubules/pathology , Myosin Heavy Chains/physiology , Nonmuscle Myosin Type IIB/physiology , Animals , Epithelium/metabolism , Female , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Type II/genetics , Myosin Type II/metabolism , Podocytes/metabolism , Podocytes/pathology , Solute Carrier Family 12, Member 1/genetics , Solute Carrier Family 12, Member 1/metabolism , Unfolded Protein Response , Uromodulin/genetics , Uromodulin/metabolism
4.
Dev Biol ; 466(1-2): 1-11, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32800756

ABSTRACT

The distal nephron and collecting duct segments of the mammalian kidney consist of intercalated cell types intermingled among principal cell types. Notch signaling ensures that a sufficient number of cells select a principal instead of an intercalated cell fate. However, the precise mechanisms by which Notch signaling patterns the distal nephron and collecting duct cell fates is unknown. Here we observed that Hes1, a direct target of Notch signaling pathway, is required within the mouse developing collecting ducts for repression of Foxi1 expression, an essential intercalated cell specific transcription factor. Interestingly, inactivation of Foxi1 in Hes1-deficient collecting ducts rescues the deficiency in principal cell fate selection, overall urine concentrating deficiency, and reduces the occurrence of hydronephrosis. However, Foxi1 inactivation does not rescue the reduction in expression of all principal cell genes in the Hes1-deficient kidney collecting duct cells that select the principal cell fate. Additionally, suppression of Notch/Hes1 signaling in mature principal cells reduces principal cell gene expression without activating Foxi1. We conclude that Hes1 is a Notch signaling target that is essential for normal patterning of the collecting ducts with intermingled cell types by repressing Foxi1, and for maintenance of principal cell gene expression independent of repressing Foxi1.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Kidney/embryology , Receptors, Notch/metabolism , Signal Transduction , Transcription Factor HES-1/deficiency , Animals , Forkhead Transcription Factors/genetics , Mice , Mice, Mutant Strains , Receptors, Notch/genetics , Transcription Factor HES-1/metabolism
5.
FASEB J ; 34(7): 9512-9530, 2020 07.
Article in English | MEDLINE | ID: mdl-32474964

ABSTRACT

Alagille syndrome patients present with loss of function mutations in either JAG1 or NOTCH2. About 40%-50% of patients have kidney abnormalities, and frequently display multicystic, dysplastic kidneys. Additionally, gain-of-function mutations in NOTCH2 are associated with cystic kidneys in Hajdu-Cheney syndrome patients. How perturbations in Notch signaling cause renal tubular cysts remains unclear. Here, we have determined that reduced Notch signaling mediated transcription by ectopic expression of dominant-negative mastermind-like (dnMaml) peptide in the nephrogenic epithelia from after the s-shaped body formation and in the developing collecting ducts results in proximal tubular and collecting duct cysts, respectively. An acute inhibition of Notch signaling for two days during kidney development is sufficient to disrupt tubule formation, and significantly increases Akap12 expression. Ectopic expression of Akap12 in renal epithelia results in abnormally long primary cilia similar to that observed in Notch-signaling-deficient epithelia. Both loss of Notch signaling and elevated Akap12 expression disrupt the ability of renal epithelial cells to form spherical structures with a single lumen when grown embedded in matrix. Interestingly, Akap12 can inhibit Notch signaling mediated transcription, which likely explains how both loss of Notch signaling and ectopic expression of Akap12 result in similar renal epithelial abnormalities. We conclude that Notch signaling regulates Akap12 expression while also ensuring normal primary cilia length and renal epithelial morphogenesis, and suggest that one aspect of diseases associated with defective Notch signaling, such as Alagille syndrome, maybe mechanistically related to ciliopathies.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cilia/physiology , Gene Expression Regulation , Kidney Tubules/cytology , Morphogenesis , Nuclear Proteins/physiology , Receptor, Notch2/metabolism , Transcription Factors/physiology , A Kinase Anchor Proteins/genetics , Animals , Cell Cycle Proteins/genetics , Female , Genes, Dominant , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Notch2/genetics
6.
Biomolecules ; 9(11)2019 11 04.
Article in English | MEDLINE | ID: mdl-31690016

ABSTRACT

Kidney development involves formation of nephrons intricately aligned with the vasculature and connected to a branched network of collecting ducts. Notch signaling plays multiple roles during kidney development involving the formation of nephrons composed of diverse epithelial cell types arranged into tubular segments, all the while maintaining a nephron progenitor niche. Here, we review the roles of Notch signaling identified from rodent kidney development and injury studies, while discussing human kidney diseases associated with aberrant Notch signaling. We also review Notch signaling requirement in maintenance of mature kidney epithelial cell states and speculate that Notch activity regulation mediates certain renal physiologic adaptations.


Subject(s)
Kidney Diseases/metabolism , Kidney/growth & development , Receptors, Notch/metabolism , Animals , Gene Expression Regulation, Developmental , Humans , Kidney/metabolism , Kidney Diseases/genetics , Organogenesis , Receptors, Notch/genetics , Signal Transduction
7.
FEBS Lett ; 593(10): 1030-1039, 2019 05.
Article in English | MEDLINE | ID: mdl-31002388

ABSTRACT

Elf5 is a transcription factor known to regulate critical developmental processes and has been shown to act as a tumour suppressor in multiple cancers. Elf5 knockout mice are embryonically lethal, limiting in vivo studies pertaining to its function. Moreover, haploinsufficiency of Elf5 limits the use of current mouse models to investigate adult tissue distribution of Elf5. Here, we successfully generated Elf5CreERT2-GFP bacterial artificial chromosome (BAC) transgenic mice and show that Elf5+ cells are present in several adult tissues, where its expression was previously not known. Our study demonstrates the unique distribution of Elf5+ cells in multiple adult organs, which will facilitate future studies investigating the function of Elf5 in these tissues during homeostasis, repair and cancer.


Subject(s)
Cell Lineage , DNA-Binding Proteins/genetics , Mice, Transgenic , Models, Animal , Transcription Factors/genetics , Animals , Chromosomes, Artificial, Bacterial , Integrases , Mice , Mice, Knockout
8.
J Am Soc Nephrol ; 30(1): 110-126, 2019 01.
Article in English | MEDLINE | ID: mdl-30514723

ABSTRACT

BACKGROUND: Notch signaling is required during kidney development for nephron formation and principal cell fate selection within the collecting ducts. Whether Notch signaling is required in the adult kidney to maintain epithelial diversity, or whether its loss can trigger principal cell transdifferentiation (which could explain acquired diabetes insipidus in patients receiving lithium) is unclear. METHODS: To investigate whether loss of Notch signaling can trigger principal cells to lose their identity, we genetically inactivated Notch1 and Notch2, inactivated the Notch signaling target Hes1, or induced expression of a Notch signaling inhibitor in all of the nephron segments and collecting ducts in mice after kidney development. We examined renal function and cell type composition of control littermates and mice with conditional Notch signaling inactivation in adult renal epithelia. In addition, we traced the fate of genetically labeled adult kidney collecting duct principal cells after Hes1 inactivation or lithium treatment. RESULTS: Notch signaling was required for maintenance of Aqp2-expressing cells in distal nephron and collecting duct segments in adult kidneys. Fate tracing revealed mature principal cells in the inner stripe of the outer medulla converted to intercalated cells after genetic inactivation of Hes1 and, to a lesser extent, lithium treatment. Hes1 ensured repression of Foxi1 to prevent the intercalated cell program from turning on in mature Aqp2+ cell types. CONCLUSIONS: Notch signaling viaHes1 regulates maintenance of mature renal epithelial cell states. Loss of Notch signaling or use of lithium can trigger transdifferentiation of mature principal cells to intercalated cells in adult kidneys.


Subject(s)
Aquaporin 2/metabolism , Lithium/pharmacology , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Water-Electrolyte Balance/genetics , Animals , Cell Differentiation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Homeostasis/genetics , Kidney/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Mice , Receptor, Notch1/drug effects , Receptor, Notch2/drug effects , Signal Transduction/genetics , Water-Electrolyte Balance/physiology
9.
Physiol Rep ; 5(23)2017 Dec.
Article in English | MEDLINE | ID: mdl-29208685

ABSTRACT

The diverse epithelial cell types of the kidneys are segregated into nephron segments and the collecting ducts in order to endow each tubular segment with unique functions. The rich diversity of the epithelial cell types is highlighted by the unique membrane channels and receptors expressed within each nephron segment. Our previous work identified a critical role for Myh9 and Myh10 in mammalian endocytosis. Here, we examined the expression patterns of Nonmuscle myosin 2 (NM2) heavy chains encoded by Myh9, Myh10, and Myh14 in mouse kidneys as these genes may confer unique nephron segment-specific membrane transport properties. Interestingly, we found that each segment of the renal tubules predominately expressed only two of the three NM2 isoforms, with isoform-specific subcellular localization, and different levels of expression within a nephron segment. Additionally, we identify Myh14 to be restricted to the intercalated cells and Myh10 to be restricted to the principal cells within the collecting ducts and connecting segments. We speculate that the distinct expression pattern of the NM2 proteins likely reflects the diversity of the intracellular trafficking machinery present within the different renal tubular epithelial segments.


Subject(s)
Kidney Tubules, Proximal/metabolism , Nonmuscle Myosin Type IIA/metabolism , Animals , Mice , Mice, Inbred C57BL , Nonmuscle Myosin Type IIA/genetics , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/metabolism
10.
Cancer Res ; 77(14): 3766-3777, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28490517

ABSTRACT

Medulloblastoma arising from the cerebellum is the most common pediatric brain malignancy, with leptomeningeal metastases often present at diagnosis and recurrence associated with poor clinical outcome. In this study, we used mouse medulloblastoma models to explore the relationship of tumor pathophysiology and dysregulated expression of the NOTCH pathway transcription factor ATOH1, which is present in aggressive medulloblastoma subtypes driven by aberrant Sonic Hedgehog/Patched (SHH/PTCH) signaling. In experiments with conditional ATOH1 mouse mutants crossed to Ptch1+/- mice, which develop SHH-driven medulloblastoma, animals with Atoh1 transgene expression developed highly penetrant medulloblastoma at a young age with extensive leptomeningeal disease and metastasis to the spinal cord and brain, resembling xenografts of human SHH medulloblastoma. Metastatic tumors retained abnormal SHH signaling like tumor xenografts. Conversely, ATOH1 expression was detected consistently in recurrent and metastatic SHH medulloblastoma. Chromatin immunoprecipitation sequencing and gene expression profiling identified candidate ATOH1 targets in tumor cells involved in development and tumorigenesis. Among these targets specific to metastatic tumors, there was an enrichment in those implicated in extracellular matrix remodeling activity, cytoskeletal network and interaction with microenvironment, indicating a shift in transcriptomic and epigenomic landscapes during metastasis. Treatment with bone morphogenetic protein or SHH pathway inhibitors decreased tumor cell proliferation and suppressed metastatic tumor growth, respectively. Our work reveals a dynamic ATOH1-driven molecular cascade underlying medulloblastoma metastasis that offers possible therapeutic opportunities. Cancer Res; 77(14); 3766-77. ©2017 AACR.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Hedgehog Proteins , Heterografts , Humans , Medulloblastoma/genetics , Mice , Mice, Transgenic , Neoplasm Metastasis , Signal Transduction
11.
Dev Biol ; 424(1): 77-89, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28215940

ABSTRACT

The mammalian kidney collecting ducts are critical for water, electrolyte and acid-base homeostasis and develop as a branched network of tubular structures composed of principal cells intermingled with intercalated cells. The intermingled nature of the different collecting duct cell types has made it challenging to identify unique and critical factors that mark and/or regulate the development of the different collecting duct cell lineages. Here we report that the canonical Notch signaling pathway components, RBPJ and Presinilin1 and 2, are involved in patterning the mouse collecting duct cell fates by maintaining a balance between principal cell and intercalated cell fates. The relatively reduced number of principal cells in Notch-signaling-deficient kidneys offered a unique genetic leverage to identify critical principal cell-enriched factors by transcriptional profiling. Elf5, which codes for an ETS transcription factor, is one such gene that is down-regulated in kidneys with Notch-signaling-deficient collecting ducts. Additionally, Elf5 is among the earliest genes up regulated by ectopic expression of activated Notch1 in the developing collecting ducts. In the kidney, Elf5 is first expressed early within developing collecting ducts and remains on in mature principal cells. Lineage tracing of Elf5-expressing cells revealed that they are committed to the principal cell lineage by as early as E16.5. Over-expression of ETS Class IIa transcription factors, including Elf5, Elf3 and Ehf, increase the transcriptional activity of the proximal promoters of Aqp2 and Avpr2 in cultured ureteric duct cell lines. Conditional inactivation of Elf5 in the developing collecting ducts results in a small but significant reduction in the expression levels of Aqp2 and Avpr2 genes. We have identified Elf5 as an early maker of the principal cell lineage that contributes to the expression of principal cell specific genes.


Subject(s)
Aquaporin 2/genetics , Cell Lineage , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Kidney/cytology , Kidney/metabolism , Receptors, Vasopressin/genetics , Transcription Factors/metabolism , Animals , Aquaporin 2/metabolism , Cell Count , Cell Line , Down-Regulation/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Integrases/metabolism , Kidney/embryology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/embryology , Kidney Tubules, Collecting/metabolism , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptors, Notch/metabolism , Receptors, Vasopressin/metabolism , Signal Transduction , Up-Regulation/genetics , Ureter/embryology , Ureter/metabolism
12.
J Am Soc Nephrol ; 26(1): 149-59, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24904084

ABSTRACT

A disintegrin and metalloproteinase domain 10 (Adam10), a member of the ADAM family of cell membrane-anchored proteins, has been linked to the regulation of the Notch, EGF, E-cadherin, and other signaling pathways. However, it is unclear what role Adam10 has in the kidney in vivo. In this study, we showed that Adam10 deficiency in ureteric bud (UB) derivatives leads to a decrease in urinary concentrating ability, polyuria, and hydronephrosis in mice. Furthermore, Adam10 deficiency led to a reduction in the percentage of aquaporin 2 (Aqp2)(+) principal cells (PCs) in the collecting ducts that was accompanied by a proportional increase in the percentage of intercalated cells (ICs). This increase was more prominent in type A ICs than in type B ICs. Foxi1, a transcription factor important for the differentiation of ICs, was upregulated in the Adam10 mutants. The observed reduction of Notch activity in Adam10 mutant collecting duct epithelium and the similar reduction of PC/IC ratios in the collecting ducts in mice deficient for mindbomb E3 ubiquitin protein ligase 1, a key regulator of the Notch and Wnt/receptor-like tyrosine kinase signaling pathways, suggest that Adam10 regulates cell fate determination through the activation of Notch signaling, probably through the regulation of Foxi1 expression. However, phenotypic differences between the Adam10 mutants, the Mib1 mutants, and the Foxi1 mutants suggest that the functions of Adam10 in determining the fate of collecting duct cells are more complex than those of a simple upstream factor in a linear pathway involving Notch and Foxi1.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Kidney/metabolism , Membrane Proteins/metabolism , ADAM10 Protein , Animals , Apoptosis , Aquaporin 2/metabolism , Cadherins/metabolism , Cell Proliferation , Epithelial Cells/cytology , Forkhead Transcription Factors/metabolism , Hydronephrosis/genetics , Kidney Tubules/cytology , Kidney Tubules, Collecting/metabolism , Ligands , Mice , Mice, Transgenic , Mutation , Polyuria/genetics , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Up-Regulation , Wnt Signaling Pathway
13.
Pediatr Nephrol ; 29(12): 2253-61, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24217784

ABSTRACT

The lysosome, an organelle central to macromolecule degradation and recycling, plays a pivotal role in normal cell processes, ranging from autophagy to redox regulation. Not surprisingly, lysosomes are an integral part of the renal epithelial molecular machinery that facilitates normal renal physiology. Two inherited diseases that manifest as kidney dysfunction are Fabry's disease and cystinosis, each of which is caused by a primary biochemical defect at the lysosome resulting from loss-of-function mutations in genes that encode lysosomal proteins. The functions of the lysosomes in the kidney and how lysosomal dysfunction might contribute to Fabry's disease and cystinosis are discussed. Unlike most other pediatric renal diseases, therapies are available for Fabry's disease and cystinosis, but require early diagnosis. Recent analysis of ceroid neuronal lipofuscinosis type 3 (Cln3) null mice, a mouse model of lysosomal disease that is primarily associated with neurological deficits, revealed renal functional abnormalities. As current and future therapeutics increase the life-span of those suffering from diseases like neuronal ceroid lipofuscinosis, it remains a distinct possibility that many more lysosomal disorders that primarily manifest as infant and juvenile neurodegenerative diseases may also include renal disease phenotypes.


Subject(s)
Kidney Diseases , Lysosomal Storage Diseases , Lysosomes , Animals , Humans
14.
PLoS Genet ; 8(3): e1002577, 2012.
Article in English | MEDLINE | ID: mdl-22457635

ABSTRACT

Notch signaling between neighboring cells controls many cell fate decisions in metazoans both during embryogenesis and in postnatal life. Previously, we uncovered a critical role for physiological Notch signaling in suppressing osteoblast differentiation in vivo. However, the contribution of individual Notch receptors and the downstream signaling mechanism have not been elucidated. Here we report that removal of Notch2, but not Notch1, from the embryonic limb mesenchyme markedly increased trabecular bone mass in adolescent mice. Deletion of the transcription factor RBPjk, a mediator of all canonical Notch signaling, in the mesenchymal progenitors but not the more mature osteoblast-lineage cells, caused a dramatic high-bone-mass phenotype characterized by increased osteoblast numbers, diminished bone marrow mesenchymal progenitor pool, and rapid age-dependent bone loss. Moreover, mice deficient in Hey1 and HeyL, two target genes of Notch-RBPjk signaling, exhibited high bone mass. Interestingly, Hey1 bound to and suppressed the NFATc1 promoter, and RBPjk deletion increased NFATc1 expression in bone. Finally, pharmacological inhibition of NFAT alleviated the high-bone-mass phenotype caused by RBPjk deletion. Thus, Notch-RBPjk signaling functions in part through Hey1-mediated inhibition of NFATc1 to suppress osteoblastogenesis, contributing to bone homeostasis in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Bone and Bones/metabolism , Cell Differentiation , Immunoglobulin J Recombination Signal Sequence-Binding Protein , NFATC Transcription Factors , Osteoblasts , Receptor, Notch2 , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone and Bones/embryology , Embryonic Development , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mesoderm/embryology , Mice , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoblasts/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Repressor Proteins/metabolism , Signal Transduction , Stem Cells/metabolism
15.
Adv Exp Med Biol ; 727: 99-113, 2012.
Article in English | MEDLINE | ID: mdl-22399342

ABSTRACT

The kidney is the body's filter, responsible for the removal of metabolic waste and the excretion or reabsorption of electrolytes to control blood composition and pH balance. The functional unit of this filter is the nephron, whose segmented architecture has been largely conserved in form and function throughout eukaryotic evolution. Not surprisingly, the core developmental pathways that regulate the formation of the nephron have also been conserved. In particular, the Notch signaling pathway functions in both primitive and advanced nephrons to pattern domains required for the kidney's diverse functions. In this chapter, we will discuss the role that Notch plays in directing cell fate decisions during embryonic development of the pronephros and metanephros. We will go on to discuss the later role of Notch signaling as a cyst-suppressor and the consequences of aberrant or absent Notch activity in disease and cancer. The work discussed here highlights the fundamental importance of Notch during development and homeostasis of the kidney and underlies the need for mechanistic understanding of its role towards the treatment of human disease.


Subject(s)
Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney/cytology , Kidney/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Humans
16.
J Am Soc Nephrol ; 22(7): 1208-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21700830

ABSTRACT

Successful regenerative renal medicine depends on understanding the molecular mechanisms by which diverse phenotypes of epithelial cells differentiate from metanephric mesenchyme to populate nephrons. Whereas many genes are maintained in a poised state within the population of pluripotent progenitors, specialized epithelial functions reflect the selective expression of a subset of genes and the repression of all others. Here we highlight some common mechanisms of cell differentiation and epigenetic regulation to discuss their implications for renal epithelial development, repair, and disease.


Subject(s)
Cell Differentiation , Chromatin/metabolism , Epigenesis, Genetic , Epithelial Cells/cytology , Kidney/cytology , Animals , Kidney/metabolism , Kidney Diseases/etiology , Regeneration , Transcription Factors/metabolism
17.
J Am Soc Nephrol ; 21(5): 819-32, 2010 May.
Article in English | MEDLINE | ID: mdl-20378824

ABSTRACT

The formation of proximal nephron segments requires canonical Notch2 signaling, but other functions of Notch signaling during renal development are incompletely understood. Here, we report that proximal tubules forming with reduced Notch signaling, resulting from delayed conditional inactivation of Notch1 and/or Notch2, are prone to cyst formation and tubular epithelial stratification. Conditional inactivation of the DNA binding factor RBP-J, which mediates Notch signaling, also resulted in multiple congenital cysts arising from the proximal tubule. Moreover, a few stratified foci/microadenomas containing hyperproliferative cells, resembling precursors of papillary renal cell carcinoma, formed in these proximal tubules. Epithelial stratification correlated neither with reduced expression of the transcriptional regulator of ciliary proteins TCF2/HNF1beta nor with loss of apical-basal polarity. Instead, Notch signaling helped to restrict the orientation of epithelial mitotic spindles to a plane parallel to the basement membrane during nephron elongation. In the absence of Notch, random spindle orientation may explain the epithelial stratification and cyst formation. Furthermore, post hoc analysis of human class 1 papillary renal cell carcinoma revealed reduced Notch activity in these tumors, resulting from abundant expression of a potent inhibitor of canonical Notch signaling, KyoT3/FHL1B. In summary, these data suggest that canonical Notch signaling maintains the alignment of cell division in the proximal tubules during nephrogenesis and that perturbations in Notch signaling may lead to cystic renal disease and tumorigenesis.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Diseases, Cystic/metabolism , Kidney Neoplasms/metabolism , Kidney Tubules, Proximal/embryology , Receptors, Notch/metabolism , Adenoma/metabolism , Animals , Basement Membrane/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Proximal/metabolism , LIM Domain Proteins , Mice , Mice, Transgenic , Morphogenesis , Muscle Proteins/metabolism , Signal Transduction , Spindle Apparatus/metabolism , Urothelium/ultrastructure
18.
Dev Biol ; 337(2): 386-95, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19914235

ABSTRACT

We previously determined that Notch2, and not Notch1, was required for forming proximal nephron segments. The dominance of Notch2 may be conserved in humans, since Notch2 mutations occur in Alagille syndrome (ALGS) 2 patients, which includes renal complications. To test whether mutations in Notch1 could increase the severity of renal complications in ALGS, we inactivated conditional Notch1 and Notch2 alleles in mice using a Six2-GFP::Cre. This BAC transgene is expressed mosaically in renal epithelial progenitors but uniformly in cells exiting the progenitor pool to undergo mesenchymal-to-epithelial transition. Although delaying Notch2 inactivation had a marginal effect on nephron numbers, it created a sensitized background in which the inactivation of Notch1 severely compromised nephron formation, function, and survival. These and additional observations indicate that Notch1 in concert with Notch2 contributes to the morphogenesis of renal vesicles into S-shaped bodies in a RBP-J-dependent manner. A significant implication is that elevating Notch1 activity could improve renal functions in ALGS2 patients. As proof of principle, we determined that conditional inactivation of Mint, an inhibitor of Notch-RBP-J interaction, resulted in a moderate rescue of Notch2 null kidneys, implying that temporal blockage of Notch signaling inhibitors downstream of receptor activation may have therapeutic benefits for ALGS patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Dosage/genetics , Nephrons/embryology , Nephrons/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Notch1/metabolism , Animals , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/metabolism , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Integrases/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mosaicism , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Receptor, Notch2/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transgenes/genetics
19.
Endocrinology ; 149(6): 2934-42, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18339714

ABSTRACT

Intense mesangial cell apoptosis contributes to the pathogenesis of diabetic nephropathy. Although reactive oxygen radicals and Wnt signaling components are potent regulators that modulate renal tissue remodeling and morphogenesis, cross-talk between oxidative stress and Wnt/beta-catenin signaling in controlling high-glucose-impaired mesangial cell survival and renal function have not been tested. In this study, high glucose induced Ras and Rac1 activation, superoxide burst, and Wnt5a/beta-catenin destabilization and subsequently promoted caspase-3 and poly (ADP-ribose) polymerase cleavage and apoptosis in mesangial cell cultures. The pharmacological and genetic suppression of superoxide synthesis by superoxide dismutase and diphenyloniodium, dominant-negative Ras (S17N), and dominant-negative Rac1 (T17N) abrogated high-glucose-induced glycogen synthase kinase (GSK-3beta) activation and caspase-3 and poly (ADP-ribose) polymerase degradation. Inactivation of Ras and Racl also reversed Wnt/beta-catenin expression and survival of mesangial cells. Stabilization of beta-catenin by the transfection of stable beta-catenin (Delta45) and kinase-inactive GSK-3beta attenuated high-glucose-mediated mesangial cell apoptosis. Exogenous superoxide dismutase administration attenuated urinary protein secretion in diabetic rats and abrogated diabetes-mediated reactive oxygen radical synthesis in renal glomeruli. Immunohistological observation revealed that superoxide dismutase treatment abrogated diabetes-induced caspase-3 cleavage and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end-labeling (TUNEL) and increased Wnt5a/beta-catenin expression in renal glomeruli. Taken together, high glucose induced oxidative stress and apoptosis in mesangial cells. The Ras and Rac1 regulation of superoxide appeared to raise apoptotic activity by activating GSK-3beta and inhibiting Wnt5a/beta-catenin signaling. Controlling oxidative stress and Wnt/beta-catenin signaling has potential for protecting renal tissue against the deleterious effect of high glucose.


Subject(s)
Apoptosis/drug effects , Glomerular Mesangium/cytology , Glomerular Mesangium/physiology , Glucose/pharmacology , Superoxides/metabolism , beta Catenin/pharmacology , Animals , Cell Line , Glomerular Mesangium/drug effects , Humans , In Situ Nick-End Labeling , Kidney , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
20.
J Am Soc Nephrol ; 18(7): 2014-20, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17568016

ABSTRACT

The structure of a mammalian kidney is parsed into large collections of polarized nephrons, and each segment is home to a diverse community of cells that specialize in renal endocrine and excretory functions. Early developmental lengthening and diversification of nephron segments along a proximal--distal axis initiate all subsequent facets of tubular growth and function. Morphogenic cues and biochemical interactions that are critical to this process are starting to emerge. The underlying principles of regional cell signaling and transcriptional control organizing early segmentation are the subject of this review.


Subject(s)
Nephrons/embryology , Animals , Cell Differentiation , Epithelium/embryology , Mesoderm/physiology , Nephrons/cytology , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...