Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Pathol ; 252(3): 274-289, 2020 11.
Article in English | MEDLINE | ID: mdl-32715474

ABSTRACT

Disturbed intrauterine development increases the risk of renal disease. Various studies have reported that Notch signalling plays a significant role in kidney development and kidney diseases. A disintegrin and metalloproteinase domain 10 (ADAM10), an upstream protease of the Notch pathway, is also reportedly involved in renal fibrosis. However, how ADAM10 interacts with the Notch pathway and causes renal fibrosis is not fully understood. In this study, using a prenatal chlorpyrifos (CPF) exposure mouse model, we investigated the role of the ADAM10/Notch axis in kidney development and fibrosis. We found that prenatal CPF-exposure mice presented overexpression of Adam10, Notch1 and Notch2, and led to premature depletion of Six2+ nephron progenitors and ectopic formation of proximal tubules (PTs) in the embryonic kidney. These abnormal phenotypic changes persisted in mature kidneys due to the continuous activation of ADAM10/Notch and showed aggravated renal fibrosis in adults. Finally, both ADAM10 and NOTCH2 expression were positively correlated with the degree of renal interstitial fibrosis in IgA nephropathy patients, and increased ADAM10 expression was negatively correlated with decreased kidney function evaluated by serum creatinine, cystatin C, and estimated glomerular filtration rate. Regression analysis also indicated that ADAM10 expression was an independent risk factor for fibrosis in IgAN. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Kidney Diseases/embryology , Kidney Diseases/pathology , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/pathology , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Fibrosis , Humans , Immunohistochemistry , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
2.
Pediatr Dev Pathol ; 23(4): 306-311, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31771449

ABSTRACT

Activating heterozygous germline mutations in the signal transducer and activator of transcription 3 (STAT3) gene are associated with the rare autoimmune disorder autoimmune disease, multisystem, infantile onset (ADMIO). The phenotype of ADMIO is typified by hypogammaglobulinemia and onset of autoimmune phenomena during early childhood that include diabetes and autoimmune enteritis. This case report describes in utero onset of precocious lymphocyte maturation, autoimmune enteropathy-like inflammation, and proximal renal tubular dysplasia associated with a novel de novo heterozygous STAT3 mutation. The findings expand the phenotype associated with activating STAT3 mutations and suggest that the impact of the immunological abnormalities associated with ADMIO can begin prior to birth.


Subject(s)
Autoimmune Diseases/pathology , Kidney Tubules, Proximal/abnormalities , Prenatal Diagnosis , STAT3 Transcription Factor/genetics , Adult , Autoimmune Diseases/diagnosis , Autoimmune Diseases/embryology , Autoimmune Diseases/genetics , Female , Fetal Death , Genetic Markers , Heterozygote , Humans , Kidney Tubules, Proximal/embryology , Mutation , Phenotype , Pregnancy
3.
Croat Med J ; 60(6): 521-531, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31894918

ABSTRACT

AIM: To explore the spatial and temporal expression patterns of DAB1 and Reelin in the developing and postnatal healthy human kidneys as potential determinants of kidney development. METHODS: Paraffin-embedded fetal kidney tissue between the 13/14th and 38th developmental weeks (dw) and postnatal tissue at 1.5 and 7 years were stained with DAB1 and Reelin antibodies by double immunofluorescence. RESULTS: During the fetal kidney development and postnatal period, DAB1 and Reelin showed specific spatial expression pattern and diverse fluorescence intensity. During the fetal period, DAB1 was strongly expressed in the distal convoluted tubules (DCT), with strong reactivity, and diversely in the proximal convoluted tubules (PCT) and glomeruli. In the postnatal period, DAB1 expression decreased. The strongest Reelin expression in early fetal stages was observed in the PCT. In the postnatal period, Reelin expression decreased dramatically in all observed structures. These two markers were colocalized during early developmental stages, mostly in PCT, DCT, and podocytes. CONCLUSION: The appearance of DAB1 and Reelin during fetal kidney development confirms their potential significant role in the formation of kidney structure or function. High DAB1 expression in the DCT implies its regulatory role in tubular formation or function maintenance during development. Reelin was highly expressed in human kidneys at early fetal stages, mostly in the PCT, while at later fetal stages and postnatal period its expression decreased.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Kidney/embryology , Kidney/growth & development , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Child , Fetal Development , Gestational Age , Humans , Infant , Kidney/metabolism , Kidney Tubules, Distal/embryology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/metabolism , Podocytes/metabolism , Reelin Protein
4.
Elife ; 72018 08 10.
Article in English | MEDLINE | ID: mdl-30095068

ABSTRACT

Wingless/Wnts are signalling molecules, traditionally considered to pattern tissues as long-range morphogens. However, more recently the spread of Wingless was shown to be dispensable in diverse developmental contexts in Drosophila and vertebrates. Here we demonstrate that release and spread of Wingless is required to pattern the proximo-distal (P-D) axis of Drosophila Malpighian tubules. Wingless signalling, emanating from the midgut, directly activates odd skipped expression several cells distant in the proximal tubule. Replacing Wingless with a membrane-tethered version that is unable to diffuse from the Wingless producing cells results in aberrant patterning of the Malpighian tubule P-D axis and development of short, deformed ureters. This work directly demonstrates a patterning role for a released Wingless signal. As well as extending our understanding about the functional modes by which Wnts shape animal development, we anticipate this mechanism to be relevant to patterning epithelial tubes in other organs, such as the vertebrate kidney.


Subject(s)
Body Patterning , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Gene Expression Regulation, Developmental , Kidney Tubules, Distal/physiology , Kidney Tubules, Proximal/physiology , Wnt1 Protein/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Kidney Tubules, Distal/embryology , Kidney Tubules, Proximal/embryology , Morphogenesis , Wnt Signaling Pathway , Wnt1 Protein/genetics
5.
Am J Physiol Renal Physiol ; 315(6): F1637-F1643, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30110569

ABSTRACT

There is currently no technique to unambiguously diagnose antemortem kidney injury on postmortem examination since postmortem tissue damage and autolysis are common. We assessed the ability to detect kidney injury molecule-1 (KIM-1) expression in adult and fetal kidneys examined at autopsy. In adult kidneys ( n = 52 subjects), we found that the intensity of KIM-1 staining significantly correlated with the antemortem level of serum creatinine, and this was independent of the extent of tissue autolysis. In addition, kidneys from a total of 52 fetal/neonatal subjects, 30 stillborns and 22 liveborns, were assessed for KIM-1 staining. Given that serum creatinine is unreliable and often unavailable in fetuses and newborns, we assessed preterminal hypoxia in fetuses by the presence of squames in pulmonary alveoli and by required intubation. KIM-1 expression correlated with these clinical indexes of hypoxia. The expression of KIM-1 was seen in a majority of the fetal and neonatal autopsy kidneys (77%, 40/52) as early as 16 wk of gestation, even in the presence of autolysis. Thus KIM-1 is a specific and stable marker of antemortem tubular injury in kidneys of adults and fetuses despite postmortem autolysis.


Subject(s)
Acute Kidney Injury/metabolism , Hepatitis A Virus Cellular Receptor 1/analysis , Kidney Tubules, Proximal/chemistry , Postmortem Changes , Acute Kidney Injury/blood , Acute Kidney Injury/embryology , Age Factors , Autolysis , Autopsy , Biomarkers/blood , Creatinine/blood , Gestational Age , Humans , Immunohistochemistry , Infant, Newborn , Kidney Tubules, Proximal/embryology , Predictive Value of Tests , Retrospective Studies
6.
Int J Dev Biol ; 62(4-5): 319-324, 2018.
Article in English | MEDLINE | ID: mdl-29877571

ABSTRACT

LRPAP1, also known as receptor associated protein (RAP) is a small protein of 40 kDa associated with six of the seven members of the evolutionary conserved family of LDL receptors. Numerous studies showed that LRPAP1 has a dual function, initially as a chaperone insuring proper formation of intermolecular disulfide bonds during biogenesis of low density lipoprotein (LDL) receptors and later as an escort protein during trafficking through the endoplasmic reticulum and the early Golgi compartment, preventing premature interaction of receptor and ligand. Because of the general influence of LRPAP1 protein on lipid metabolism, we analyzed the temporal and spatial expression of the Xenopus laevis ortholog of lrpap1. Here, we show that lrpap1 was expressed in the developing neural system, the eye and ear anlagen, the branchial arches, the developing skin and the pronephric kidney. The very high expression level of lrpap1 specifically in the proximal tubules of the developing pronephros establishes this gene as a novel marker for the analysis of pronephros formation.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Kidney Tubules, Proximal/embryology , LDL-Receptor Related Protein-Associated Protein/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Biomarkers/metabolism , Embryonic Development/physiology , Kidney Tubules, Proximal/metabolism , LDL-Receptor Related Protein-Associated Protein/genetics , Organogenesis/physiology , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
7.
Development ; 144(24): 4530-4539, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29113990

ABSTRACT

Notch signaling plays important roles during mammalian nephrogenesis. To investigate whether Notch regulates nephron segmentation, we performed Notch loss-of-function and gain-of-function studies in developing nephrons in mice. Contrary to the previous notion that Notch signaling promotes the formation of proximal tubules and represses the formation of distal tubules in the mammalian nephron, we show that inhibition of Notch blocks the formation of all nephron segments and that constitutive activation of Notch in developing nephrons does not promote or repress the formation of a specific segment. Cells lacking Notch fail to form the S-shaped body and show reduced expression of Lhx1 and Hnf1b Consistent with this, we find that constitutive activation of Notch in mesenchymal nephron progenitors causes ectopic expression of Lhx1 and Hnf1b and that these cells eventually form a heterogeneous population that includes proximal tubules and other types of cells. Our data suggest that Notch signaling is required for the formation of all nephron segments and that it primes nephron progenitors for differentiation rather than directing their cell fates into a specific nephron segment.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Kidney Tubules, Proximal/embryology , Organogenesis/physiology , Receptors, Notch/metabolism , Animals , Cell Differentiation , Enzyme Activation/genetics , Hepatocyte Nuclear Factor 1-beta/biosynthesis , LIM-Homeodomain Proteins/biosynthesis , Mice , Mice, Transgenic , Receptors, Notch/genetics , Signal Transduction/physiology , Transcription Factors/biosynthesis , Wnt4 Protein/metabolism
8.
Development ; 144(19): 3625-3632, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28851704

ABSTRACT

Single-cell RNA-seq is a powerful technique. Nevertheless, there are important limitations, including the technical challenges of breaking down an organ or tissue into a single-cell suspension. Invariably, this has required enzymatic incubation at 37°C, which can be expected to result in artifactual changes in gene expression patterns. Here, we describe a dissociation method that uses a protease with high activity in the cold, purified from a psychrophilic microorganism. The entire procedure is carried out at 6°C or colder, at which temperature mammalian transcriptional machinery is largely inactive, thereby effectively 'freezing in' the in vivo gene expression patterns. To test this method, we carried out RNA-seq on 20,424 single cells from postnatal day 1 mouse kidneys, comparing the results of the psychrophilic protease method with procedures using 37°C incubation. We show that the cold protease method provides a great reduction in gene expression artifacts. In addition, the results produce a single-cell resolution gene expression atlas of the newborn mouse kidney, an interesting time in development when mature nephrons are present yet nephrogenesis remains extremely active.


Subject(s)
Artifacts , Kidney/embryology , Organogenesis , Peptide Hydrolases/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Gene Expression Profiling , Gene Expression Regulation, Developmental , Kidney/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/embryology , Mice , Stromal Cells/cytology , Stromal Cells/metabolism , Temperature , Time Factors
9.
Nephron ; 135(1): 77-86, 2017.
Article in English | MEDLINE | ID: mdl-27665619

ABSTRACT

BACKGROUND: Apoptosis regulates embryogenesis, organ metamorphosis and tissue homeostasis. Mitochondrial signaling is an apoptotic pathway, in which Cyt-c and Apaf-1 are transformed into an apoptosome, which activates procaspase-9 and triggers apoptosis. This study evaluated Cyt-c, Apaf-1 and caspase-9 expression during renal development. METHODS: Kidneys from embryonic (E) 16-, 18-, and 20-day-old fetuses and postnatal (P) 1-, 3-, 5-, 7-, 14-, and 21-day-old pups were obtained. Immunohistochemical analysis, dual-labeled immunofluorescence, terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) technique assay and Western blot were performed in addition to histological analysis. RESULTS: Immunohistochemistry showed that Cyt-c was strongly expressed in proximal and distal tubules (DTs) at all time points. Caspase-9 and Apaf-1 were strongly expressed in proximal tubules (PTs) but only weakly expressed in DTs. Dual-labeled immunofluorescence showed that most tubules expressed both Cyt-c and Apaf-1, except for some tubules that only expressed Cyt-c. The TUNEL assay showed a greater percentage of apoptotic cells in PTs compared to DTs. Apaf-1 and cleaved caspase-9 protein expression gradually increased during the embryonic period and peaked during the early postnatal period but apparently declined from P7. Cyt-c protein expression was weak during the embryonic period but obviously increased after P1. CONCLUSION: This study showed that PTs are more sensitive to apoptosis than DTs during rat renal development, even though both tubule segments contain a large number of mitochondria. Furthermore, Cyt-c-mediated mitochondrial apoptosis-related proteins play an important role in PTs during the early postnatal kidney development.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cytochromes c/metabolism , Kidney Tubules, Proximal/growth & development , Kidney Tubules, Proximal/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Female , Immunohistochemistry , Kidney Tubules, Proximal/embryology , Pregnancy , Rats , Rats, Sprague-Dawley
10.
Dev Dyn ; 245(4): 472-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26773453

ABSTRACT

BACKGROUND: Notch signaling in pronephros development has been shown to regulate establishment of glomus and proximal tubule, but how Notch signal works on competency of pronephric anlagen during the generation of pronephric components remains to be understood. RESULTS: We investigated how components of pronephros (glomus, proximal tubule, intermediate tubule, distal tubule, and connecting tubule) were generated in Xenopus embryos by timed overactivation and suppression of Notch signaling. Notch activation resulted in expansion of the glomus and disruption of the proximal tubule formation. Inhibition of Notch signaling reduced expression of wt1 and XSMP-30. In addition, when Notch signaling was overactivated at stage 20 on, intermediate, distal, and connecting tubule markers, gremlin and clcnkb, were decreased while Notch down-regulation increased gremlin and clcnkb. Similar changes were observed with segmental markers, cldn19, cldn14, and rhcg on activation or inhibition of Notch. Although Notch did not affect the expression of pan-pronephric progenitor marker, pax2, its activation inhibited lumen formation in the pronephros. CONCLUSIONS: Notch signal is essential for glomus and proximal tubule development and inhibition of Notch is critical for the differentiation of the intermediate, distal, and connecting tubule.


Subject(s)
Cell Differentiation/physiology , Embryo, Nonmammalian/embryology , Kidney Tubules, Proximal/embryology , Pronephros/embryology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Antigens, Differentiation/biosynthesis , Embryo, Nonmammalian/cytology , Gene Expression Regulation, Developmental/physiology , Kidney Tubules, Proximal/cytology , Pronephros/cytology , Xenopus laevis
11.
Nature ; 526(7574): 564-8, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26444236

ABSTRACT

The human kidney contains up to 2 million epithelial nephrons responsible for blood filtration. Regenerating the kidney requires the induction of the more than 20 distinct cell types required for excretion and the regulation of pH, and electrolyte and fluid balance. We have previously described the simultaneous induction of progenitors for both collecting duct and nephrons via the directed differentiation of human pluripotent stem cells. Paradoxically, although both are of intermediate mesoderm in origin, collecting duct and nephrons have distinct temporospatial origins. Here we identify the developmental mechanism regulating the preferential induction of collecting duct versus kidney mesenchyme progenitors. Using this knowledge, we have generated kidney organoids that contain nephrons associated with a collecting duct network surrounded by renal interstitium and endothelial cells. Within these organoids, individual nephrons segment into distal and proximal tubules, early loops of Henle, and glomeruli containing podocytes elaborating foot processes and undergoing vascularization. When transcription profiles of kidney organoids were compared to human fetal tissues, they showed highest congruence with first trimester human kidney. Furthermore, the proximal tubules endocytose dextran and differentially apoptose in response to cisplatin, a nephrotoxicant. Such kidney organoids represent powerful models of the human organ for future applications, including nephrotoxicity screening, disease modelling and as a source of cells for therapy.


Subject(s)
Cell Lineage , Induced Pluripotent Stem Cells/cytology , Models, Biological , Nephrons/cytology , Nephrons/embryology , Organogenesis , Organoids/cytology , Animals , Coculture Techniques , Feeder Cells , Fetus/anatomy & histology , Fetus/cytology , Fetus/embryology , Fibroblasts/cytology , Humans , Kidney Tubules, Collecting/cytology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/physiology , Mesoderm/cytology , Mice , Nephrons/anatomy & histology , Nephrons/physiology , Organoids/embryology , Tissue Culture Techniques
12.
Kidney Int ; 87(6): 1125-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25651362

ABSTRACT

MicroRNAs, activated by the enzyme Dicer1, control post-transcriptional gene expression. Dicer1 has important roles in the epithelium during nephrogenesis, but its function in stromal cells during kidney development is unknown. To study this, we inactivated Dicer1 in renal stromal cells. This resulted in hypoplastic kidneys, abnormal differentiation of the nephron tubule and vasculature, and perinatal mortality. In mutant kidneys, genes involved in stromal cell migration and activation were suppressed as were those involved in epithelial and endothelial differentiation and maturation. Consistently, polarity of the proximal tubule was incorrect, distal tubule differentiation was diminished, and elongation of Henle's loop attenuated resulting in lack of inner medulla and papilla in stroma-specific Dicer1 mutants. Glomerular maturation and capillary loop formation were abnormal, whereas peritubular capillaries, with enhanced branching and increased diameter, formed later. In Dicer1-null renal stromal cells, expression of factors associated with migration, proliferation, and morphogenic functions including α-smooth muscle actin, integrin-α8, -ß1, and the WNT pathway transcriptional regulator LEF1 were reduced. Dicer1 mutation in stroma led to loss of expression of distinct microRNAs. Of these, miR-214, -199a-5p, and -199a-3p regulate stromal cell functions ex vivo, including WNT pathway activation, migration, and proliferation. Thus, Dicer1 activity in the renal stromal compartment regulates critical stromal cell functions that, in turn, regulate differentiation of the nephron and vasculature during nephrogenesis.


Subject(s)
Cell Differentiation/genetics , DEAD-box RNA Helicases/physiology , Neovascularization, Physiologic/genetics , Nephrons/embryology , Ribonuclease III/physiology , Actins/metabolism , Animals , Capillaries/embryology , Cell Movement/genetics , Cell Proliferation/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Gene Expression , Integrin alpha Chains/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/cytology , Kidney Glomerulus/embryology , Kidney Tubules/blood supply , Kidney Tubules/cytology , Kidney Tubules/embryology , Kidney Tubules, Distal/blood supply , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/embryology , Kidney Tubules, Proximal/blood supply , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/embryology , Loop of Henle/blood supply , Loop of Henle/cytology , Loop of Henle/embryology , Mice , MicroRNAs/genetics , Nephrons/abnormalities , Nephrons/cytology , Organogenesis/genetics , Podocytes/physiology , Ribonuclease III/genetics , Ribonuclease III/metabolism , Stromal Cells/physiology , Transcriptome , Ureter/abnormalities , Wnt Signaling Pathway/genetics
13.
Dev Biol ; 399(1): 100-116, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25542995

ABSTRACT

The mechanisms that establish nephron segments are poorly understood. The zebrafish embryonic kidney, or pronephros, is a simplified yet conserved genetic model to study this renal development process because its nephrons contain segments akin to other vertebrates, including the proximal convoluted and straight tubules (PCT, PST). The zebrafish pronephros is also associated with the corpuscles of Stannius (CS), endocrine glands that regulate calcium and phosphate homeostasis, but whose ontogeny from renal progenitors is largely mysterious. Initial patterning of zebrafish renal progenitors in the intermediate mesoderm (IM) involves the formation of rostral and caudal domains, the former being reliant on retinoic acid (RA) signaling, and the latter being repressed by elevated RA levels. Here, using expression profiling to gain new insights into nephrogenesis, we discovered that the gene single minded family bHLH transcription factor 1a (sim1a) is dynamically expressed in the renal progenitors-first marking the caudal domain, then becoming restricted to the proximal segments, and finally exhibiting specific CS expression. In loss of function studies, sim1a knockdown expanded the PCT and abrogated both the PST and CS populations. Conversely, overexpression of sim1a modestly expanded the PST and CS, while it reduced the PCT. These results show that sim1a activity is necessary and partially sufficient to induce PST and CS fates, and suggest that sim1a may inhibit PCT fate and/or negotiate the PCT/PST boundary. Interestingly, the sim1a expression domain in renal progenitors is responsive to altered levels of RA, suggesting that RA regulates sim1a, directly or indirectly, during nephrogenesis. sim1a deficient embryos treated with exogenous RA formed nephrons that were predominantly composed of PCT segments, but lacked the enlarged PST observed in RA treated wild-types, indicating that RA is not sufficient to rescue the PST in the absence of sim1a expression. Alternately, when sim1a knockdowns were exposed to the RA inhibitor diethylaminobenzaldehyde (DEAB), the CS was abrogated rather than expanded as seen in DEAB treated wild-types, revealing that CS formation in the absence of sim1a cannot be rescued by RA biosynthesis abrogation. Taken together, these data reveal previously unappreciated roles for sim1a in zebrafish pronephric proximal tubule and CS patterning, and are consistent with the model that sim1a acts downstream of RA to mitigate the formation of these lineages. These findings provide new insights into the genetic pathways that direct nephron development, and may have implications for understanding renal birth defects and kidney reprogramming.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Kidney Tubules, Proximal/metabolism , Nephrons/metabolism , Repressor Proteins/genetics , Tretinoin/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , In Situ Hybridization , Kidney Tubules, Proximal/embryology , Nephrons/embryology , Organogenesis/genetics , Repressor Proteins/metabolism , Time Factors , Tretinoin/pharmacology , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
14.
Dev Biol ; 394(1): 54-64, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25127994

ABSTRACT

The kidney is a homeostatic organ required for waste excretion and reabsorption of water, salts and other macromolecules. To this end, a complex series of developmental steps ensures the formation of a correctly patterned and properly proportioned organ. While previous studies have mainly focused on the individual signaling pathways, the formation of higher order receptor complexes in lipid rafts is an equally important aspect. These membrane platforms are characterized by differences in local lipid and protein compositions. Indeed, the cells in the Xenopus pronephric kidney were positive for the lipid raft markers ganglioside GM1 and Caveolin-1. To specifically interfere with lipid raft function in vivo, we focused on the Sterol Carrier Protein 2 (scp2), a multifunctional protein that is an important player in remodeling lipid raft composition. In Xenopus, scp2 mRNA was strongly expressed in differentiated epithelial structures of the pronephric kidney. Knockdown of scp2 did not interfere with the patterning of the kidney along its proximo-distal axis, but dramatically decreased the size of the kidney, in particular the proximal tubules. This phenotype was accompanied by a reduction of lipid rafts, but was independent of the peroxisomal or transcriptional activities of scp2. Finally, disrupting lipid micro-domains by inhibiting cholesterol synthesis using Mevinolin phenocopied the defects seen in scp2 morphants. Together these data underscore the importance for localized signaling platforms in the proper formation of the Xenopus kidney.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Kidney Tubules, Proximal/embryology , Membrane Microdomains/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Anticholesteremic Agents/pharmacology , Body Patterning/genetics , Cell Line , Cholesterol/biosynthesis , Gene Knockdown Techniques , HEK293 Cells , Humans , Kidney Tubules, Proximal/physiology , Lovastatin/pharmacology , Membrane Microdomains/physiology , Morpholinos , RNA, Messenger/biosynthesis , Transcription, Genetic
15.
PLoS One ; 9(6): e97806, 2014.
Article in English | MEDLINE | ID: mdl-24905828

ABSTRACT

Kidney tubule plays a critical role in recovering or secreting solutes, but the detailed morphogenesis remains unclear. Our previous studies have found that grouper tshß (gtshß) is also expressed in kidney, however, the distribution significance is still unknown. To understand the gtshß role and kidney tubule morphogenesis, here, we have generated a transgenic zebrafish line Tg(gtshß:GFP) with green fluorescent protein driven by the gtshß promoter. Similar to the endogenous tshß in zebrafish or in grouper, the gtshß promoter-driven GFP is expressed in pituitary and kidney, and the developing details of proximal kidney tubule are marked in the transgenic zebrafish line. The gfp initially transcribes at 16 hours post fertilization (hpf) above the dorsal mesentery, and partially co-localizes with pronephric tubular markers slc20a1a and cdh17. Significantly, the GFP specifically localizes in proximal pronephric segments during embryogenesis and resides at kidney duct epithelium in adult fish. To test whether the gtshß promoter-driven GFP may serve as a readout signal of the tubular development, we have treated the embryos with retinoic acid signaing (RA) reagents, in which exogenous RA addition results in a distal extension of the proximal segments, while RA inhibition induces a weakness and shortness of the proximal segments. Therefore, this transgenic line provides a useful tool for genetic or chemical analysis of kidney tubule.


Subject(s)
Gene Expression Regulation, Developmental , Kidney Tubules, Proximal/metabolism , Thyrotropin, beta Subunit/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Epithelial Cells/metabolism , Kidney Tubules, Proximal/embryology , Promoter Regions, Genetic , Thyrotropin, beta Subunit/genetics , Zebrafish , Zebrafish Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 111(17): 6335-40, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733901

ABSTRACT

MicroRNAs (miRNAs) are major posttranscriptional regulators of a wide variety of biological processes. However, redundancy among most miRNAs has made it difficult to identify their in vivo functions. We previously demonstrated that global inhibition of miRNA biogenesis in Xenopus resulted in a dramatically smaller pronephric kidney. This suggested that microRNAs play a pivotal role in organ size control. Here we now provide a detailed mechanistic explanation for this phenotype. We identified that the activation of the mechanistic target of rapamycin complex 1 (mTORC1) by Insulin and insulin-like growth factor (Igf) 2 is an important regulator in kidney growth, which in turn is modulated by microRNAs. Molecular analyses demonstrate that microRNAs set a threshold for mTORC1 signaling by down-regulating one of its core negative regulators, tuberous sclerosis 1 (Tsc1). Most importantly, this rheostat can be reprogrammed experimentally. Whereas knockdown of miRNAs causes growth arrest, concomitant knockdown of Tsc1 restores mTORC1 activity and proximal tubular size. Together, these data establish a previously unidentified in vivo paradigm for the importance of posttranscriptional regulation in organ size control.


Subject(s)
Kidney/anatomy & histology , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Xenopus/genetics , Animals , Gene Expression Regulation, Developmental , Insulin/metabolism , Insulin-Like Growth Factor II/metabolism , Kidney/embryology , Kidney/metabolism , Kidney Tubules, Proximal/anatomy & histology , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/metabolism , LLC-PK1 Cells , Mechanistic Target of Rapamycin Complex 1 , MicroRNAs/genetics , Organ Size/genetics , Signal Transduction/genetics , Swine , Tuberous Sclerosis Complex 1 Protein , Xenopus/embryology
17.
J Am Soc Nephrol ; 25(6): 1211-25, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24357672

ABSTRACT

Human pluripotent stem cells (hPSCs) can generate a diversity of cell types, but few methods have been developed to derive cells of the kidney lineage. Here, we report a highly efficient system for differentiating human embryonic stem cells and induced pluripotent stem cells (referred to collectively as hPSCs) into cells expressing markers of the intermediate mesoderm (IM) that subsequently form tubule-like structures. Treatment of hPSCs with the glycogen synthase kinase-3ß inhibitor CHIR99021 induced BRACHYURY(+)MIXL1(+) mesendoderm differentiation with nearly 100% efficiency. In the absence of additional exogenous factors, CHIR99021-induced mesendodermal cells preferentially differentiated into cells expressing markers of lateral plate mesoderm with minimal IM differentiation. However, the sequential treatment of hPSCs with CHIR99021 followed by fibroblast growth factor-2 and retinoic acid generated PAX2(+)LHX1(+) cells with 70%-80% efficiency after 3 days of differentiation. Upon growth factor withdrawal, these PAX2(+)LHX1(+) cells gave rise to apically ciliated tubular structures that coexpressed the proximal tubule markers Lotus tetragonolobus lectin, N-cadherin, and kidney-specific protein and partially integrated into embryonic kidney explant cultures. With the addition of FGF9 and activin, PAX2(+)LHX1(+) cells specifically differentiated into cells expressing SIX2, SALL1, and WT1, markers of cap mesenchyme nephron progenitor cells. Our findings demonstrate the effective role of fibroblast growth factor signaling in inducing IM differentiation in hPSCs and establish the most rapid and efficient system whereby hPSCs can be differentiated into cells with features characteristic of kidney lineage cells.


Subject(s)
Cell Differentiation/physiology , Kidney Tubules, Proximal/cytology , Mesoderm/cytology , Pluripotent Stem Cells/cytology , Animals , Antineoplastic Agents/pharmacology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Foreskin/cytology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/metabolism , LIM-Homeodomain Proteins/metabolism , Male , Mesoderm/embryology , Mesoderm/metabolism , Mice , PAX2 Transcription Factor/metabolism , Pluripotent Stem Cells/drug effects , Pregnancy , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factors/metabolism , Tretinoin/pharmacology
18.
Mol Pharmacol ; 84(6): 808-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038112

ABSTRACT

The transcriptional regulation of drug-metabolizing enzymes and transporters (here collectively referred to as DMEs) in the developing proximal tubule (PT) is not well understood. As in the liver, DME regulation in the PT may be mediated through nuclear receptors, which are thought to "sense" deviations from homeostasis by being activated by ligands, some of which are handled by DMEs, including drug transporters. Systems analysis of transcriptomic data during kidney development predicted a set of upstream transcription factors, including hepatocyte nuclear factor 4α (Hnf4a) and Hnf1a, as well as Nr3c1 (Gr), Nfe2l2 (Nrf2), peroxisome proliferator-activated receptor α (Pparα), and Tp53. Motif analysis of cis-regulatory enhancers further suggested that Hnf4a and Hnf1a are the main transcriptional regulators of DMEs in the PT. Available expression data from tissue-specific Hnf4a knockout tissues revealed that distinct subsets of DMEs were regulated by Hnf4a in a tissue-specific manner. Chromatin immunoprecipitation combined with massively parallel DNA sequencing was performed to characterize the PT-specific binding sites of Hnf4a in rat kidneys at three developmental stages (prenatal, immature, adult), which further supported a major role for Hnf4a in regulating PT gene expression, including DMEs. In ex vivo kidney organ culture, an antagonist of Hnf4a (but not a similar inactive compound) led to predicted changes in DME expression, including among others Fmo1, Cyp2d2, Cyp2d4, Nqo2, as well as organic cation transporters and organic anion transporters Slc22a1 (Oct1), Slc22a2 (Oct2), Slc22a6 (Oat1), Slc22a8 (Oat3), and Slc47a1 (Mate1). Conversely, overexpression of Hnf1a and Hnf4a in primary mouse embryonic fibroblasts, sometimes considered a surrogate for mesenchymal stem cells, induced expression of several of these proximal tubule DMEs, as well as epithelial markers and a PT-enriched brush border marker Ggt1. These cells had organic anion transporter function. Taken together, the data strongly supports a critical role for HNF4a and Hnf1a in the tissue-specific regulation of drug handling and differentiation toward a PT-like cellular identity. We discuss our data in the context of the "remote sensing and signaling hypothesis" (Ahn and Nigam, 2009; Wu et al., 2011).


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Inactivation, Metabolic/genetics , Kidney/metabolism , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/genetics , Kidney/embryology , Kidney/growth & development , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/growth & development , Kidney Tubules, Proximal/metabolism , Lentivirus/genetics , Metabolic Detoxication, Phase I/genetics , Metabolic Detoxication, Phase II/genetics , Mice , Mice, Knockout , Protein Binding , Rats , Tissue Culture Techniques
19.
PLoS One ; 8(2): e56731, 2013.
Article in English | MEDLINE | ID: mdl-23468876

ABSTRACT

Maternal hyperglycemia can inhibit morphogenesis of ureteric bud branching, Glial cell line-derived neurotrophilic factor (GDNF) is a key regulator of the initiation of ureteric branching. Early growth response gene-1 (EGR-1) is an immediate early gene. Preliminary study found EGR-1 persistently expressed with GDNF in hyperglycemic environment. To evaluate the potential relationship of hyperglycemia-GDNF-EGR-1 pathway, in vitro human renal proximal tubular epithelial (HRPTE) cells as target and in vivo streptozotocin-induced mice model were used. Our in vivo microarray, real time-PCR and confocal morphological observation confirmed apoptosis in hyperglycemia-induced fetal nephropathy via activation of the GDNF/MAPK/EGR-1 pathway at E12-E15. Detachment between ureteric branch and metanephrons, coupled with decreasing number and collapse of nephrons on Day 1 newborn mice indicate hyperglycemic environment suppress ureteric bud to invade metanephric rudiment. In vitro evidence proved that high glucose suppressed HRPTE cell migration and enhanced GDNF-EGR-1 pathway, inducing HRPTE cell apoptosis. Knockdown of EGR-1 by siRNA negated hyperglycemic suppressed GDNF-induced HRPTE cells. EGR-1 siRNA also reduced GDNF/EGR-1-induced cRaf/MEK/ERK phosphorylation by 80%. Our findings reveal a novel mechanism of GDNF/MAPK/EGR-1 activation playing a critical role in HRPTE cell migration, apoptosis and fetal hyperglycemic nephropathy.


Subject(s)
Hyperglycemia/complications , Kidney Diseases/etiology , Maternal Exposure/adverse effects , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Embryonic Development/genetics , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Genome-Wide Association Study , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Hyperglycemia/genetics , Kidney Diseases/genetics , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis/genetics , Phosphorylation/drug effects , Pregnancy , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
20.
Dev Biol ; 376(1): 31-42, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23352791

ABSTRACT

In the kidney, proximal tubules are very important for the reabsorption of water, ions and organic solutes from the primary urine. They are composed of highly specialized epithelial cells that are characterized by an elaborate apical brush border to increase transport efficiency. Using the pronephric kidney of Xenopus laevis we discovered that the G-protein modulator cholera toxin resulted in a dramatic reduction of the proximal tubular size. This phenotype was accompanied by changes in the cytoarchitecture characterized by ectopic expression of the distal tubular marker 4A6 and an impairment of yolk platelet degradation. In addition, cholera toxin caused edema formation. However, this phenotype was not due to kidney defects, but rather due to impaired vasculature development. Based on experiments with antisense morpholino oligomers as well as pharmacological agonists and antagonists, we could show that the complex phenotype of cholera toxin in the pronephric kidney was caused by the hyperactivation of a single G-protein alpha subunit, Gnas. This-in turn-caused elevated cAMP levels, triggered a Rapgef4-dependent signaling cassette and perturbed exo- and endocytosis. This perturbation of the secretory pathway by Ctx was not only observed in Xenopus embryos. Also, in a human proximal tubular cell line, cholera toxin or a Rapgef4-specific agonist increased uptake and decreased secretion of FITC-labeled Albumin. Based on these data we propose that the Gnas/cAMP/Rapgef4 pathway regulates the signals inducing the proliferation of proximal tubules to acquire their final organ size.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Proteins/metabolism , Kidney Tubules, Proximal/embryology , Pronephros/embryology , Signal Transduction/physiology , Xenopus laevis/embryology , Albumins , Animals , Cell Line , Cholera Toxin/pharmacology , Cyclic AMP/metabolism , DNA Primers/genetics , Fluorescein-5-isothiocyanate , Guanine Nucleotide Exchange Factors/metabolism , Histological Techniques , Humans , Immunohistochemistry , In Situ Hybridization , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Microscopy, Electron, Transmission , Pronephros/metabolism , Pronephros/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...