Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(6): e2309466121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38300866

ABSTRACT

Congenital anomalies of the lower genitourinary (LGU) tract are frequently comorbid due to genetically linked developmental pathways, and are among the most common yet most socially stigmatized congenital phenotypes. Genes involved in sexual differentiation are prime candidates for developmental anomalies of multiple LGU organs, but insufficient prospective screening tools have prevented the rapid identification of causative genes. Androgen signaling is among the most influential modulators of LGU development. The present study uses SpDamID technology in vivo to generate a comprehensive map of the pathways actively regulated by the androgen receptor (AR) in the genitalia in the presence of the p300 coactivator, identifying wingless/integrated (WNT) signaling as a highly enriched AR-regulated pathway in the genitalia. Transcription factor (TF) hits were then assayed for sexually dimorphic expression at two critical time points and also cross-referenced to a database of clinically relevant copy number variations to identify 252 TFs exhibiting copy variation in patients with LGU phenotypes. A subset of 54 TFs was identified for which LGU phenotypes are statistically overrepresented as a proportion of total observed phenotypes. The 252 TF hitlist was then subjected to a functional screen to identify hits whose silencing affects genital mesenchymal growth rates. Overlap of these datasets results in a refined list of 133 TFs of both functional and clinical relevance to LGU development, 31 of which are top priority candidates, including the well-documented renal progenitor regulator, Sall1. Loss of Sall1 was examined in vivo and confirmed to be a powerful regulator of LGU development.


Subject(s)
DNA Copy Number Variations , Urinary Tract , Humans , Prospective Studies , Androgens/metabolism , Genitalia/metabolism , Urinary Tract/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Development ; 150(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38014633

ABSTRACT

Sall1 and Sall4 (Sall1/4), zinc-finger transcription factors, are expressed in the progenitors of the second heart field (SHF) and in cardiomyocytes during the early stages of mouse development. To understand the function of Sall1/4 in heart development, we generated heart-specific Sall1/4 functionally inhibited mice by forced expression of the truncated form of Sall4 (ΔSall4) in the heart. The ΔSall4-overexpression mice exhibited a hypoplastic right ventricle and outflow tract, both of which were derived from the SHF, and a thinner ventricular wall. We found that the numbers of proliferative SHF progenitors and cardiomyocytes were reduced in ΔSall4-overexpression mice. RNA-sequencing data showed that Sall1/4 act upstream of the cyclin-dependent kinase (CDK) and cyclin genes, and of key transcription factor genes for the development of compact cardiomyocytes, including myocardin (Myocd) and serum response factor (Srf). In addition, ChIP-sequencing and co-immunoprecipitation analyses revealed that Sall4 and Myocd form a transcriptional complex with SRF, and directly bind to the upstream regulatory regions of the CDK and cyclin genes (Cdk1 and Ccnb1). These results suggest that Sall1/4 are critical for the proliferation of cardiac cells via regulation of CDK and cyclin genes that interact with Myocd and SRF.


Subject(s)
Cyclin-Dependent Kinases , Myocytes, Cardiac , Animals , Mice , Cell Proliferation/genetics , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , Myocytes, Cardiac/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism , Transcription Factors/metabolism
3.
Development ; 150(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37519269

ABSTRACT

Changes in gene expression represent an important source of phenotypic innovation. Yet how such changes emerge and impact the evolution of traits remains elusive. Here, we explore the molecular mechanisms associated with the development of masculinizing ovotestes in female moles. By performing integrative analyses of epigenetic and transcriptional data in mole and mouse, we identified the co-option of SALL1 expression in mole ovotestes formation. Chromosome conformation capture analyses highlight a striking conservation of the 3D organization at the SALL1 locus, but an evolutionary divergence of enhancer activity. Interspecies reporter assays support the capability of mole-specific enhancers to activate transcription in urogenital tissues. Through overexpression experiments in transgenic mice, we further demonstrate the capability of SALL1 to induce kidney-related gene programs, which are a signature of mole ovotestes. Our results highlight the co-option of gene expression, through changes in enhancer activity, as a plausible mechanism for the evolution of traits.


Subject(s)
Kidney , Moles , Animals , Female , Mice , Kidney/metabolism , Mice, Transgenic , Moles/genetics
4.
Nephrology (Carlton) ; 29(8): 541-546, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38584358

ABSTRACT

Townes-Brocks syndrome (TBS) is an autosomal dominant disorder characterised by the triad of anorectal, thumb, and ear malformations. It may also be accompanied by defects in kidney, heart, eyes, hearing, and feet. TBS has been demonstrated to result from heterozygous variants in the SALL1 gene, which encodes zinc finger protein believed to function as a transcriptional repressor. The clinical characteristics of an atypical TBS phenotype patient from a Chinese family are described, with predominant manifestations including external ear dysplasia, unilateral renal hypoplasia with mild renal dysfunction, and hearing impairment. A novel heterozygous variant c.3060T>A (p.Tyr1020*) in exon 2 of the SALL1 gene was identified in this proband. Pyrosequencing of the complementary DNA of the proband revealed that the variant transcript accounted for 48% of the total transcripts in peripheral leukocytes, indicating that this variant transcript has not undergone nonsense-mediated mRNA decay. This variant c.3060T > A is located at the terminal end of exon 2, proximal to the 3' end of the SALL1 gene, and exerts a relatively minor impact on protein function. We suggest that the atypical TBS phenotype observed in the proband may be attributed to the truncated protein retaining partial SALL1 function.


Subject(s)
Abnormalities, Multiple , Hearing Loss, Sensorineural , Transcription Factors , Female , Humans , Male , Abnormalities, Multiple/genetics , Anus, Imperforate/genetics , Anus, Imperforate/diagnosis , China , DNA Mutational Analysis , Ear/abnormalities , East Asian People/genetics , Genetic Predisposition to Disease , Heredity , Heterozygote , Mutation , Pedigree , Phenotype , Thumb/abnormalities , Tracheoesophageal Fistula/genetics , Transcription Factors/genetics
5.
Adv Exp Med Biol ; 1441: 505-534, 2024.
Article in English | MEDLINE | ID: mdl-38884729

ABSTRACT

Ventricular septal defects (VSDs) are recognized as one of the commonest congenital heart diseases (CHD), accounting for up to 40% of all cardiac malformations, and occur as isolated CHDs as well as together with other cardiac and extracardiac congenital malformations in individual patients and families. The genetic etiology of VSD is complex and extraordinarily heterogeneous. Chromosomal abnormalities such as aneuploidy and structural variations as well as rare point mutations in various genes have been reported to be associated with this cardiac defect. This includes both well-defined syndromes with known genetic cause (e.g., DiGeorge syndrome and Holt-Oram syndrome) and so far undefined syndromic forms characterized by unspecific symptoms. Mutations in genes encoding cardiac transcription factors (e.g., NKX2-5 and GATA4) and signaling molecules (e.g., CFC1) have been most frequently found in VSD cases. Moreover, new high-resolution methods such as comparative genomic hybridization enabled the discovery of a high number of different copy number variations, leading to gain or loss of chromosomal regions often containing multiple genes, in patients with VSD. In this chapter, we will describe the broad genetic heterogeneity observed in VSD patients considering recent advances in this field.


Subject(s)
Heart Septal Defects, Ventricular , Humans , Chromosome Aberrations , DNA Copy Number Variations/genetics , Genetic Predisposition to Disease/genetics , Heart Septal Defects, Ventricular/genetics , Mutation , Transcription Factors/genetics
6.
J Gene Med ; 25(7): e3127, 2023 07.
Article in English | MEDLINE | ID: mdl-31693770

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is a serious health threat for humans worldwide. Recent studies have revealed that microRNAs are associated with the progression of human cancers, including PCa. However, no study has been performed aiming to investigate the role of microNA-4286 (miR-4286) on PCa. METHODS: A quantitative reverse transcriptase-polymerase chain reaction was conducted to analyze the expression level of miR-4286 in PCa cells. The connection of miR-4286 and spalt like transcription factor 1 (SALL1) was analyzed with a bioinformatic analysis tool, a dual-luciferase activity reporter assay and western blotting. The effects of miR-4286 and SALL1 on PCa cell behaviors were examined in vitro. RESULTS: We showed miR-4286 expression was significantly increased in PCa cells compared to a normal cell line. Knockdown of miR-4286 could inhibit PCa cell proliferation but promote cell apoptosis by targeting SALL1. CONCLUSIONS: The results of the present study suggest that miR-4286 overexpression represents a tumor promoter role in PCa.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Humans , Male , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/pathology
7.
Glia ; 70(9): 1720-1733, 2022 09.
Article in English | MEDLINE | ID: mdl-35567352

ABSTRACT

Microglia play many critical roles in neural development. Recent single-cell RNA-sequencing studies have found diversity of microglia both across different stages and within the same stage in the developing brain. However, how such diversity is controlled during development is poorly understood. In this study, we first found the expression of the macrophage mannose receptor CD206 in early-stage embryonic microglia on mouse brain sections. This expression showed a sharp decline between E12.5 and E13.5 across the central nervous system. We next tested the roles of the microglia-expressed zinc finger transcription factor SALL1 in this early transition of gene expression. By deleting Sall1 specifically in microglia, we found that many microglia continued to express CD206 when it is normally downregulated. In addition, the mutant microglia continued to show less ramified morphology in comparison with controls even into postnatal stages. Thus, SALL1 is required for early microglia to transition into a more mature status during development.


Subject(s)
Gene Expression Regulation, Developmental , Microglia , Neurogenesis , Transcription Factors , Zinc Fingers , Animals , Gene Expression Regulation, Developmental/genetics , Mice , Microglia/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers/genetics , Zinc Fingers/physiology
8.
Am J Hum Genet ; 102(2): 249-265, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395072

ABSTRACT

Townes-Brocks syndrome (TBS) is characterized by a spectrum of malformations in the digits, ears, and kidneys. These anomalies overlap those seen in a growing number of ciliopathies, which are genetic syndromes linked to defects in the formation or function of the primary cilia. TBS is caused by mutations in the gene encoding the transcriptional repressor SALL1 and is associated with the presence of a truncated protein that localizes to the cytoplasm. Here, we provide evidence that SALL1 mutations might cause TBS by means beyond its transcriptional capacity. By using proximity proteomics, we show that truncated SALL1 interacts with factors related to cilia function, including the negative regulators of ciliogenesis CCP110 and CEP97. This most likely contributes to more frequent cilia formation in TBS-derived fibroblasts, as well as in a CRISPR/Cas9-generated model cell line and in TBS-modeled mouse embryonic fibroblasts, than in wild-type controls. Furthermore, TBS-like cells show changes in cilia length and disassembly rates in combination with aberrant SHH signaling transduction. These findings support the hypothesis that aberrations in primary cilia and SHH signaling are contributing factors in TBS phenotypes, representing a paradigm shift in understanding TBS etiology. These results open possibilities for the treatment of TBS.


Subject(s)
Abnormalities, Multiple/genetics , Anus, Imperforate/genetics , Cilia/metabolism , Hearing Loss, Sensorineural/genetics , Mutation/genetics , Thumb/abnormalities , Transcription Factors/genetics , Animals , Cytoplasm/metabolism , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Infant, Newborn , Mice , Phenotype , Protein Binding , Proteomics , Signal Transduction
9.
Am J Med Genet A ; 185(3): 937-944, 2021 03.
Article in English | MEDLINE | ID: mdl-33438842

ABSTRACT

Townes-Brocks syndrome (TBS) is a rare autosomal dominant syndrome, resulting from heterozygous variant in SALL1 gene and initially characterized by the triad of anorectal, thumb, and ear malformations. Essentially described in children, adult case reports are uncommon. Renal involvement has already been reported in adults and children but poorly described. Structural abnormalities such as hypodysplasia, unilateral renal agenesis or multicystic kidneys have been described, as well as functional impairment (with or without structural abnormalities) that may progress to end-stage renal disease (ESRD). We report two adult cases (mother and daughter) which exhibited kidney hypoplasia (focal and segmental glomerulosclerosis for the mother) and ESRD. The mother had unilateral polydactyly. TBS was suggested after physical examination. TBS diagnosis was confirmed by identification of a SALL1 variant. We conducted a literature review to evaluate the renal anomalies in TBS cases diagnosed in adulthood. Among 44 adult cases of TBS with genetic confirmation (including our two cases), 10 had kidney disease. The circumstances of renal failure diagnosis were incidental findings (2/5), gout (2/5), or repeated episodes of pyelonephritis (1/5). The median age of kidney disease diagnosis was 30 years old and of renal transplant 49 years old. The most frequent renal malformation was bilateral kidney hypoplasia. TBS is probably underestimated in adulthood and this report highlights that less obvious elements of morphology such as dysplasic ears can facilitate the diagnosis of TBS. As long-term prognosis of renal involvement in TBS patients remains largely unknown, a regular evaluation is required throughout life for patients.


Subject(s)
Anus, Imperforate/complications , Hearing Loss, Sensorineural/complications , Kidney Failure, Chronic/etiology , Thumb/abnormalities , Transcription Factors/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abortion, Habitual/genetics , Anus, Imperforate/diagnosis , Anus, Imperforate/genetics , Delayed Diagnosis , Ear, External/abnormalities , Female , Hammer Toe Syndrome/genetics , Hearing Loss, Bilateral/genetics , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Humans , Kidney Failure, Chronic/surgery , Kidney Failure, Chronic/therapy , Kidney Transplantation , Middle Aged , Pedigree , Peripheral Nervous System Diseases/genetics , Phenotype , Polydactyly/genetics , Renal Dialysis , Retinal Dystrophies/genetics
10.
Dev Dyn ; 249(10): 1217-1242, 2020 10.
Article in English | MEDLINE | ID: mdl-32492250

ABSTRACT

BACKGROUND: Understanding the mechanisms that regulate hair cell (HC) differentiation in the organ of Corti (OC) is essential to designing genetic therapies for hearing loss due to HC loss or damage. We have previously identified Fibroblast Growth Factor 20 (FGF20) as having a key role in HC and supporting cell differentiation in the mouse OC. To investigate the genetic landscape regulated by FGF20 signaling in OC progenitors, we employ Translating Ribosome Affinity Purification combined with Next Generation RNA Sequencing (TRAPseq) in the Fgf20 lineage. RESULTS: We show that TRAPseq targeting OC progenitors effectively enriched for RNA from this rare cell population. TRAPseq identified differentially expressed genes (DEGs) downstream of FGF20, including Etv4, Etv5, Etv1, Dusp6, Hey1, Hey2, Heyl, Tectb, Fat3, Cpxm2, Sall1, Sall3, and cell cycle regulators such as Cdc20. Analysis of Cdc20 conditional-null mice identified decreased cochlea length, while analysis of Sall1-null and Sall1-ΔZn2-10 mice, which harbor a mutation that causes Townes-Brocks syndrome, identified a decrease in outer hair cell number. CONCLUSIONS: We present two datasets: genes with enriched expression in OC progenitors, and DEGs downstream of FGF20 in the embryonic day 14.5 cochlea. We validate select DEGs via in situ hybridization and in vivo functional studies in mice.


Subject(s)
Fibroblast Growth Factors/physiology , Organ of Corti/metabolism , Ribosomes/metabolism , Animals , Cell Differentiation , Fibroblast Growth Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Therapy , Hair Cells, Auditory, Outer/metabolism , Hearing , Mice , Mice, Transgenic , Mutation , Neurogenesis , Organ of Corti/embryology , Phenotype , Protein Biosynthesis , Sequence Analysis, RNA , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Development ; 144(17): 3080-3094, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28760814

ABSTRACT

The formation of the proper number of nephrons requires a tightly regulated balance between renal progenitor cell self-renewal and differentiation. The molecular pathways that regulate the transition from renal progenitor to renal vesicle are not well understood. Here, we show that Sall1interacts with the nucleosome remodeling and deacetylase complex (NuRD) to inhibit premature differentiation of nephron progenitor cells. Disruption of Sall1-NuRD in vivo in knock-in mice (ΔSRM) resulted in accelerated differentiation of nephron progenitors and bilateral renal hypoplasia. Transcriptional profiling of mutant kidneys revealed a striking pattern in which genes of the glomerular and proximal tubule lineages were either unchanged or upregulated, and those in the loop of Henle and distal tubule lineages were downregulated. These global changes in gene expression were accompanied by a significant decrease in THP-, NKCC2- and AQP1-positive loop of Henle nephron segments in mutant ΔSRM kidneys. These findings highlight an important function of Sall1-NuRD interaction in the regulation of Six2-positive multipotent renal progenitor cells and formation of the loop of Henle.


Subject(s)
Loop of Henle/embryology , Loop of Henle/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Multipotent Stem Cells/cytology , Organogenesis , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Gene Ontology , Homozygote , Kidney Tubules/metabolism , Loop of Henle/abnormalities , Mice , Multipotent Stem Cells/metabolism , Mutation/genetics , Organogenesis/genetics , Protein Binding/genetics , Transcription Factors/chemistry , Ureter/embryology , Ureter/metabolism
12.
Mol Cancer ; 17(1): 78, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29625565

ABSTRACT

BACKGROUND: SALL1 is a multi-zinc finger transcription factor that regulates organogenesis and stem cell development, but the role of SALL1 in tumor biology and tumorigenesis remains largely unknown. METHODS: We analyzed SALL1 expression levels in human and murine breast cancer cells as well as cancer tissues from different types of breast cancer patients. Using both in vitro co-culture system and in vivo breast tumor models, we investigated how SALL1 expression in breast cancer cells affects tumor cell growth and proliferation, metastasis, and cell fate. Using the gain-of function and loss-of-function strategies, we dissected the molecular mechanism responsible for SALL1 tumor suppressor functions. RESULTS: We demonstrated that SALL1 functions as a tumor suppressor in breast cancer, which is significantly down-regulated in the basal like breast cancer and in estrogen receptor (ER), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER2) triple negative breast cancer patients. SALL1 expression in human and murine breast cancer cells inhibited cancer cell growth and proliferation, metastasis, and promoted cell cycle arrest. Knockdown of SALL1 in breast cancer cells promoted cancer cell growth, proliferation, and colony formation. Our studies revealed that tumor suppression was mediated by recruitment of the Nucleosome Remodeling and Deacetylase (NuRD) complex by SALL1, which promoted cancer cell senescence. We further demonstrated that the mechanism of inhibition of breast cancer cell growth and invasion by SALL1-NuRD depends on the p38 MAPK, ERK1/2, and mTOR signaling pathways. CONCLUSION: Our studies indicate that the developmental control gene SALL1 plays a critical role in tumor suppression by recruiting the NuRD complex and thereby inducing cell senescence in breast cancer cells.


Subject(s)
Down-Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
13.
Adv Exp Med Biol ; 1074: 209-215, 2018.
Article in English | MEDLINE | ID: mdl-29721946

ABSTRACT

Retinal degeneration often accompanies microglial activation and infiltration of monocyte-derived macrophages into the retina, resulting in the coexistence of microglia and monocyte-derived macrophages in the retina. We previously showed that the Sall1 zinc-finger transcriptional factor is expressed specifically in microglia within the retinal phagocyte pool, and analyses of Sall1 knockout mice revealed that microglial morphology changed from a ramified to a more amoeboid appearance in the developing retina. To investigate further whether Sall1 functions autonomously in microglia, we generated Sall1 conditional knockout mice, in which Sall1 was depleted specifically in the Cx3cr1+ microglial compartment of the developing retina. Sall1-deficient microglia exhibited morphological abnormalities on embryonic day 18 that strikingly resembled the phenotype observed in Sall1 knockout mice, demonstrating that Sall1 regulates microglial morphology cell autonomously. Analysis of the postnatal retina revealed that Sall1-deficient microglia extended their processes and their morphology became comparable to that of wild-type microglia on postnatal day 21, indicating that Sall1 is essential for microglial ramification in the developing retina, but not in the postnatal retina.


Subject(s)
Eye Proteins/physiology , Microglia/ultrastructure , Retina/cytology , Transcription Factors/physiology , Animals , CX3C Chemokine Receptor 1/biosynthesis , CX3C Chemokine Receptor 1/genetics , Cell Shape , Cell Surface Extensions/ultrastructure , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/biosynthesis , Retina/embryology , Retina/growth & development , Tamoxifen/pharmacology , Transcription Factors/deficiency
14.
Development ; 141(5): 1047-58, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24550112

ABSTRACT

The formation of the proper number of functional nephrons requires a delicate balance between renal progenitor cell self-renewal and differentiation. The molecular factors that regulate the dramatic expansion of the progenitor cell pool and differentiation of these cells into nephron precursor structures (renal vesicles) are not well understood. Here we show that Sall1, a nuclear transcription factor, is required to maintain the stemness of nephron progenitor cells. Transcriptional profiling of Sall1 mutant cells revealed a striking pattern, marked by the reduction of progenitor genes and amplified expression of renal vesicle differentiation genes. These global changes in gene expression were accompanied by ectopic differentiation at E12.5 and depletion of Six2+Cited1+ cap mesenchyme progenitor cells. These findings highlight a novel role for Sall1 in maintaining the stemness of the progenitor cell pool by restraining their differentiation into renal vesicles.


Subject(s)
Cell Differentiation/physiology , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Female , Immunohistochemistry , In Situ Hybridization , Kidney/cytology , Mice , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
15.
BMC Med Genet ; 18(1): 125, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29110636

ABSTRACT

BACKGROUND: Townes-Brocks syndrome (TBS) is a rare autosomal dominant condition characterized by renal, anal, limb, and auditory abnormalities. TBS diagnosis can be challenging in settings where genetic analysis is not readily available. TBS traits overlap with those of Goldenhar and VACTERL syndromes. CASE PRESENTATION: Here, we present the case of a 5-year-old Brazilian boy born with an anorectal abnormality, limb and external ears malformations, genitourinary anomalies, and a congenital heart defect. Genetic analysis revealed a SALL1 nonsense mutation. The case is discussed in the context of the current literature. CONCLUSIONS: Because of the variability in TBS clinical presentation, genetic analysis is key to the differential diagnosis of TBS relative to phenotypically similar syndromes.


Subject(s)
Abnormalities, Multiple/genetics , Anus, Imperforate/genetics , Codon, Nonsense , Genetic Predisposition to Disease/genetics , Hearing Loss, Sensorineural/genetics , Thumb/abnormalities , Transcription Factors/genetics , Abnormalities, Multiple/diagnosis , Anal Canal/abnormalities , Anus, Imperforate/diagnosis , Brazil , Child, Preschool , Diagnosis, Differential , Esophagus/abnormalities , Genotype , Hearing Loss, Sensorineural/diagnosis , Heart Defects, Congenital/diagnosis , Humans , Kidney/abnormalities , Limb Deformities, Congenital/diagnosis , Male , Phenotype , Spine/abnormalities , Trachea/abnormalities
16.
Pediatr Nephrol ; 32(2): 195-200, 2017 02.
Article in English | MEDLINE | ID: mdl-26868670

ABSTRACT

Studies of kidney regeneration using stem cells have progressed rapidly in recent years. Our group has developed a protocol to induce nephron progenitors from both mouse and human pluripotent stem cells which is based on a revised model of early stage kidney specification. The induced progenitors readily reconstitute three-dimensional nephron structures, including glomeruli and renal tubules, in vitro. We can further generate human induced pluripotent stem cells (iPSCs), in which nephrin-expressing glomerular podocytes are tagged with green fluorescent protein (GFP). The sorted GFP-positive cells retain the podocyte-specific molecular and structural features. Upon transplantation, mouse endothelial cells of the host animals are integrated into the human iPSC-derived glomeruli, and the podocytes show further maturation. Other laboratories have reported different protocols to induce nephron structures from human iPSCs in vitro. These findings will accelerate our understanding of kidney development and diseases in humans.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Kidney Glomerulus/cytology , Kidney Tubules/cytology , Animals , Cell Culture Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Mice
17.
Am J Physiol Renal Physiol ; 309(10): F852-63, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26311113

ABSTRACT

It has been postulated that developmental pathways are reutilized during repair and regeneration after injury, but functional analysis of many genes required for kidney formation has not been performed in the adult organ. Mutations in SALL1 cause Townes-Brocks syndrome (TBS) and nonsyndromic congenital anomalies of the kidney and urinary tract, both of which lead to childhood kidney failure. Sall1 is a transcriptional regulator that is expressed in renal progenitor cells and developing nephrons in the embryo. However, its role in the adult kidney has not been investigated. Using a mouse model of TBS (Sall1TBS), we investigated the role of Sall1 in response to acute kidney injury. Our studies revealed that Sall1 is expressed in terminally differentiated renal epithelia, including the S3 segment of the proximal tubule, in the mature kidney. Sall1TBS mice exhibited significant protection from ischemia-reperfusion injury and aristolochic acid-induced nephrotoxicity. This protection from acute injury is seen despite the presence of slowly progressive chronic kidney disease in Sall1TBS mice. Mice containing null alleles of Sall1 are not protected from acute kidney injury, indicating that expression of a truncated mutant protein from the Sall1TBS allele, while causative of congenital anomalies, protects the adult kidney from injury. Our studies further revealed that basal levels of the preconditioning factor heme oxygenase-1 are elevated in Sall1TBS kidneys, suggesting a mechanism for the relative resistance to injury in this model. Together, these studies establish a functional role for Sall1 in the response of the adult kidney to acute injury.


Subject(s)
Abnormalities, Multiple/metabolism , Acute Kidney Injury/metabolism , Anus, Imperforate/metabolism , Hearing Loss, Sensorineural/metabolism , Mutant Proteins/metabolism , Thumb/abnormalities , Transcription Factors/metabolism , Abnormalities, Multiple/genetics , Acute Kidney Injury/genetics , Animals , Anus, Imperforate/genetics , Disease Models, Animal , Hearing Loss, Sensorineural/genetics , Heme Oxygenase-1/genetics , Mice, Transgenic , Mutation/genetics , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Transcription Factors/genetics
18.
Pediatr Int ; 56(5): e75-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25336016

ABSTRACT

Microdeletion of 16q12 is a rare chromosomal abnormality. We present the cases of two Japanese patients with developmental and renal symptoms of differing clinical severity. Both patients had 16q12 interstitial microdeletions that included the entire SALL1 gene. Patient 1 was a 15-year-old Japanese boy clinically diagnosed with branchio-oto-renal syndrome with mild developmental delay, but with no imperforate anus or polydactyly. Array comparative genome hybridization (aCGH) indicated a 5.2 Mb deletion in 16q12, which included SALL1. Patient 2 was a 13-year-old Japanese boy diagnosed with Townes-Brocks syndrome and severe developmental delay, epilepsy, and renal insufficiency requiring renal replacement therapy. Fluorescence in situ hybridization indicated deletion of the entire SALL1 gene. Subsequent aCGH showed a 6 Mb deletion in 16q12q13, which included SALL1. Precise analysis of the present two cases will give us some clues to elucidate the pathogenic mechanisms of 16q12 microdeletion syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Aberrations , Adolescent , Asian People , Gene Deletion , Humans , Male , Syndrome
19.
Genetics ; 227(1)2024 05 07.
Article in English | MEDLINE | ID: mdl-38386912

ABSTRACT

Vertebrate limbs start to develop as paired protrusions from the lateral plate mesoderm at specific locations of the body with forelimb buds developing anteriorly and hindlimb buds posteriorly. During the initiation process, limb progenitor cells maintain active proliferation to form protrusions and start to express Fgf10, which triggers molecular processes for outgrowth and patterning. Although both processes occur in both types of limbs, forelimbs (Tbx5), and hindlimbs (Isl1) utilize distinct transcriptional systems to trigger their development. Here, we report that Sall1 and Sall4, zinc finger transcription factor genes, regulate hindlimb initiation in mouse embryos. Compared to the 100% frequency loss of hindlimb buds in TCre; Isl1 conditional knockouts, Hoxb6Cre; Isl1 conditional knockout causes a hypomorphic phenotype with only approximately 5% of mutants lacking the hindlimb. Our previous study of SALL4 ChIP-seq showed SALL4 enrichment in an Isl1 enhancer, suggesting that SALL4 acts upstream of Isl1. Removing 1 allele of Sall4 from the hypomorphic Hoxb6Cre; Isl1 mutant background caused loss of hindlimbs, but removing both alleles caused an even higher frequency of loss of hindlimbs, suggesting a genetic interaction between Sall4 and Isl1. Furthermore, TCre-mediated conditional double knockouts of Sall1 and Sall4 displayed a loss of expression of hindlimb progenitor markers (Isl1, Pitx1, Tbx4) and failed to develop hindlimbs, demonstrating functional redundancy between Sall1 and Sall4. Our data provides genetic evidence that Sall1 and Sall4 act as master regulators of hindlimb initiation.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Developmental , Hindlimb , LIM-Homeodomain Proteins , Transcription Factors , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , Mice , Hindlimb/embryology , Hindlimb/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Limb Buds/metabolism , Limb Buds/embryology , Mice, Knockout , Embryo, Mammalian/metabolism , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
20.
Kidney Int Rep ; 9(6): 1810-1816, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38899216

ABSTRACT

Introduction: Townes-Brocks syndrome (TBS), a rare autosomal dominant genetic condition associated with SALL1 (Spalt like Transcription Factor 1), is reported to be present in 1:238,000 individuals in the general population. TBS is characterized by the triad of anorectal malformations, dysplastic ears, with or without hearing impairment, and hand or thumb anomalies. Although kidney involvement is less common in TBS, the disease can progress to kidney failure. Here, we sought to characterize the incidence of SALL1 variants in individuals undergoing broad-based genetic testing with a kidney gene panel and to quantify the presence of (extra)renal features. Methods: A retrospective analysis of the genetic data from a 385-gene panel identified cases with a pathogenic (P) or likely pathogenic (LP) variant in SALL1. Data including age, features, and disease progression were collected. Results: Of 35,044 samples, P or LP variants in SALL1 were identified in 22, yielding a prevalence of 1:1592 among patients tested for monogenic kidney disease, and 1:342 among cases identified with a monogenic kidney disease. Among this cohort, the median patient age was 23 years (range: 3 months-62 years) with chronic kidney disease (CKD) reported in 91% (20/22) of cases. Reported kidney features included renal agenesis/hypoplasia (7/22; 32%), focal segmental glomerulosclerosis (4/22; 18%), and kidney cysts (3/22; 14%). Confirmed extrarenal features included hearing loss and/or ear features (7/22; 32%), anorectal malformations (6/22; 27%) and hand or thumb abnormalities (4/22; 18%). Three patients (3/22; 14%) had both a priori TBS diagnoses and the traditional "triad." Conclusion: Traditionally, a molecular diagnosis was ascertained primarily in individuals presenting with cardinal features of TBS; therefore, individuals with mild or atypical presentations were often overlooked clinically. Our findings reveal that SALL1 P/LP variants could be a consequential contributor to monogenic kidney disease.

SELECTION OF CITATIONS
SEARCH DETAIL