Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.033
Filter
1.
Taiwan J Obstet Gynecol ; 63(4): 561-564, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39004488

ABSTRACT

OBJECTIVE: We present prenatal diagnosis of familial 3p26.3p25.3 deletion in a pregnancy associated with a favorable fetal outcome and asymptomatic carrier parent and family members in three generations. CASE REPORT: A 35-year-old, gravida 2, para 1, woman underwent amniocentesis at 17 weeks of gestation because of advanced maternal age and the carrier of distal 3p deletion. She was phenotypically normal, and there was no family history of congenital anomalies. Amniocentesis revealed a karyotype of 46,XY,del(3)(p26.1). Repeat amniocentesis at 21 weeks of gestation revealed a karyotype of 46,XY,del(3)(p25.3). Simultaneous array comparative genomic hybridization (aCGH) analysis on the DNA extracted from uncultured amniocytes showed the result of arr 3p26.3p25.3 (117,735-8,709,972) × 1.0 [GRCh37 (hg19)] with an 8.59-Mb deletion of 3p26.3p25.3 encompassing 14 OMIM genes of CHL1, CNTN6, CNTN4, IL5RA, TRNT1, CRBN, SETMAR, SUMF1, ITPR1, BHLHE40, ARL8B, GRM7, LMCD1 and SSUH2. Cytogenetic analysis of parental bloods revealed a karyotype of 46,XX,del (3) (p25.3) in the mother and 46,XY in the father. The woman's 69-year-old mother and her 2-year-old elder son carried the same aberrant chromosome of 3p25.3→p26.3 deletion by conventional cytogenetic analysis but manifested no phenotypic abnormality. aCGH analysis of the peripheral bloods showed that the woman's mother and her elder son had the same 8.59-Mb deletion of 3p26.3p25.3. The woman was advised to continue the pregnancy. At 39 weeks of gestation, a 3040-g healthy male baby was delivered. When follow-up at age 2½ years, the neonate was normal in development and showed no apparent phenotypic abnormality. CONCLUSION: Distal 3p deletion of 3p26.3p25.3 involving the OMIM genes from CHL1 to SSUH2 can be associated with no apparent phenotypic abnormality.


Subject(s)
Amniocentesis , Chromosome Deletion , Chromosomes, Human, Pair 3 , Comparative Genomic Hybridization , Pedigree , Humans , Female , Pregnancy , Chromosomes, Human, Pair 3/genetics , Adult , Male , Heterozygote , Infant, Newborn
2.
Nagoya J Med Sci ; 86(2): 216-222, 2024 May.
Article in English | MEDLINE | ID: mdl-38962417

ABSTRACT

Psychiatric disorders are highly inheritable, and most psychiatric disorders exhibit genetic overlap. Recent studies associated the 3q29 recurrent deletion with schizophrenia (SCZ) and autism spectrum disorder (ASD). In this study, we investigated the association of genes in the 3q29 region with SCZ and ASD. TM4SF19 and PAK2 were chosen as candidate genes for this study based on evidence from previous research. We sequenced TM4SF19 and PAK2 in 437 SCZ cases, 187 ASD cases and 524 controls in the Japanese population. Through targeted sequencing, we identified 6 missense variants among the cases (ASD & SCZ), 3 missense variants among controls, and 1 variant common to both cases and controls; however, no loss-of-function variants were identified. Fisher's exact test showed a significant association of variants in TM4SF19 among cases (p=0.0160). These results suggest TM4SF19 variants affect the etiology of SCZ and ASD in the Japanese population. Further research examining 3q29 region genes and their association with SCZ and ASD is thus needed.


Subject(s)
Asian People , Autism Spectrum Disorder , Genetic Predisposition to Disease , Schizophrenia , Humans , Autism Spectrum Disorder/genetics , Schizophrenia/genetics , Female , Male , Japan , Asian People/genetics , Genetic Predisposition to Disease/genetics , p21-Activated Kinases/genetics , Chromosomes, Human, Pair 3/genetics , Adult , Mutation, Missense/genetics , Case-Control Studies , Genetic Association Studies , East Asian People
3.
Sci Rep ; 14(1): 15058, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38956433

ABSTRACT

Since the COVID-19 pandemic, the diversity of clinical manifestations in patients has been a tremendous challenge. It seems that genetic variations, as one of the players, contribute to the variety of symptoms. Genome-wide association studies have demonstrated the influence of certain genomic regions on the disease prognosis. Particularly, a haplotype at 3p21.31 locus, inherited from Neanderthals, showed an association with COVID-19 severity. Despite several studies regarding this haplotype, some key variants are not sufficiently addressed. In the present study, we investigated the association of rs17713054 at 3p21.31 with COVID-19 severity. We analyzed the genotype of 251 Iranian COVID-19 patients (151 patients with asymptomatic to mild form as control and 100 patients with severe to critical symptoms without any comorbidities as case group) using the ARMS-PCR method. Results demonstrated that the A allele confers an almost twofold increased risk for COVID-19 severity (P value = 0.008). The AA genotype also raises the risk by more than 11 times following the recessive model (P value = 0.013). In conclusion, the A allele in rs17713054 was a risk allele in Iranian patients and was independently associated with COVID-19 severity. More studies are beneficial to confirm these findings in other populations and to develop strategies for risk assessment, prevention, and personalized medicine.


Subject(s)
COVID-19 , Genetic Predisposition to Disease , Neanderthals , Polymorphism, Single Nucleotide , SARS-CoV-2 , Severity of Illness Index , Humans , COVID-19/genetics , COVID-19/virology , COVID-19/epidemiology , Iran/epidemiology , Neanderthals/genetics , Male , Female , Middle Aged , Animals , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Adult , Haplotypes , Chromosomes, Human, Pair 3/genetics , Alleles , Genome-Wide Association Study , Genotype , Aged
4.
Croat Med J ; 65(3): 209-219, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38868967

ABSTRACT

AIM: To precisely identify and analyze alpha-satellite higher-order repeats (HORs) in T2T-CHM13 assembly of human chromosome 3. METHODS: From the recently sequenced complete T2T-CHM13 assembly of human chromosome 3, the precise alpha satellite HOR structure was computed by using the novel high-precision GRM2023 algorithm with global repeat map (GRM) and monomer distance (MD) diagrams. RESULTS: The major alpha satellite HOR array in chromosome 3 revealed a novel cascading HOR, housing 17mer HOR copies with subfragments of periods 15 and 2. Within each row in the cascading HOR, the monomers were of different types, but different rows within the same cascading 17mer HOR contained more than one monomer of the same type. Each canonical 17mer HOR copy comprised 17 monomers belonging to 16 different monomer types. Another pronounced 10mer HOR array was of the regular Willard's type. CONCLUSION: Our findings emphasize the complexity within the chromosome 3 centromere as well as deviations from expected highly regular patterns.


Subject(s)
Chromosomes, Human, Pair 3 , DNA, Satellite , Humans , DNA, Satellite/genetics , Chromosomes, Human, Pair 3/genetics , Centromere/genetics , Algorithms , Repetitive Sequences, Nucleic Acid/genetics
5.
Nat Commun ; 15(1): 4739, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834613

ABSTRACT

The overexpression of the ecotropic viral integration site-1 gene (EVI1/MECOM) marks the most lethal acute myeloid leukemia (AML) subgroup carrying chromosome 3q26 abnormalities. By taking advantage of the intersectionality of high-throughput cell-based and gene expression screens selective and pan-histone deacetylase inhibitors (HDACis) emerge as potent repressors of EVI1. To understand the mechanism driving on-target anti-leukemia activity of this compound class, here we dissect the expression dynamics of the bone marrow leukemia cells of patients treated with HDACi and reconstitute the EVI1 chromatin-associated co-transcriptional complex merging on the role of proliferation-associated 2G4 (PA2G4) protein. PA2G4 overexpression rescues AML cells from the inhibitory effects of HDACis, while genetic and small molecule inhibition of PA2G4 abrogates EVI1 in 3q26 AML cells, including in patient-derived leukemia xenografts. This study positions PA2G4 at the crosstalk of the EVI1 leukemogenic signal for developing new therapeutics and urges the use of HDACis-based combination therapies in patients with 3q26 AML.


Subject(s)
Chromosomes, Human, Pair 3 , Histone Deacetylase Inhibitors , Leukemia, Myeloid, Acute , MDS1 and EVI1 Complex Locus Protein , Proteogenomics , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chromosomes, Human, Pair 3/genetics , Gene Expression Regulation, Leukemic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , MDS1 and EVI1 Complex Locus Protein/metabolism , MDS1 and EVI1 Complex Locus Protein/genetics , Proteogenomics/methods , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Xenograft Model Antitumor Assays
6.
Invest Ophthalmol Vis Sci ; 65(6): 7, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38833258

ABSTRACT

Purpose: The purpose of this study was to analyze the extent of DNA breaks in primary uveal melanoma (UM) with regard to radiotherapy dose delivery (single-dose versus fractionated) and monosomy 3 status. Methods: A total of 54 patients with UM were included. Stereotactic radiotherapy (SRT) was performed in 23 patients, with 8 undergoing single-dose SRT (sdSRT) treatment and 15 receiving fractionated SRT (fSRT). DNA breaks in the enucleated or endoresected tumors were visualized by a TUNEL assay and quantified by measuring the TUNEL-positive area. Protein expression was analyzed by immunohistochemistry. Co-detection of chromosome 3 with proteins was performed by immuno-fluorescent in situ hybridization. Results: The amount of DNA breaks in the total irradiated group was increased by 2.7-fold (P < 0.001) compared to non-irradiated tissue. Tumors treated with fSRT were affected more severely, showing 2.1-fold more DNA damage (P = 0.007) compared to the cases after single (high) dose irradiation (sdSRT). Monosomy 3 tumors showed less DNA breaks compared to disomy 3 samples (P = 0.004). The presence of metastases after radiotherapy correlated with monosomy 3 and less DNA breaks compared to patients with non-metastatic cancer in the combined group with fSRT and sdSRT (P < 0.05). Conclusions: Fractionated irradiation led to more DNA damage than single-dose treatment in primary UM. As tumors with monosomy 3 showed less DNA breaks than those with disomy 3, this may indicate that they are less radiosensitive, which may influence the efficacy of irradiation.


Subject(s)
Chromosomes, Human, Pair 3 , DNA Damage , Melanoma , Uveal Neoplasms , Humans , Uveal Neoplasms/radiotherapy , Uveal Neoplasms/genetics , Melanoma/radiotherapy , Melanoma/genetics , Female , Chromosomes, Human, Pair 3/genetics , Male , Middle Aged , Aged , Adult , Aged, 80 and over , In Situ Hybridization, Fluorescence , In Situ Nick-End Labeling , Radiotherapy Dosage , Immunohistochemistry , Radiosurgery/adverse effects , Radiosurgery/methods , Dose-Response Relationship, Radiation
7.
Int J Hematol ; 120(2): 203-211, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38748089

ABSTRACT

STUDY AIMS: To evaluate the outcomes of patients with 3q26.2/MECOM-rearranged chronic myeloid leukemia (CML). METHODS: We reviewed consecutive adult patients with 3q26.2/MECOM-rearranged CML between January 1, 1998 and February 16, 2023. Rearrangements of 3q26.2/MECOM were confirmed by conventional cytogenetics, and fluorescence in situ hybridization starting in 2015. RESULTS: We identified 55 patients with MECOM-rearranged CML, including 23 in chronic phase (CP) or accelerated phase (AP) and 32 in blast phase (BP). Nine patients (16%) achieved a major cytogenetic response (MCyR) or deeper. At a median follow-up of 89 months, median survival was 14 months. The 5-year survival rate was 19% overall, 23% in CML-CP/AP, and 15% in CML-BP. In the 6-month landmark analysis, the 5-year survival rate was 41% for allogeneic stem cell transplantation (allo-SCT) recipients versus 17% for non-recipients (P = 0.050). Multivariate analysis showed that the percentage of marrow blasts and achievement of MCyR or deeper could predict survival. CONCLUSION: Outcomes of 3q26.2/MECOM-rearranged CML are poor despite the availability of multiple BCR::ABL1 tyrosine kinase inhibitors (TKIs). Third-generation TKIs in combination with novel agents and possible allo-SCT could be considered given the poor outcomes and resistance to second-generation TKIs.


Subject(s)
Chromosomes, Human, Pair 3 , Gene Rearrangement , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Adult , Middle Aged , Male , Female , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Chromosomes, Human, Pair 3/genetics , Aged , Survival Rate , Young Adult , Treatment Outcome , Retrospective Studies , Adolescent , Follow-Up Studies
8.
Article in English | MEDLINE | ID: mdl-38765507

ABSTRACT

Endometriosis is a complex disease that affects 10-15% of women of reproductive age. Familial studies show that relatives of affected patients have a higher risk of developing the disease, implicating a genetic role for this disorder. Little is known about the impact of germline genomic copy number variant (CNV) polymorphisms on the heredity of the disease. In this study, we describe a rare CNV identified in two sisters with familial endometriosis, which contain genes that may increase the susceptibility and progression of this disease. We investigated the presence of CNVs from the endometrium and blood of the sisters with endometriosis and normal endometrium of five women as controls without the disease using array-CGH through the Agilent 2x400K platform. We excluded common CNVs that were present in the database of genomic variation. We identified, in both sisters, a rare CNV gain affecting 113kb at band 3q12.2 involving two candidate genes: ADGRG7 and TFG. The CNV gain was validated by qPCR. ADGRG7 is located at 3q12.2 and encodes a G protein-coupled receptor influencing the NF-kappaß pathway. TFG participates in chromosomal translocations associated with hematologic tumor and soft tissue sarcomas, and is also involved in the NF-kappa B pathway. The CNV gain in this family provides a new candidate genetic marker for future familial endometriosis studies. Additional longitudinal studies of affected families must confirm any associations between this rare CNV gain and genes involved in the NF-kappaß pathway in predisposition to endometriosis.


Subject(s)
DNA Copy Number Variations , Endometriosis , Humans , Endometriosis/genetics , Female , Adult , Chromosomes, Human, Pair 3/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(5): 617-621, 2024 May 10.
Article in Chinese | MEDLINE | ID: mdl-38684312

ABSTRACT

OBJECTIVE: To explore the characteristics of a fetus with chromosome 1p36 deletion syndrome and 3p26.3p25.2 duplication. METHODS: A pregnant woman who had attended the Genetic Counseling Clinic of Linyi People's Hospital on February 22, 2022 and her fetus were selected as the study subjects. Clinical data were collected. Chromosomal karyotyping, fluorescence in situ hybridization (FISH) and chromosomal microarray analysis (CMA) were carried out for the prenatal diagnosis. RESULTS: Ultrasonography at 24th gestational week revealed that the fetus had ventricular septal defect, single umbilical artery, and slight widening of left lateral ventricle (12 mm). The woman was found to have a karyotype of 46,XX,t(1;3)(p36.22;p25.2), and the result of FISH was t(1;3)(3pter+,1qter+;1pter+,3qter+). The fetus was found to have a karyotype of 46,X?,add(1)(p36), and CMA confirmed that it has a 9.0 Mb deletion at 1p36.33p36.22 and a 12.6 Mb duplication at 3p26.3p25.2. Combining the maternal karyotype, the molecular karyotype of the fetus was determined as 46,X?,der(1)t(1;3)(p36.22;p25.2)mat.arr[hg19]1p36.33p36.22(849467_9882666)×1, 3p26.3p25.2(61892_12699607)×3, with the former known to be associated with 1p36 deletion syndrome. CONCLUSION: The fetus was diagnosed with 1p36 deletion syndrome, and its 1p36.33p36.22 deletion and 3p26.3p25.2 duplication had both derived from the balanced translocation carried by its mother.


Subject(s)
Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 3 , Karyotyping , Prenatal Diagnosis , Humans , Female , Chromosomes, Human, Pair 1/genetics , Pregnancy , Chromosomes, Human, Pair 3/genetics , Adult , Trisomy/genetics , Trisomy/diagnosis , Chromosome Disorders/genetics , Chromosome Disorders/embryology , Chromosome Disorders/diagnosis , In Situ Hybridization, Fluorescence , Fetus/abnormalities
10.
Am J Med Genet A ; 194(8): e63621, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38567931

ABSTRACT

GATA2 and ZNF148 have both been mapped to chromosome 3q. Pathogenic variants in GATA2 have been associated with immunodeficiency and high risk for myelodysplasia, acute myeloid leukemia, and chronic myelomonocytic leukemia. Gain-of-function variants in ZNF148 have previously been suggested as a mechanism for agenesis of the corpus callosum (ACC). Here, we report a novel 10.4 Mb interstitial deletion on 3q12.33q22.1 including GATA2 and ZNF148 in a child with developmental delay, agenesis of the corpus callosum, and vertebral segmentation defects. With this diagnosis, we were able to suggest preemptive referrals to hematology/oncology and allergy/immunology for close monitoring of early myelodysplasia. We also propose a possible link between ZNF148 loss of function variants and ACC.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 3 , GATA2 Transcription Factor , Transcription Factors , Humans , GATA2 Transcription Factor/genetics , Chromosomes, Human, Pair 3/genetics , Transcription Factors/genetics , Male , DNA-Binding Proteins/genetics , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/pathology , Female , Developmental Disabilities/genetics , Developmental Disabilities/pathology
11.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673877

ABSTRACT

Monosomy 3 in uveal melanoma (UM) increases the risk of lethal metastases, mainly in the liver, which serves as the major site for the storage of excessive glucose and the metabolization of the dietary flavonoid quercetin. Although primary UMs with monosomy 3 exhibit a higher potential for basal glucose uptake, it remains unknown as to whether glycolytic capacity is altered in such tumors. Herein, we initially analyzed the expression of n = 151 genes involved in glycolysis and its interconnected branch, the "pentose phosphate pathway (PPP)", in the UM cohort of The Cancer Genome Atlas Study and validated the differentially expressed genes in two independent cohorts. We also evaluated the effects of quercetin on the growth, survival, and glucose metabolism of the UM cell line 92.1. The rate-limiting glycolytic enzyme PFKP was overexpressed whereas the ZBTB20 gene (locus: 3q13.31) was downregulated in the patients with metastases in all cohorts. Quercetin was able to impair proliferation, viability, glucose uptake, glycolysis, ATP synthesis, and PPP rate-limiting enzyme activity while increasing oxidative stress. UMs with monosomy 3 display a stronger potential to utilize glucose for the generation of energy and biomass. Quercetin can prevent the growth of UM cells by interfering with glucose metabolism.


Subject(s)
Cell Proliferation , Glucose , Glycolysis , Melanoma , Quercetin , Uveal Neoplasms , Quercetin/pharmacology , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Melanoma/drug therapy , Humans , Uveal Neoplasms/metabolism , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Uveal Neoplasms/drug therapy , Glucose/metabolism , Glycolysis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Pentose Phosphate Pathway/drug effects , Chromosomes, Human, Pair 3/genetics
12.
Brain Res ; 1833: 148867, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38499234

ABSTRACT

The rate of early neurological deterioration (END) differs in different subtypes of ischaemic stroke. Previous studies showed PLCL2 gene is a novel susceptibility locus for the occurrence of atherosclerosis and thrombotic events. The objective of this research is to examine the efficacy that PLCL2 may have on the risk of END in large artery atherosclerotic (LAA) stroke. Tagged single nucleotide polymorphisms (SNPs) were identified by a strategy of fine-mapping. The genotyping of the selected SNPs was performed by SNPscan. The impact of PLCL2 on indicating the susceptibility of END in LAA patients was evaluated by binary logistic regression. The SNP-SNP interactions of PLCL2 for END was assessed by generalized multifactor dimensionality reduction (GMDR). A total of 1527 LAA stroke patients were recruited, 582 patients (38 %) experienced END. Compared to participants without END, participants experienced END were much older (P = 0.018), more likely to suffer pre-existing diabetes mellitus (P = 0.036), higher frequent in active tobacco users (P = 0.022) and had much higher median NIHSS on admission (P < 0.001). Rs4685423 was identified to be a predictor to the risk of END: the frequency of END in AA genotype patients is lower than that in AC or CC genotype patients (multivariate-adjusted, OR 0.63; 95 % CI 0.49-0.80; P < 0.001). The SNP-SNP interactions analysis indicates rs4685423 has the greatest impacton the risk of END for LAA patients. The time from admission diagnosis to END onset in AA genotype patients is much later than that in CA or CC genotype patients (log-rank, P = 0.005). In summary, the PLCL2 rs4685423 SNP is probably associated with the END risk in LAA stroke patients.


Subject(s)
Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Stroke , Humans , Male , Female , Polymorphism, Single Nucleotide/genetics , Aged , Middle Aged , Stroke/genetics , Genetic Predisposition to Disease/genetics , Atherosclerosis/genetics , Chromosomes, Human, Pair 3/genetics , Risk Factors , Genotype , Intracranial Arteriosclerosis/genetics
13.
Am J Med Genet A ; 194(7): e63531, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38421086

ABSTRACT

Duplications of the 3q29 cytoband are rare chromosomal copy number variations (CNVs) (overlapping or recurrent ~1.6 Mb 3q29 duplications). They have been associated with highly variable neurodevelopmental disorders (NDDs) with various associated features or reported as a susceptibility factor to the development of learning disabilities and neuropsychiatric disorders. The smallest region of overlap and the phenotype of 3q29 duplications remain uncertain. We here report a French cohort of 31 families with a 3q29 duplication identified by chromosomal microarray analysis (CMA), including 14 recurrent 1.6 Mb duplications, eight overlapping duplications (>1 Mb), and nine small duplications (<1 Mb). Additional genetic findings that may be involved in the phenotype were identified in 11 patients. Focusing on apparently isolated 3q29 duplications, patients present mainly mild NDD as suggested by a high rate of learning disabilities in contrast to a low proportion of patients with intellectual disabilities. Although some are de novo, most of the 3q29 duplications are inherited from a parent with a similar mild phenotype. Besides, the study of small 3q29 duplications does not provide evidence for any critical region. Our data suggest that the overlapping and recurrent 3q29 duplications seem to lead to mild NDD and that a severe or syndromic clinical presentation should warrant further genetic analyses.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 3 , DNA Copy Number Variations , Phenotype , Humans , Female , Male , Chromosomes, Human, Pair 3/genetics , Chromosome Duplication/genetics , Child , DNA Copy Number Variations/genetics , Child, Preschool , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Adolescent , Cohort Studies , Intellectual Disability/genetics , Intellectual Disability/pathology , Adult , Infant
14.
Ital J Pediatr ; 49(1): 17, 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36759911

ABSTRACT

BACKGROUND: Duplications of the long arm of chromosome 3 are rare, and associated to a well-defined contiguous gene syndrome known as partial trisomy 3q syndrome. It has been first described in 1966 by Falek et al., and since then around 100 patients have been reported. Clinical manifestations include characteristic facial dysmorphic features, microcephaly, hirsutism, congenital heart disease, genitourinary anomalies, hand and feet abnormalities, growth disturbances and intellectual disability. Most of cases are due to unbalanced translocations, inherited from a parent carrying a balanced aberration (reciprocal translocation or inversion), and rarely the genomic anomaly arises de novo. Very few studies report on the prenatal identification of such rearrangements. CASE PRESENTATION: Hereby, we report on a newborn with a rare pure duplication of the long arm of chromosome 3. Noninvasive prenatal test (cell free fetal DNA analysis on maternal blood), performed for advanced parental age and use of assisted reproductive technique, evidenced a partial 3q trisomy. Then, invasive cytogenetic (standard and molecular) investigations, carried out through amniocentesis, confirmed and defined a 3q27.1-q29 duplication spanning 10.9 Mb, and including about 80 genes. Our patient showed clinical findings (typical facial dysmorphic features, esotropia, short neck, atrial septal defect, hepatomegaly, mild motor delay) compatible with partial trisomy 3q syndrome diagnosis, in addition to pre- and postnatal overgrowth. CONCLUSIONS: Advanced parental age increases the probability of chromosomal and/or genomic anomalies, while ART that of epigenomic defects. Both conditions, thus, deserve more careful prenatal monitoring and screening/diagnostic investigations. Among the latter, cell free fetal DNA testing can detect large segmental aneuploidies, along with chromosomal abnormalities. It identified in our patient a wide 3q rearrangement, then confirmed and defined through invasive molecular cytogenetic analysis. Neonatologists and pediatricians must be aware of the potential risks associated to duplication syndromes. Therefore, they should offer to affected subjects an adequate management and early and careful follow-up. These may be able to guarantee to patients satisfactory growth and development profiles, prevent and/or limit neurodevelopmental disorders, and timely recognition of complications.


Subject(s)
Abnormalities, Multiple , Trisomy , Pregnancy , Infant, Newborn , Female , Humans , Trisomy/diagnosis , Trisomy/genetics , Chromosomes, Human, Pair 3/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , DNA
15.
Invest Ophthalmol Vis Sci ; 64(1): 15, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36656566

ABSTRACT

Purpose: Choroidal melanoma (CM) and ciliary body melanoma (CBM) are the two most common subtypes of uveal melanoma. Starting from the observation that CBM tends to have a higher metastatic potential than CM, we hypothesized that specific cytogenetic abnormalities could be associated with tumor location - reflecting distinct genetic signatures that would drive the risk of distant spread. Methods: Chromosomal alterations were investigated by molecular cytogenetic techniques in 217 and 97 patients with CM and CBM, respectively. Cox proportional hazards regression analysis was used to identify the independent predictors of distant metastasis. Results: Patients with CBM had larger tumor sizes (P < 0.001), higher disease stages (P < 0.001), and more frequently showed distant metastasis (P = 0.002) than those with CM. On analyzing the entire study cohort, we found that specific chromosomal alterations - including chromosome 8p loss (P < 0.001), 1p loss (P < 0.001), and monosomy 3 (P < 0.005) - were independent predictors of distant metastasis. Based on a decision-tree learning algorithm, we identified three specific subgroups of patients with uveal melanoma at high risk of distant spread. Monosomy 3 occurred significantly more frequently in patients with T3 CBM tumors. Conclusions: Specific cytogenetic abnormalities - including chromosome 8p loss, 1p loss, and monosomy 3 - are independent risk factors for distant metastasis in uveal melanoma. Larger tumor size at presentation and monosomy 3 contribute to a higher metastatic risk in patients with CBM.


Subject(s)
Choroid Neoplasms , Melanoma , Uveal Neoplasms , Humans , Ciliary Body/pathology , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Melanoma/genetics , Melanoma/pathology , Choroid Neoplasms/genetics , Choroid Neoplasms/pathology , Chromosome Deletion , Monosomy , Risk Factors , Chromosomes, Human, Pair 3/genetics
16.
Cancer Genomics Proteomics ; 19(6): 740-746, 2022.
Article in English | MEDLINE | ID: mdl-36316043

ABSTRACT

BACKGROUND/AIM: The Von Hippel-Lindau (VHL) gene encodes a protein (pVHL) that plays an important role in proteasome degradation of hypoxia inducible factor α (HIFα) through E3 activation. Accumulation of HIFα by loss of functional pVHL promotes tumorigenesis, thus, VHL has tumor suppressor gene capability in clear cell renal cell carcinoma (ccRCC). VHL is the most frequently mutated gene in ccRCC. The complete loss of VHL is mainly achieved by loss of chromosome 3p, which has a VHL coding region in combination with mutation or hypermethylation of the remaining copy of VHL. Given the risk of constitutional chromosome 3 translocation for RCC, it is important to detect the translocation and understand the mechanism underlying the development of multifocal ccRCC. CASE REPORT: A 67-year-old female patient diagnosed with multifocal RCC underwent robot-assisted partial nephrectomy (RAPN) for three kidney tumors. A cancer gene panel test using next generation sequencing (NGS) detected differential VHL mutations (c.533T>G; p.L178R, c.465_466insTA; p.T157Ifs*3, c.343C>A; p.H115N), while VHL mutation was not detected in peripheral blood DNA. A tendency toward copy number loss of genes on der(3) was also detected in all tumors, but not in the germline one. A karyotype analysis revealed a germline translocation between 3 and 6, t(3;6)(q12;q14). CONCLUSION: Chromosome 3 translocation and loss of derivative chromosome containing 3p and subsequent somatic differential VHL mutations in this case strongly support the previously proposed three-step model to explain the development of familial conventional ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Female , Humans , Aged , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Chromosomes, Human, Pair 3/genetics , Kidney Neoplasms/pathology , Mutation , Translocation, Genetic , Von Hippel-Lindau Tumor Suppressor Protein/genetics
17.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35193979

ABSTRACT

There are genetic risk factors that influence the outcome of COVID-19 [COVID-19 Host Genetics Initiative, Nature 600, 472-477 (2021)]. The major genetic risk factor for severe COIVD-19 resides on chromosome 3 and is inherited from Neandertals [H. Zeberg, S. Pääbo, Nature 587, 610-612 (2020)]. The risk-associated DNA segment modulates the expression of several chemokine receptors, among them CCR5, a coreceptor for HIV which is down-regulated in carriers of the COVID-19 risk haplotype. Here I show that carriers of the risk variant have an ∼27% lower risk of HIV infection.


Subject(s)
COVID-19/genetics , Chromosomes, Human, Pair 3/genetics , HIV Infections/genetics , HIV-1 , Haplotypes , SARS-CoV-2 , COVID-19/prevention & control , HIV Infections/prevention & control , Humans , Patient Acuity , Risk Factors
18.
EBioMedicine ; 75: 103806, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34998241

ABSTRACT

BACKGROUND: To identify host genetic variants (SNPs) associated with COVID-19 disease severity, a number of genome-wide association studies (GWAS) have been conducted. Since most of the identified variants are located at non-coding regions, such variants are presumed to affect the expression of neighbouring genes, thereby influencing COVID-19 disease severity. However, it remains largely unknown which genes are influenced by such COVID-19 GWAS loci. METHODS: CRISPRi (interference)-mediated gene expression analysis was performed to identify genes functionally regulated by COVID-19 GWAS loci by targeting regions near the loci (SNPs) in lung epithelial cell lines. The expression of CRISPRi-identified genes was investigated using COVID-19-contracted human and monkey lung single-nucleus/cell (sn/sc) RNA-seq datasets. FINDINGS: CRISPRi analysis indicated that a region near rs11385942 at chromosome 3p21.31 (locus of highest significance with COVID-19 disease severity at intron 5 of LZTFL1) significantly affected the expression of LZTFL1 (P<0.05), an airway cilia regulator. A region near rs74956615 at chromosome 19p13.2 (locus located at the 3' untranslated exonic region of RAVER1), which is associated with critical illness in COVID-19, affected the expression of RAVER1 (P<0.05), a coactivator of MDA5 (IFIH1), which induces antiviral response genes, including ICAM1. The sn/scRNA-seq datasets indicated that the MDA5/RAVER1-ICAM1 pathway was activated in lung epithelial cells of COVID-19-resistant monkeys but not those of COVID-19-succumbed humans. INTERPRETATION: Patients with risk alleles of rs11385942 and rs74956615 may be susceptible to critical illness in COVID-19 in part through weakened airway viral clearance via LZTFL1-mediated ciliogenesis and diminished antiviral immune response via the MDA5/RAVER1 pathway, respectively. FUNDING: NIH.


Subject(s)
COVID-19/genetics , CRISPR-Cas Systems , Genetic Loci , Polymorphism, Single Nucleotide , Ribonucleoproteins/genetics , SARS-CoV-2/genetics , Transcription Factors/genetics , Animals , COVID-19/metabolism , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 19/metabolism , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 3/metabolism , Databases, Nucleic Acid , Genome-Wide Association Study , Haplorhini , Humans , RNA-Seq , Ribonucleoproteins/metabolism , SARS-CoV-2/metabolism , Transcription Factors/metabolism
19.
Ophthalmology ; 129(4): 421-430, 2022 04.
Article in English | MEDLINE | ID: mdl-34780841

ABSTRACT

PURPOSE: Individuals with gray, blue, or green eyes have a higher chance of developing uveal melanoma (UM) than those with brown eyes. We wondered whether iris pigmentation might be related not only to predisposition to UM but also to its behavior; therefore, we compared the clinical, histopathologic, and genetic characteristics of UM between eyes with different colors. DESIGN: We determined iris color in a large cohort of patients enucleated for UM. Clinical and histopathologic tumor characteristics, chromosome status, and survival were compared among 3 groups on the basis of iris color. PARTICIPANTS: A total of 412 patients with choroidal/ciliary body UM, who had undergone primary enucleation at the Leiden University Medical Center, Leiden, The Netherlands, between 1993 and 2019, were divided into 3 groups based on iris color: gray/blue, green/hazel, and brown. The validation cohort included 934 patients with choroidal/ciliary body UM treated at Wills Eye Hospital (WEH). METHODS: Comparison of clinical, histopathologic, and genetic characteristics of UM in patients with different iris colors. MAIN OUTCOME MEASURES: Melanoma-related survival in UM patients, divided over 3 iris color groups, in relation to the tumor's chromosome 3 and 8q status. RESULTS: Moderate and heavy tumor pigmentations were especially seen in eyes with a brown iris (P < 0.001). Survival did not differ between patients with different iris colors (P = 0.27); however, in patients with a light iris, copy number changes in chromosome 3 and 8q had a greater influence on survival than in patients with a dark iris. Likewise, chromosome 3 and chromosome 8q status affected survival more among patients with lightly pigmented tumors than in patients with heavily pigmented tumors. The WEH cohort similarly showed a greater influence of chromosome aberrations in light-eyed individuals. CONCLUSIONS: Although iris color by itself did not relate to UM-related survival, chromosome 3 and 8q aberrations had a larger influence on survival in patients with a light iris than those with a brown iris. This suggests a synergistic effect of iris pigmentation and chromosome status in the regulation of oncogenic behavior of UM. Iris color should be taken into consideration when calculating a patient's risk for developing metastases.


Subject(s)
Melanoma , Uveal Neoplasms , Chromosome Aberrations , Chromosomes, Human, Pair 3/genetics , Eye Color/genetics , Humans , Iris/pathology , Melanoma/pathology , Prognosis , Uveal Neoplasms/pathology
20.
Nat Genet ; 53(11): 1606-1615, 2021 11.
Article in English | MEDLINE | ID: mdl-34737427

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS­CoV­2) disease (COVID-19) pandemic has caused millions of deaths worldwide. Genome-wide association studies identified the 3p21.31 region as conferring a twofold increased risk of respiratory failure. Here, using a combined multiomics and machine learning approach, we identify the gain-of-function risk A allele of an SNP, rs17713054G>A, as a probable causative variant. We show with chromosome conformation capture and gene-expression analysis that the rs17713054-affected enhancer upregulates the interacting gene, leucine zipper transcription factor like 1 (LZTFL1). Selective spatial transcriptomic analysis of lung biopsies from patients with COVID-19 shows the presence of signals associated with epithelial-mesenchymal transition (EMT), a viral response pathway that is regulated by LZTFL1. We conclude that pulmonary epithelial cells undergoing EMT, rather than immune cells, are likely responsible for the 3p21.31-associated risk. Since the 3p21.31 effect is conferred by a gain-of-function, LZTFL1 may represent a therapeutic target.


Subject(s)
COVID-19/complications , Chromosomes, Human, Pair 3/genetics , Epithelial-Mesenchymal Transition , Lung/virology , Polymorphism, Single Nucleotide , SARS-CoV-2/isolation & purification , Transcription Factors/genetics , COVID-19/transmission , COVID-19/virology , Case-Control Studies , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Genome-Wide Association Study , Humans , Lung/metabolism , Lung/pathology , Male , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL