Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters











Publication year range
1.
Ital J Pediatr ; 50(1): 121, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38915054

ABSTRACT

BACKGROUND: Townes-Brocks syndrome (TBS) is a rare genetic disorder characterized by imperforate anus, dysplastic ears, thumb malformations, and other abnormalities. Previous studies have revealed that mutations in the SALL1 gene can disrupt normal development, resulting in the characteristic features of Townes-Brocks syndrome. Spalt-like transcription factors (SALLs) are highly conserved proteins that play important roles in various cellular processes, including embryonic development, cell differentiation, and cell survival. Over 400 different variants or mutations have been reported in the SALL1 gene in individuals with TBS. Most of these variants lead to the formation of premature termination codons (PTCs), also known as nonsense mutations. The majority of these PTCs occur in a specific region of the SALL1 gene called the "hotspot region", which is particularly susceptible to mutation. METHODS: In this study, we conducted whole-exome sequencing on a three-generation Chinese family with anorectal malformations. RESULTS: We identified a novel heterozygous mutation (chr16:51175376:c.757 C > T p.Gln253*) in the SALL1 gene. Molecular analysis revealed a heterozygous C to T transition at nucleotide position 757 in exon 2 of the SALL1 (NM_002968) gene. This mutation is predicted to result in the substitution of the Gln253 codon with a premature stop codon (p.Gln253*). The glutamine-rich domain forms a long alpha helix, enabling the mutant protein to interact with the wild-type SALL1 protein. This interaction may result in steric hindrance effects on the wild-type SALL1 protein. CONCLUSIONS: Our findings have expanded the mutation database of the SALL1 gene, which is significant for genetic counseling and clinical surveillance in the affected family. Furthermore, our study enhances the understanding of Townes-Brocks syndrome and has the potential to improve its diagnosis and treatment.


Subject(s)
Abnormalities, Multiple , Anus, Imperforate , Pedigree , Transcription Factors , Female , Humans , Male , Abnormalities, Multiple/genetics , Anorectal Malformations/genetics , Anus, Imperforate/genetics , Asian People/genetics , China , East Asian People , Hearing Loss, Sensorineural , Mutation , Rare Diseases/genetics , Thumb/abnormalities , Transcription Factors/genetics
2.
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.

3.
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
4.
J Pediatr Genet ; 13(2): 139-143, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38721582

ABSTRACT

Townes-Brocks syndrome (TBS) is a rare syndrome characterized by triad of anal, ear, and thumb anomalies. Further malformations/anomalies include congenital heart diseases, foot malformations, sensorineural and/or conductive hearing impairment, genitourinary malformations, and anomalies of eye and nervous system. Definitive diagnosis for TBS is confirmed by molecular analysis for mutations in the SALL1 gene. Only one known case of TBS with absent pulmonary valve syndrome (APVS) has been previously described to our knowledge. Here, we report a newborn diagnosed with TBS with APVS and tetralogy of Fallot (TOF) who was found to carry the most common pathogenic SALL1 gene mutation c.826C > T (p.R276X), with its surgical repair and postoperative follow-up. To our knowledge, this is the first genotyped case of TBS from Turkey to date. TBS should be suspected in the presence of ear, anal, and thumb malformations in a neonate. If a patient with TBS and TOF-APVS needs preoperative ventilation within the first months of life, this implies prolonged postoperative intubation and increased risk of mortality.

5.
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
6.
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
7.
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
8.
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
9.
Pathol Res Pract ; 250: 154827, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37741137

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most usual malignant tumors, and its incidence continues to rise. Our purpose was to explore the function and potential regulatory mechanisms of SALL1, a differentially methylated gene in CRC, in vivo and in vitro. METHODS: Firstly, methylation differential gene SALL1 in CRC was screened and validated. SALL1 overexpression plasmids or SALL1 siRNAs were transfected in HT-29 and SW480 cells. Moreover, 10 µM T-5224 was added in SALL1-overexpressed CRC cells. CCK-8, flow cytometry and transwell assays were utilized to assess cell proliferation, cycle, migration, and invasion, respectively. Then CRC organoids were cultured. Next, HT-29 and SW480 cells transfected with SALL1 overexpression lentivirus were analyzed by transcriptome sequencing. Finally, in vivo tumorigenesis was used to analyze the effect of SALL1 overexpression on subcutaneous tumorigenesis in nude mice. RESULTS: The methylation level of CpG island in SALL1 promoter was increased in CRC tissues and could distinguish tumor tissues. Overexpression of SALL1 accelerated proliferation, migration and invasion of HT-29 and SW480 cells, and silencing of SALL1 attenuated proliferation, migration and invasion of HT-29 and SW480 cells. Through analysis and validation, we found that overexpression of SALL1 also could upregulate p-p65 and p-JUN expressions. Besides, c-Fos/activator protein (AP)- 1 inhibitor (T-5224) could reverse the induction of CRC progression by SALL1 overexpression. In vivo, we also proved that overexpression of SALL1 significantly increased tumor volume, tumor weight, and p-JUN expression. CONCLUSIONS: SALL1 could promote the proliferation, migration, and invasion of CRC cells and activate phosphorylation of p65 and JUN.

10.
Orphanet J Rare Dis ; 18(1): 250, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37644569

ABSTRACT

BACKGROUND: Townes-Brocks syndrome is a rare autosomal dominant genetic syndrome caused by mutations in SALL1. The clinical features of Townes-Brocks syndrome are highly heterogonous. Identification of new SALL1 mutations and study of the relation between SALL1 mutations and clinical features can facilitate diagnosis of Townes-Brocks syndrome. METHODS: We collected clinical data and blood samples of the two patients and their family members for whole-exome sequencing and Sanger sequencing. Prediction analysis of the SALL1variation protein structure was achieved using Alphafold. The clinical materials and gene sequencing results were analyzed. The clinical materials and gene sequencing results were analyzed. The related literature of Townes-Brocks syndrome were searched and the genotype-renal phenotype analysis was performed combined with this two cases. RESULTS: Based on the clinical features and gene sequencing results, the two patients were diagnosed as Townes-Brocks syndrome. Two novel SALL1 mutations (c.878-887del and c.1240G > T) were identified, both of which were pathogenic mutations. The correlation between genotypes and renal phenotypes in Townes-Brocks syndrome patients caused by SALL1 mutation were summarized. CONCLUSION: This study identified two novel mutations and provided new insights into the correlation of genotypes and renal phenotypes of Townes-Brocks syndrome.


Subject(s)
Arthrogryposis , East Asian People , Humans , Asian People , Mutation/genetics
11.
World J Clin Cases ; 11(23): 5567-5572, 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37637690

ABSTRACT

BACKGROUND: Townes-Brocks syndrome (TBS) is a rare autosomal dominant syndrome that is characterized by a triad of imperforate anus, dysplastic ears, and thumb malformations. Heterozygous variants of SALL1 are responsible for this syndrome. Renal structural abnormalities and functional impairments are often reported in TBS patients. CASE SUMMARY: We report a case of TBS in a Chinese family. The index patients showed obvious renal atrophy and renal failure. TBS was suggested after a physical examination and pedigree analysis. Whole exome sequencing revealed a heterozygous variant of SALL1. The variant (NM_001127892 c.1289_c.1290 insC) led to a read-frame shift of the encoded protein, which was confirmed by Sanger sequencing. The variant cosegregated with the phenotype among affected members. CONCLUSION: A novel variant in SALL1 gene may be the molecular pathogenic basis of this disorder.

12.
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
13.
Genes (Basel) ; 14(2)2023 01 19.
Article in English | MEDLINE | ID: mdl-36833185

ABSTRACT

SALL1 heterozygous pathogenic variants cause Townes-Brocks syndrome (TBS), a condition with variable clinical presentation. The main features are a stenotic or imperforate anus, dysplastic ears, and thumb malformations, and other common concerns are hearing impairments, foot malformations, and renal and heart defects. Most of the pathogenic SALL1 variants are nonsense and frameshift, likely escaping nonsense-mediated mRNA decay and causing disease via a dominant-negative mechanism. Haploinsufficiency may result in mild phenotypes, but only four families with distinct SALL1 deletions have been reported to date, with a few more being of larger size and also affecting neighboring genes. We report on a family with autosomal dominant hearing impairment and mild anal and skeletal anomalies, in whom a novel 350 kb SALL1 deletion, spanning exon 1 and the upstream region, was identified by array comparative genomic hybridization. We review the clinical findings of known individuals with SALL1 deletions and point out that the overall phenotype is milder, especially when compared with individuals who carry the recurrent p.Arg276Ter mutation, but with a possible higher risk of developmental delay. Chromosomal microarray analysis is still a valuable tool in the identification of atypical/mild TBS cases, which are likely underestimated.


Subject(s)
Anus, Imperforate , Syndrome , Transcription Factors , Humans , Anus, Imperforate/genetics , Comparative Genomic Hybridization , Haploinsufficiency , Microarray Analysis , Phenotype , Thumb/abnormalities , Transcription Factors/genetics
14.
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
15.
Front Pediatr ; 10: 988374, 2022.
Article in English | MEDLINE | ID: mdl-36238604

ABSTRACT

Congenital lower urinary tract obstructions (LUTO) are most often caused by posterior urethral valves (PUV), a male limited anatomical obstruction of the urethra affecting 1 in 4,000 male live births. Little is known about the genetic background of PUV. Here, we report the largest genome-wide association study (GWAS) for PUV in 4 cohorts of patients and controls. The final meta-analysis included 756 patients and 4,823 ethnicity matched controls and comprised 5,754,208 variants that were genotyped or imputed and passed quality control in all 4 cohorts. No genome-wide significant locus was identified, but 33 variants showed suggestive significance (P < 1 × 10-5). When considering only loci with multiple variants residing within < 10 kB of each other showing suggestive significance and with the same effect direction in all 4 cohorts, 3 loci comprising a total of 9 variants remained. These loci resided on chromosomes 13, 16, and 20. The present GWAS and meta-analysis is the largest genetic study on PUV performed to date. The fact that no genome-wide significant locus was identified, can be explained by lack of power or may indicate that common variants do not play a major role in the etiology of PUV. Nevertheless, future studies are warranted to replicate and validate the 3 loci that yielded suggestive associations.

16.
Eur J Med Genet ; 65(12): 104642, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36252910

ABSTRACT

This report describes a novel truncating c.709C > T p.(Gln237*) SALL1 variant in two siblings exhibiting sagittal craniosynostosis as a unique feature of Townes-Brocks syndrome (TBS, OMIM #107480). TBS is a rare autosomal dominant syndrome with variable phenotypes, including anorectal, renal, limb, and ear abnormalities, which results from heterozygous variants in the SALL1 gene, predominantly located in the 802 bp "hot spot region" within exon 2. Recent studies have suggested that aberrations in primary cilia and sonic hedgehog signalling contribute to the TBS phenotypes. The presence of the novel c.709C > T p.(Gln237*) SALL1 variant was confirmed in both the siblings and their father, whereas no mutations currently associated with craniosynostosis were detected. We hypothesise that the truncating c.709C > T p.(Gln237*) SALL1 variant, which occurs outside the "hot spot region" and inside the glutamine-rich domain coding region, could interfere with ciliary signalling and mechanotransduction, contributing to premature fusion of calvarial sutures. This report broadens the genetic and phenotypic spectrum of TBS and provides the first clinical evidence of craniosynostosis as a novel feature of the syndrome.


Subject(s)
Craniosynostoses , Siblings , Humans , Craniosynostoses/genetics , Hedgehog Proteins , Mechanotransduction, Cellular , Syndrome , Transcription Factors/genetics
17.
World J Clin Cases ; 10(20): 7068-7075, 2022 Jul 16.
Article in English | MEDLINE | ID: mdl-36051141

ABSTRACT

BACKGROUND: Approximately 10% of adults and nearly all children who receive renal replacement therapy have inherited risk factors or are related to genetic factors. In the past, due to the limitations of detection technology and the nonspecific manifestations of uraemia, the etiological diagnosis is unclear. In addition to common monogenic diseases and complex disorders, advanced testing techniques have led to the recognition of more hereditary renal diseases. Here, we report a four-generation Chinese family in which four individuals had a novel SALL1 mutation and presented with uraemia or abnormal urine tests. CASE SUMMARY: A 32-year-old man presented with end-stage renal disease with a 4-year history of dialysis. His father and paternal aunt both had a history of unexplained renal failure with haemodialysis, and his 10-year-old daughter presented with proteinuria. The patient had multiple congenital abnormalities, including bilateral overlapping toes, unilateral dysplastic external ears, and sensorineural hearing loss. His family members also presented with similar defects. Genetic testing revealed that the proband carried a novel heterozygous shift mutation in SALL1_exon 2 (c.3437delG), and Sanger sequencing confirmed the same mutation in all affected family members. CONCLUSION: We report a novel SALL1 exon 2 (c.3437delG) mutation and clinical syndrome with kidney disease, bilateral overlapping toes, unilateral dysplastic external ears, and sensorineural hearing loss in a four-generation Chinese family.

18.
Glob Med Genet ; 9(2): 124-128, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35707773

ABSTRACT

Sterile α motif domain-containing protein 9 (SAMD9) is a regulatory protein centrally involved in cell proliferation and apoptosis. Mapped to 7p21.2, variants in SAMD9 have been reported in <50 pediatric cases worldwide, typically with early lethality. Germline gain-of-function SAMD9 variants are associated with MIRAGE syndrome (myelodysplasia, infection, restricted growth, adrenal hypoplasia, genital anomalies, and enteropathy). Spalt like transcription factor 1 (SALL1) is a zinc finger transcriptional repressor located at 16q12.1 where only two transcript variants in SALL1 are known. RUNX2 (6p21.1) encodes a nuclear protein with a Runt DNA-binding domain critical for osteoblastic differentiation, skeletal morphogenesis, and serves as a scaffold for nucleic acids and regulatory factors involved in skeletal gene expression. RUNX2 and SALL1 are thus both "master regulators" of tissue organization and embryo development. Here, we describe exome sequencing and copy number variants in two previously unknown mutations-R824Q in SAMD9, and Q253H in SALL1. A multiexon 3' terminal duplication of RUNX2 not previously encountered is also reported. This is the first known phenotype assessment for an intersection of all three variants in a healthy 46,XX adult. Focusing on developmental progress, ultrastructural renal anatomy, and selected reproductive aspects, we describe this unique genotype diagnosed incidentally during coronavirus disease 2019 (COVID-19) illness. Individually, disruption in SAMD9, RUNX2, or SALL1 would be expected to give a bleak prognosis. However, this variant convergence appears to dampen severe pathology perhaps by cross-gene silencing of effects normally deleterious when such changes occur alone.

19.
Tissue Cell ; 76: 101811, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35567907

ABSTRACT

BACKGROUND: This study compared the effect of ultrasound microbubble-mediated miR-503-5p downregulation with that of pure liposome-mediated miR-503-5p downregulation on colorectal cancer (CRC) progression and explored the downstream mechanism. METHODS: Bioinformatics tools were utilized to predict miR-503-5p-targeted genes and CRC progression-associated genes. MiR-503-5p and sal-like 1 (SALL1) expressions in CRC cells and tissues were analyzed by qRT-PCR and/or bioinformatics tools; their correlations with overall survival and clinicopathological features of CRC patients were presented, and their interaction was validated by dual-luciferase reporter assay. CRC cells received ultrasound microbubble-mediated miR-503-5p downregulation and/or liposome-mediated miR-503-5p downregulation or SALL1 silencing. Cell phenotype changes were evaluated by flow cytometry, as well as MTT, Wound healing, Transwell and tube formation assays. E-cadherin, N-cadherin, Vimentin, B-cell lymphoma (Bcl)- 2, Cleaved caspase-3, and SALL1 expressions in cells were analyzed by Western blot. RESULTS: Upregulated miR-503-5p in CRC tissues and cells was detected, associated with poorer cell differentiation, easier lymph node metastasis and higher TNM stages, and related to poorer prognoses of CRC patients. Ultrasound microbubble-mediated miR-503-5p downregulation relative to pure liposome-mediated miR-503-5p downregulation better decreased viability, inhibited migration, invasion and tube formation, enhanced apoptosis, upregulated SALL1, E-cadherin and Cleaved caspase-3, and downregulated miR-503-5p, N-cadherin, Vimentin and Bcl-2 in CRC cells. SALL1 was targeted by miR-503-5p, low-expressed in CRC tissues and cells and positively related to CRC patients' survival. Silencing SALL1 exerted the opposite effects, which reversed the effects of ultrasound microbubble-mediated miR-503-5p downregulation and vice versa. CONCLUSION: Ultrasound microbubble-mediated miR-503-5p downregulation suppressed in vitro CRC progression via promoting SALL1 expression.


Subject(s)
MicroRNAs , Cadherins/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Humans , Liposomes , MicroRNAs/metabolism , Microbubbles , Vimentin/metabolism
20.
Neurochem Int ; 157: 105347, 2022 07.
Article in English | MEDLINE | ID: mdl-35490895

ABSTRACT

BACKGROUND: Abnormal expression of lncRNA is involved in a diversity of diseases and plays a vital role in targeted therapy. However, few studies have been conducted on lncRNA PART1 in glioma. We aimed to investigate the function and the potential regulatory mechanism of lncRNA PART1/miR-374b/SALL1 axis in glioma. METHODS: qRT-PCR and western blotting detected genes and proteins expression. Dual-luciferase reporter assay was performed to examine the binding relationship of lncRNA PART1, miR-374b, and SALL1. MTT assay and clone formation assay were performed to detect the cell viability and proliferation. Transwell assay detected glioma cell migration. In vivo tumor development experiments detected changes in tumor size, volume, and weight of the tumor after overexpression of lncRNA PART1. Immunohistochemistry was used to detect ki-67, E-cadherin, and N-cadherin expression. RESULTS: The expression of lncRNA PART1 and SALL1 were down-regulated and miR-374b was up-regulated in different glioma cell lines. Overexpression of lncRNA PART1 inhibited glioma cell proliferation, migration, and epithelial mesenchymal transition (EMT). LncRNA PART1 targeted miR-374b to promote SALL1 expression. The knockdown of miR-374b inhibited glioma cell proliferation and migration and EMT by SALL1. What's more, overexpression of miR-374b or knockdown of SALL1 reversed the inhibitory effect of lncRNA PART1 on the proliferation, migration, and EMT of glioma cells. Furthermore, overexpression of lncRNA PART1 inhibited glioma growth in vivo. CONCLUSION: LncRNA PART1 inhibited glioma proliferation and migration via miR-374b/SALL1 axis. These results might provide new insights for comprehending the complex lncRNA-miRNA network in gliomas.


Subject(s)
Glioma , MicroRNAs , RNA, Long Noncoding , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL