860 results on '"GNAS"'
Search Results
2. Steroids-producing nodules: a two-layered adrenocortical nodular structure as a precursor lesion of cortisol-producing adenoma
- Author
-
Fukumoto, Tazuru, Umakoshi, Hironobu, Iwahashi, Norifusa, Ogasawara, Tatsuki, Yokomoto-Umakoshi, Maki, Kaneko, Hiroki, Fujita, Masamichi, Uchida, Naohiro, Nakao, Hiroshi, Kawamura, Namiko, Matsuda, Yayoi, Sakamoto, Ryuichi, Miyazawa, Takashi, Seki, Masahide, Eto, Masatoshi, Oda, Yoshinao, Suzuki, Yutaka, Ogawa, Seishi, and Ogawa, Yoshihiro
- Published
- 2024
- Full Text
- View/download PDF
3. Umschriebenes choroidales Hämangiom bei Sturge-Weber-Syndrom.
- Author
-
Vetter, Anna, Zimpfer, Annette, Schneider, Björn, Erbersdobler, Andreas, Brockmann, Tobias, Fuchsluger, Thomas, and Brockmann, Claudia
- Abstract
Copyright of Die Pathologie is the property of Springer Nature and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2025
- Full Text
- View/download PDF
4. Heterodisomy in the GNAS locus is also a cause of pseudohypoparathyroidism type 1B (iPPSD3).
- Author
-
Manero-Azua, Africa, Vado, Yerai, Gonzàlez Morlà, Judith, Mogas, Eduard, Pereda, Arrate, and Perez de Nanclares, Guiomar
- Subjects
GENETIC regulation ,SINGLE nucleotide polymorphisms ,LOCUS (Genetics) ,CHROMOSOMES ,EPIGENETICS - Abstract
Objective: To identify the genetic cause underlying the methylation defect in a patient with clinical suspicion of PHP1B/iPPSD3. Design: Imprinting is an epigenetic mechanism that allows the regulation of gene expression. The GNAS locus is one of the loci within the genome that is imprinted. When the methylation pattern is affected, it causes pseudohypoparathyroidism type 1B (PHP1B) or inactivating PTH/PTHrP signaling disorder 3 (iPPSD3). Paternal uniparental isodisomy (iUPDpat) of the chromosomal region comprising the GNAS locus has been described as one of the possible underlying genetic causes of the methylation alteration. Methods: We present the case of a patient clinically diagnosed with iPPSD3. We performed a commercial methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA), single-nucleotide polymorphism (SNP) array, and microsatellite study. In addition, we designed a custom MS-MLPA to analyze GNAS and nearby differentially methylated regions (DMRs). Results: A methylation defect at the four GNAS -DMRs was detected, confirming the clinical diagnosis. Complementary techniques revealed the presence of a mixed isodisomy and heterodisomy of chromosome 20. Surprisingly, the GNAS locus was located on the heterodisomic zone. Conclusions: Paternal uniparental heterodisomy (hUPD) at the GNAS locus is also a genetic defect associated with iPPSD3. In the absence of parental samples, our custom MS-MLPA allows for the detection of a methylation defect at the GNAS locus and flanking DMRs, suggestive of uniparental disomy (UPD). We also suggest updating the actual guidelines to include hUPD at the GNAS locus as a cause of iPPSD3. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Next-generation sequencing improves diagnostic accuracy of imaging and carcinoembryonic antigen alone for pancreatic cystic neoplasms.
- Author
-
Belfrage, Hanna, Boyd, Sonja, Louhimo, Johanna, Kytölä, Soili, Johansson, Katarina, Tenca, Andrea, Puustinen, Lauri, Kokkola, Arto, Arkkila, Perttu, Arola, Johanna, and Seppänen, Hanna
- Abstract
New tools are needed to determine the pancreatic cysts that require surgical resection. This study aimed to evaluate whether next-generation sequencing (NGS) is useful for identifying mucinous, malignant, or pre-malignant cysts leading to surgery. Laboratory, cytological, and histological data from 97 patients with worrisome features on imaging or an unclear pancreatic cystic lesion (PCL) who were indicated for further investigation and who underwent endoscopic ultrasound (EUS) and fine-needle aspiration (FNA) between 2018 and 2022 were analyzed. A multidisciplinary team evaluated MRI, CT, EUS-FNA, and NGS findings. Among the 40 mucinous cysts, 53 % had KRAS and/or GNAS mutations, yielding a sensitivity of 53 % and specificity of 92 % compared to 33 and 100 % for cytology and 53 and 89 % for cystic fluid CEA. Combining NGS findings with CEA levels increased sensitivity and specificity in detecting mucinous lesions to 78 and 87 %, respectively. Surgically treated high-grade dysplasia PCLs did not show worrisome mutations in cyst fluid, while 80 % of the malignant lesions had mutations typical for advanced lesions. The advanced neoplasias showed 95 % specificity for worrisome gene mutations, with the highest diagnostic accuracy observed for NGS mutations, achieving an AUC of 0.777 in the ROC curve analysis compared to 0.631 for CEA. Patients with worrisome gene mutations were offered surgical treatment. NGS results contributed to the decision to operate in 11 out of 23 cases. In 71 % of all cases, NGS supported the diagnosis, with 3 % false positives and 12 % false negatives. NGS analysis of pancreatic cyst fluid demonstrates high specificity and may serve as an additional diagnostic tool to CEA. Combining cystic fluid CEA and NGS increases the accuracy in determining whether a lesion is mucinous and NGS showed a higher diagnostic accuracy in advanced lesions compared to CEA. While the absence of alarming NGS findings should not preclude surgical treatment, patients with alarming mutations should be considered for surgery. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. PRKACB is a novel imprinted gene in marsupials
- Author
-
Trent Newman, Donna M. Bond, Teruhito Ishihara, Phoebe Rizzoli, Quentin Gouil, Timothy A. Hore, Geoff Shaw, and Marilyn B. Renfree
- Subjects
Genomic imprinting ,PRKACB ,GNAS ,Convergent evolution ,Differential methylation ,Genetics ,QH426-470 - Abstract
Abstract Background Genomic imprinting results in parent-of-origin-specific gene expression and, among vertebrates, is found only in therian mammals: marsupials and eutherians. A differentially methylated region (DMR), in which the methylation status of CpG dinucleotides differs between the two alleles, can mark the parental identity of imprinted genes. We developed a computational pipeline that detected CpG islands (CGIs) marked by both methylated and unmethylated signals in whole genome bisulfite sequencing data. This approach identified candidate marsupial DMRs in a publicly available koala methylome. One of these candidate DMRs was associated with PRKACB, a gene encoding the protein kinase A catalytic subunit beta. Nothing is known about the imprinting status of PRKACB in eutherian mammals although mutations of this gene are associated with endocrine neoplasia and other developmental disorders. Results In the tammar wallaby and brushtail possum there was parent-of-origin-specific DNA methylation in the PRKACB DMR in which the maternal allele was methylated and the paternal allele was unmethylated. There were multiple RNAs transcribed from this locus. Allele-specific expression analysis identified paternal expression of a PRKACB lncRNA and an mRNA isoform. Comparison of the PRKACB gene start site between marsupials and eutherians demonstrated that the CGI is longer in marsupials. The PRKACB gene product functions in the same signalling pathway as the guanine nucleotide-binding protein alpha subunit encoded at the GNAS locus, a known eutherian imprinted gene. In a mouse methylome Gnas had three differentially methylated CGIs, while in the koala methylome the GNAS locus had two unmethylated CGIs. Conclusions We conclude that PRKACB is a novel, DMR-associated marsupial imprinted gene. Imprinting of PRKACB in marsupials and GNAS in eutherians may indicate a conserved selection pressure for imprinting of the protein kinase A signalling pathway in therians with the two lineages adapting by imprinting different genes.
- Published
- 2024
- Full Text
- View/download PDF
7. Skipping of Exon 20 in EP300: A Novel Variant Linked to Rubinstein–Taybi Syndrome With Atypical and Severe Clinical Manifestations.
- Author
-
Pavinato, Lisa, Carestiato, Silvia, Trajkova, Slavica, Sorasio, Lorena, Mantovani, Giovanna, De Sanctis, Luisa, Kerkhof, Jennifer, Haley, Rzasa, Jessica, Todd, Emily, Balzo, Maria, Cardaropoli, Simona, Bruselles, Alessandro, De Rubeis, Silvia, Buxbaum, Joseph D., Tartaglia, Marco, Sadikovic, Bekim, Ferrero, Giovanni Battista, and Brusco, Alfredo
- Subjects
- *
AUTISM spectrum disorders , *HEARING disorders , *SYMPTOMS , *SHORT stature , *DEVELOPMENTAL delay - Abstract
ABSTRACT Rubinstein–Taybi syndrome (RSTS) is a rare autosomal dominant neurodevelopmental disorder linked to haploinsufficiency of CREBBP (RSTS1) and EP300 (RSTS2) genes. Characteristic features often include distinctive facial traits, broad thumbs and toes, short stature, and various degrees of intellectual disability. The clinical presentation of RSTS is notably variable, making it challenging to establish a clear genotype–phenotype correlation, except for specific variants which cause the allelic Menke–Hennekam syndrome. Trio exome analysis, data collection via networking and GeneMatcher platforms, transcript processing analysis, and DNA methylation profiling were performed. We identified two unrelated patients with de novo variants in EP300 (NM_001429.4: c.3671+5G>C; c.3671+5_3671+8delGTAA) predicted to cause in‐frame exon 20 skipping, confirmed in one patient. In silico 3D protein modeling suggested that exon 20 deletion (comprising 27 amino acids) likely alters the structural conformation between the RING_CBP‐p300 and HAT‐KAT11 domains. Clinically, both patients displayed severe RSTS2‐like clinical features, including autism spectrum disorder, speech delay, hearing loss, microcephaly, developmental delay, and intellectual disability, alongside ocular, respiratory, and cardiovascular abnormalities. Additionally, one patient developed early‐onset colorectal cancer. DNA methylation profiling in Subject #1 confirmed RSTS but did not align with the specific episignatures for RSTS1 or RSTS2. We propose that skipping of exon 20 in EP300 is associated with a distinct form of Rubinstein–Taybi syndrome featuring clinical characteristics not fully aligning with RSTS1 or RSTS2. Our findings increase the understanding of RSTS genetic and molecular basis and stress the need for further research to establish definitive genotype–phenotype correlations. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Genotype–Phenotype Correlation of GNAS Gene: Review and Disease Management of a Hotspot Mutation.
- Author
-
Cipriano, Lorenzo, Ferrigno, Rosario, Andolfo, Immacolata, Russo, Roberta, Cioffi, Daniela, Savanelli, Maria Cristina, Pellino, Valeria, Klain, Antonella, Iolascon, Achille, and Piscopo, Carmelo
- Subjects
- *
HETEROTOPIC ossification , *GENETIC variation , *INTELLECTUAL disabilities , *DISEASE management - Abstract
Defects of the GNAS gene have been mainly associated with pseudohypoparathyroidism Ia. To date, pathogenic missense, frameshift, non-sense and splicing variants have been described in all the 13 exons of the GNAS gene. Of them, a specific mutation, namely the 4 bp deletion c.565_568delGACT, is currently considered a mutation hotspot. Recent articles performed genotype–phenotype correlations in patients with GNAS-related pseudohypoparathyroidism Ia (PHP1a) but a specific focus on this hotspot is still lacking. We reported two cases, from our department, of PHP1a associated with c.565_568delGACT deletion and performed a literature review of all the previously reported cases of the 4 bp deletion hotspot. We found a higher prevalence of brachydactyly, round face, intellectual disability and subcutaneous/heterotopic ossifications in patients with the c.565_568delGACT as compared to the other variants in the GNAS gene. The present study highlights the different prevalence of some clinical features in patients with the c.565_568delGACT variant in the GNAS gene, suggesting the possibility of a personalized diagnostic follow-up and surveillance for these patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Mutational Landscape of Gastric Adenocarcinoma of the Fundic Gland Type Revealed by Whole Genome Sequencing.
- Author
-
Wei, Hu, Chen, Ze Min, Xue, Xiu Fen, Xia Xi, Li, Yang, Gen Hua, Zhai, Zhi Yong, Huang, Zhao Yu, Zhou, Ping, Bao, Chong Ju, You, Li Juan, Ou Yang, Mei Ping, Xia, Gui Li, Zeng, Zhi Yu, Cui, Xiao Bing, Pei, Xiao Juan, and Gong, Wei
- Subjects
- *
WHOLE genome sequencing , *FOCAL adhesions , *STOMACH cancer , *CELLULAR signal transduction , *PROTEOGLYCANS - Abstract
Background: Gastric adenocarcinoma of the fundic gland type (GA‐FG) is a newly described variant of gastric adenocarcinoma with lack of knowledges regarding its genetic features. Methods: We performed whole‐genome sequencing (WGS) in formalin‐fixed paraffin‐embedded (FFPE) tumor tissues and matched adjacent noncancerous specimens from 21 patients with GA‐FG, and integrated published datasets from 1105 patients with traditional gastric adenocarcinoma with the purpose of dissecting genetic determinants both common to conventional gastric adenocarcinoma and unique to GA‐FG disease. Results: We characterized the genomic architecture of GA‐FG disease, revealing the predominant proportion of C > T substitution among the six types of SNVs. GNAS was the most significantly mutated driver gene (14.29%). 42.8% of samples harbored "Kataegis." Distinct genomic alterations between GA‐FG and conventional gastric cancer were identified. Specifically, low mutational burden and relatively moderate mutational frequencies of significantly mutated driver genes, coupled with the absence of non‐silent alterations of formerly well‐known drivers such as TP53, PIK3CA and KRAS were identified in GA‐FG patients. Oncogenic signaling pathway analysis revealed mutational processes associated with focal adhesions and proteoglycans in cancer, highlighting both common and specific procedures during the development of GA‐FG and conventional gastric cancer. Conclusion: Our study is the first to comprehensively depict the genomic landscape highlighting the multidimensional perturbations in GA‐FG patients. These discoveries offered mechanistic insights for novel diagnostic and therapeutic strategies for patients with such disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. PRKACB is a novel imprinted gene in marsupials.
- Author
-
Newman, Trent, Bond, Donna M., Ishihara, Teruhito, Rizzoli, Phoebe, Gouil, Quentin, Hore, Timothy A., Shaw, Geoff, and Renfree, Marilyn B.
- Subjects
GENOMIC imprinting ,G proteins ,GENE expression ,WHOLE genome sequencing ,PROTEIN kinases - Abstract
Background: Genomic imprinting results in parent-of-origin-specific gene expression and, among vertebrates, is found only in therian mammals: marsupials and eutherians. A differentially methylated region (DMR), in which the methylation status of CpG dinucleotides differs between the two alleles, can mark the parental identity of imprinted genes. We developed a computational pipeline that detected CpG islands (CGIs) marked by both methylated and unmethylated signals in whole genome bisulfite sequencing data. This approach identified candidate marsupial DMRs in a publicly available koala methylome. One of these candidate DMRs was associated with PRKACB, a gene encoding the protein kinase A catalytic subunit beta. Nothing is known about the imprinting status of PRKACB in eutherian mammals although mutations of this gene are associated with endocrine neoplasia and other developmental disorders. Results: In the tammar wallaby and brushtail possum there was parent-of-origin-specific DNA methylation in the PRKACB DMR in which the maternal allele was methylated and the paternal allele was unmethylated. There were multiple RNAs transcribed from this locus. Allele-specific expression analysis identified paternal expression of a PRKACB lncRNA and an mRNA isoform. Comparison of the PRKACB gene start site between marsupials and eutherians demonstrated that the CGI is longer in marsupials. The PRKACB gene product functions in the same signalling pathway as the guanine nucleotide-binding protein alpha subunit encoded at the GNAS locus, a known eutherian imprinted gene. In a mouse methylome Gnas had three differentially methylated CGIs, while in the koala methylome the GNAS locus had two unmethylated CGIs. Conclusions: We conclude that PRKACB is a novel, DMR-associated marsupial imprinted gene. Imprinting of PRKACB in marsupials and GNAS in eutherians may indicate a conserved selection pressure for imprinting of the protein kinase A signalling pathway in therians with the two lineages adapting by imprinting different genes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. GNAS, not a Highly Mutated Gene, Has Prognostic Significance and Carcinogenic Effects in Osteosarcoma.
- Author
-
Jin Qi, Yanjiao Huang, and Yaogang Bian
- Subjects
- *
TUMORS in children , *ANIMAL experimentation , *GENE expression , *TUMOR growth , *IMMUNOHISTOCHEMISTRY - Abstract
Background/Aims: Osteosarcoma is a prevalent and aggressive primary malignant bone tumor affecting children and adolescents. Despite advancements in sequencing technologies, there remains a lack of reliable prognostic biomarkers and effective targeted therapies for osteosarcoma. This study focuses on identifying key prognostic genes, particularly the role of GNAS, in osteosarcoma progression. Methods: Bioinformatics analyses were performed on osteosarcoma datasets from the Gene Expression Omnibus (GEO). Differential gene expression analysis, weighted correlation network analysis (WGCNA), and survival analysis identified potential prognostic hub genes. The expression and function of these genes were validated through immunohistochemistry and animal experiments. Specifically, the role of GNAS was investigated through siRNA-mediated knockdown in osteosarcoma cell lines and nude mice models. Results: Five hub genes (PROP1, GNAS, CYP4F2, LHX3, CNGB1) were identified as significantly related to osteosarcoma prognosis. Among these, GNAS was found to be highly expressed in osteosarcoma tissues compared to normal tissues. Immunohistochemical analysis confirmed the elevated expression of GNAS in osteosarcoma samples. GNAS mutation analysis revealed a low mutation rate in osteosarcoma, suggesting its oncogenic role is independent of mutational status. Animal experiments demonstrated that knocking down GNAS significantly inhibited tumor growth and induced apoptosis in osteosarcoma cells. Conclusion: GNAS is highly expressed in osteosarcoma and associated with poor prognosis, acting as an oncogene in osteosarcoma progression. Targeting GNAS could be a potential therapeutic strategy for osteosarcoma. Further studies on GNAS-related signaling pathways may provide deeper insights into the molecular mechanisms driving osteosarcoma malignancy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. KRAS and GNAS mutations in cell‐free DNA and in circulating epithelial cells in patients with intraductal papillary mucinous neoplasms—an observational pilot study.
- Author
-
Nitschke, Christine, Tölle, Marie, Walter, Philipp, Meißner, Kira, Goetz, Mara, Kropidlowski, Jolanthe, Berger, Andreas W., Izbicki, Jakob R., Nickel, Felix, Hackert, Thilo, Pantel, Klaus, Wikman, Harriet, and Uzunoglu, Faik G.
- Subjects
- *
CELL-free DNA , *POLYMERASE chain reaction , *RAS oncogenes , *EPITHELIAL cells , *PANCREATIC cancer , *CIRCULATING tumor DNA - Abstract
Intraductal papillary mucinous neoplasms (IPMNs) are potential precursor lesions of pancreatic cancer. We assessed the efficacy of screening for KRAS proto‐oncogene, GTPase (KRAS), and GNAS complex locus (GNAS) mutations in cell‐free DNA (cfDNA)—using digital droplet polymerase chain reaction (ddPCR) and circulating epithelial cell (CEC) detection—as biomarkers for risk stratification in IPMN patients. We prospectively collected plasma samples from 25 resected patients at risk of malignant progression, and 23 under clinical surveillance. Our findings revealed KRAS mutations in 10.4% and GNAS mutations in 18.8% of the overall cohort. Among resected IPMN patients, KRAS and GNAS mutation detection rates were 16.0% and 32.0%, respectively, whereas both rates were 4.0% in conservatively managed IPMN. GNAS mutations in cfDNA were significantly more prevalent in resected IPMN (P = 0.024) compared with IPMN under surveillance. No CECs were detected. The absence of KRAS and GNAS mutations could be a reliable marker for branch duct IPMN without worrisome features. The emergence of GNAS mutations could prompt enhanced imaging surveillance. Neither the presence of established worrisome features nor GNAS or KRAS mutations appear effective in identifying high‐grade dysplasia among IPMN patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Heterodisomy in the GNAS locus is also a cause of pseudohypoparathyroidism type 1B (iPPSD3)
- Author
-
Africa Manero-Azua, Yerai Vado, Judith Gonzàlez Morlà, Eduard Mogas, Arrate Pereda, and Guiomar Perez de Nanclares
- Subjects
PHP1B ,iPPSD3 ,GNAS ,heterodisomy ,upd(20)pat ,MS-MLPA ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
ObjectiveTo identify the genetic cause underlying the methylation defect in a patient with clinical suspicion of PHP1B/iPPSD3.DesignImprinting is an epigenetic mechanism that allows the regulation of gene expression. The GNAS locus is one of the loci within the genome that is imprinted. When the methylation pattern is affected, it causes pseudohypoparathyroidism type 1B (PHP1B) or inactivating PTH/PTHrP signaling disorder 3 (iPPSD3). Paternal uniparental isodisomy (iUPDpat) of the chromosomal region comprising the GNAS locus has been described as one of the possible underlying genetic causes of the methylation alteration.MethodsWe present the case of a patient clinically diagnosed with iPPSD3. We performed a commercial methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA), single-nucleotide polymorphism (SNP) array, and microsatellite study. In addition, we designed a custom MS-MLPA to analyze GNAS and nearby differentially methylated regions (DMRs).ResultsA methylation defect at the four GNAS-DMRs was detected, confirming the clinical diagnosis. Complementary techniques revealed the presence of a mixed isodisomy and heterodisomy of chromosome 20. Surprisingly, the GNAS locus was located on the heterodisomic zone.ConclusionsPaternal uniparental heterodisomy (hUPD) at the GNAS locus is also a genetic defect associated with iPPSD3. In the absence of parental samples, our custom MS-MLPA allows for the detection of a methylation defect at the GNAS locus and flanking DMRs, suggestive of uniparental disomy (UPD). We also suggest updating the actual guidelines to include hUPD at the GNAS locus as a cause of iPPSD3.
- Published
- 2024
- Full Text
- View/download PDF
14. Unlocking the Genetic Secrets of Acromegaly: Exploring the Role of Genetics in a Rare Disorder
- Author
-
Ioana Balinisteanu, Lavinia Caba, Andreea Florea, Roxana Popescu, Laura Florea, Maria-Christina Ungureanu, Letitia Leustean, Eusebiu Vlad Gorduza, and Cristina Preda
- Subjects
acromegaly ,genetics ,PitNET ,AIP ,FIPA ,GNAS ,Biology (General) ,QH301-705.5 - Abstract
Acromegaly is a rare endocrine disorder characterized by the excessive production of growth hormone (GH) in adulthood. Currently, it is understood that certain pituitary neuroendocrine tumors (PitNETs) exhibit a hereditary predisposition. These tumors’ genetic patterns fall into two categories: isolated and syndromic tumors. The isolated forms are characterized by molecular defects that predispose exclusively to PitNETs, including familial isolated pituitary adenomas (FIPAs) and sporadic genetic defects not characterized by hereditary predisposition. All the categories involve either germline or somatic mutations, or both, each associated with varying levels of penetrance and different phenotypes. This highlights the importance of genetic testing and the need for a more comprehensive view of the whole disease. Despite the availability of multiple treatment options, diagnosis often occurs after several years, and management is still difficult. Early detection and intervention are crucial for preventing complications and enhancing the quality of life for affected individuals. This review aims to elucidate the molecular, clinical, and histological characteristics of GH-secreting PitNETs, providing insights into their prevalence, treatment nuances, and the benefits of genetic testing for each type of genetic disorder associated with acromegaly.
- Published
- 2024
- Full Text
- View/download PDF
15. The prevalence, diagnostic accuracy and genotype-phenotype correlation of GNAS mutations in fibrous dysplasia: a meta-analysis.
- Author
-
Ao-Bo Zhang, Jian-Yun Zhang, Jiang Xue, Zhen-Chao Wu, Zhi-Xiu Xu, Li-Sha Sun, and Tie-Jun Li
- Subjects
RECEIVER operating characteristic curves ,DYSPLASIA ,DATABASE searching ,SCIENTIFIC observation - Abstract
Background: There is inconsistent evidence regarding the accuracy of GNAS mutations identification forthe diagnosis of FD/MAS. This study was performed to estimate the prevalence and diagnostic accuracy of GNAS mutations detection and to preliminarily investigate the genotype-phenotype correlation in FD patients. Methods: Five electronic databases were searched from 1995 to 2024 using search terms related to GNAS and fibrous dysplasia. Observational studies of FD patients undergoing GNAS mutation detection in FD were included. Results: A total of 878 FD patients were included. The pooled prevalence of GNAS mutations in FD based on the random effects model was 74% (95% CI = 64%-83%). Regarding diagnostic accuracy, a sensitivity of 0.83 (95% CI, 0.65-0.96), specificity of 0.99 (95% CI, 0.98-1.00) and the area under the receiver operating characteristic curve of 98.38% were found. Additionally, meta-analysis and Fisher's test showed the GNAS mutation types were significantly associated with FD types (OR = 3.51, 95% CI = 1.05 to 11.72; p < 0.05). Conclusion: A high detection rate of GNAS mutations occurred in FD, and its detection is reliable for diagnosing FD. Additionally, GNAS mutation type was types were significantly associated with FD type. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. A Novel Maternally Inherited GNAS Variant in a Family With Hyperphagia and Obesity: 3 Cases.
- Author
-
Ramakrishnan, Anand, Popat, Dillon, Purushothaman, Preetha, Chan, Li F., and Gevers, Evelien F.
- Subjects
- *
OBESITY genetics , *MELANOCORTIN receptors , *BODY mass index , *CHILDHOOD obesity , *HORMONE receptors - Abstract
GNAS variants were recently described in 1% of patients not known to have pseudohypoparathyroidism/inactivating PTH/PTHrP signalling disorder 2 in the UK Genetics of Obesity Study. We describe a new missense GNAS variant, c.791A > C, p.(Asp264Thr), in a family with obesity, hyperphagia and mild PTH resistance. A 6-year-old female (body mass index +4.3 SD score [SDS], height +1.9 SDS) presented with hyperphagia and obesity from age 3 years. She had subtle brachydactyly, macrocephaly, and mildly delayed development. The 12-year-old brother (height +2.1 SDS, body mass index +2.9 SDS) had hyperphagia, obesity, mildly delayed development, and autism. He had subtle brachydactyly, as did the affected mother. We assessed the functional effect of the mutant, measuring cAMP production in cells transfected with wild type and mutant GNAS after ligand stimulation. Cells with the mutant GNAS showed impaired cAMP generation through melanocortin receptor 4, GH releasing hormone receptor, and PTH receptor. These cases demonstrate the clinical heterogeneity of monogenic disease, suggesting a need to test for PHP1A in children with obesity even without classical signs of PHP1A. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. STX16 exon 5–7 deletion in a patient with pseudohypoparathyroidism type 1B.
- Author
-
Chen, Li, Yang, Chuanbin, Zhang, Xiaoxiao, Chen, Beibei, Zheng, Peibing, Li, Tingting, Song, Wenjing, Gao, Hua, Yue, Xiaofang, and Yang, Jiajun
- Abstract
Pseudohypoparathyroidism (PHP) comprises a cluster of heterogeneous diseases characterized by hypocalcemia and hyperphosphatemia due to parathyroid hormone (PTH) resistance. PHP type 1B (PHP1B) is caused by heterozygous maternal deletions within GNAS or STX16. STX16 exon 2–6 deletion is commonly observed in autosomal dominant (AD)-PHP1B, while sporadic PHP1B commonly results from methylation abnormalities of maternal differentially methylated regions and remains unclear at the molecular level. A 39-year-old male patient with PHP1B, who had his first seizure at 15 years of age, presented to our hospital. The methylation-specific multiplex ligation-dependent probe amplification results showed a half-reduced copy number of STX16 exon 5–7 and loss of methylation at GNAS exon A/B. His mother also had a half-reduced copy number of STX16 exon 5–7 but with normal methylation of GNAS. His father has a normal copy number of STX16 and normal methylation of GNAS. For the recognition and early diagnosis of this kind of disease, here we report the clinical symptoms, auxiliary examinations, genetic testing characteristics, and treatment of the patient. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. OSTEODISTROFIA HEREDITARIA DE ALBRIGHT. ESTUDIO MOLECULAR Y SU ASOCIACIÓN FENOTÍPICA.
- Author
-
Verónica Cuello, Jimena, Silberkasten, Malena, and Eugenia Andrés, María
- Subjects
OBESITY complications ,INBORN errors of metabolism diagnosis ,METALS in the body ,INBORN errors of metabolism ,HOSPITALS ,GENES ,PHENOTYPES ,SYMPTOMS - Abstract
Copyright of Actualización en Nutrición is the property of Sociedad Argentina de Nutricion and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
19. GNAS mutation inhibits growth and induces phosphodiesterase 4D expression in colorectal cancer cell lines.
- Author
-
Nummela, Pirjo, Zafar, Sadia, Veikkolainen, Erika, Ukkola, Iiris, Cinella, Vincenzo, Ayo, Abiodun, Asghar, Muhammad Yasir, Välimäki, Niko, Törnquist, Kid, Karhu, Auli, Laakkonen, Pirjo, Aaltonen, Lauri A., and Ristimäki, Ari
- Subjects
GENE expression ,COLORECTAL cancer ,CELL lines ,GAIN-of-function mutations ,CANCER cells ,CANCER cell growth ,CYCLIC-AMP-dependent protein kinase ,CETUXIMAB - Abstract
Approximately 5% of colorectal cancers (CRCs) have a gain‐of‐function mutation in the GNAS gene, which leads to the activation of cAMP‐dependent signaling pathways and associates with poor prognosis. We investigated the effect of an activating GNAS mutation in CRC cell lines on gene expression and cell proliferation in vitro, and tumor growth in vivo. GNAS‐mutated (GNASmt) HCT116 cells showed stimulated synthesis of cAMP as compared to parental (Par) cells. The most upregulated gene in the GNASmt cells was cAMP‐hydrolyzing phosphodiesterase 4D (PDE4D) as detected by RNA sequencing. To further validate our finding, we analyzed PDE4D expression in a set of human CRC tumors (n = 35) and demonstrated overexpression in GNAS mutant CRC tumors as compared to GNAS wild‐type tumors. The GNASmt HCT116 cells proliferated more slowly than the Par cells. PDE4 inhibitor Ro 20‐1724 and PDE4D subtype selective inhibitor GEBR‐7b further suppressed the proliferation of GNASmt cells without an effect on Par cells. The growth inhibitory effect of these inhibitors was also seen in the intrinsically GNAS‐mutated SK‐CO‐1 CRC cell line having high levels of cAMP synthesis and PDE4D expression. In vivo, GNASmt HCT116 cells formed smaller tumors than the Par cells in nude mice. In conclusion, our findings demonstrate that GNAS mutation results in the growth suppression of CRC cells. Moreover, the GNAS mutation‐induced overexpression of PDE4D provides a potential avenue to impede the proliferation of CRC cells through the use of PDE4 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Pseudohypoparathyroidism
- Author
-
Germain-Lee, Emily L., Levine, Michael A., Radovick, Sally, editor, and Misra, Madhusmita, editor
- Published
- 2024
- Full Text
- View/download PDF
21. Pseudohypoparathyroidism Type IB with Subclinical Hypothyroidism: a Pedigree Investigation and Literature Review
- Author
-
Liu J, Lu L, Wei Y, Li Y, Wang Q, Yu L, Zhuang L, Jin G, and Pei X
- Subjects
hypocalcemia ,parathyroid hormone resistance ,gnas ,stx16 ,molecular genetic mechanism ,Specialties of internal medicine ,RC581-951 - Abstract
Jie Liu, Lijuan Lu, Yu Wei, Yu Li, Qiong Wang, Lei Yu, Langen Zhuang, Guoxi Jin, Xiaoyan Pei Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui Province, People’s Republic of ChinaCorrespondence: Guoxi Jin; Xiaoyan Pei, Department of Endocrinology, the First Affiliated Hospital of Bengbu Medical University, No. 287 Changhuai Road, Bengbu, Anhui Province, 233000, People’s Republic of China, Email jyzjyz1999@163.com; 245899985@qq.comAbstract: Pseudohypoparathyroidism (PHP) is a rare genetic disease characterized by hypocalcemia, hyperphosphatemia, and elevated parathyroid hormone (PTH) in serum. Here, we report a case of a patient with pseudohypoparathyroidism type IB (PHPIB) and subclinical hypothyroidism, analyze the clinical and genetic data of his family members, review the relevant literature, and classify and discuss the pathogenesis and clinical characteristics of each subtype. Finally, we discuss the treatment approach to improve clinicians’ understanding of the disease.Keywords: hypocalcemia, parathyroid hormone resistance, GNAS, STX16, molecular genetic mechanism
- Published
- 2024
22. Intramuscular myxoma: unusual observation of spontaneous tumor size shrinkage
- Author
-
Khoury, Charbel El, Taihi, Lokmane, Evrard, Robin, De Roo, An-Katrien, Lecouvet, Frédéric, and Schubert, Thomas
- Published
- 2024
- Full Text
- View/download PDF
23. Targeted silencing of GNAS in a human model of osteoprogenitor cells results in the deregulation of the osteogenic differentiation program.
- Author
-
Elli, Francesca Marta, Mattinzoli, Deborah, Ikehata, Masami, Bagnaresi, Francesca, Maffini, Maria A., Del Sindaco, Giulia, Pagnano, Angela, Lucca, Camilla, Messa, Piergiorgio, Arosio, Maura, Castellano, Giuseppe, Alfieri, Carlo M., and Mantovani, Giovanna
- Subjects
MESENCHYMAL stem cell differentiation ,BONE growth ,TRANSGENIC mice ,GENE silencing - Abstract
Introduction: The dysregulation of cell fate toward osteoprecursor cells associated with most GNAS-based disorders may lead to episodic de novo extraskeletal or ectopic bone formation in subcutaneous tissues. The bony lesion distribution suggests the involvement of abnormal differentiation of mesenchymal stem cells (MSCs) and/or more committed precursor cells. Data from transgenic mice support the concept that GNAS is a crucial factor in regulating lineage switching between osteoblasts (OBs) and adipocyte fates. The mosaic nature of heterotopic bone lesions suggests that GNAS genetic defects provide a sensitized background for ectopic osteodifferentiation, but the underlying molecular mechanism remains largely unknown. Methods: The effect of GNAS silencing in the presence and/or absence of osteoblastic stimuli was evaluated in the human L88/5 MSC line during osteodifferentiation. A comparison of the data obtained with data coming from a bony lesion from a GNAS-mutated patient was also provided. Results: Our study adds some dowels to the current fragmented notions about the role of GNAS during osteoblastic differentiation, such as the premature transition of immature OBs into osteocytes and the characterization of the differences in the deposed bone matrix. Conclusion: We demonstrated that our cell model partially replicates the in vivo behavior results, resulting in an applicable human model to elucidate the pathophysiology of ectopic bone formation in GNAS-based disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Medulloblastoma in a child with osteoma cutis – a rare association due to loss of GNAS expression.
- Author
-
Suntharesan, Jananie, Lyulcheva-Bennett, Ekaterina, Hart, Rachel, Pizer, Barry, Hayden, James, and Ramakrishnan, Renuka
- Abstract
Inactivating GNAS mutations result in varied phenotypes depending on parental origin. Maternally inherited mutations typically lead to hormone resistance and Albright's hereditary osteodystrophy (AHO), characterised by short stature, round facies, brachydactyly and subcutaneous ossifications. Paternal inheritance presents with features of AHO or ectopic ossification without hormone resistance. This report describes the case of a child with osteoma cutis and medulloblastoma. The objective of this report is to highlight the emerging association between inactivating germline GNAS mutations and medulloblastoma, aiming to shed light on its implications for tumor biology and promote future development of targeted surveillance strategies to improve outcomes in paediatric patients with these mutations. A 12-month-old boy presented with multiple plaque-like skin lesions. Biopsy confirmed osteoma cutis, prompting genetic testing which confirmed a heterozygous inactivating GNAS mutation. At 2.5 years of age, he developed neurological symptoms and was diagnosed with a desmoplastic nodular medulloblastoma, SHH molecular group, confirmed by MRI and histology. Further analysis indicated a biallelic loss of GNAS in the tumor. This case provides important insights into the role of GNAS as a tumor suppressor and the emerging association between inactivating GNAS variants and the development of medulloblastoma. The case underscores the importance of careful neurological assessment and ongoing vigilance in children with known inactivating GNAS variants or associated phenotypes. Further work to establish genotype–phenotype correlations is needed to inform optimal management of these patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Wnt pathway inhibition with the porcupine inhibitor LGK974 decreases trabecular bone but not fibrosis in a murine model with fibrotic bone.
- Author
-
Lung, Hsuan, Wentworth, Kelly L, Moody, Tania, Zamarioli, Ariane, Ram, Apsara, Ganesh, Gauri, Kang, Misun, Ho, Sunita, and Hsiao, Edward C
- Subjects
CANCELLOUS bone ,G protein coupled receptors ,PORCUPINES ,WNT signal transduction ,BONE growth ,BONE mechanics ,FEMUR - Abstract
G protein-coupled receptors (GPCRs) mediate a wide spectrum of physiological functions, including the development, remodeling, and repair of the skeleton. Fibrous dysplasia (FD) of the bone is characterized by fibrotic, expansile bone lesions caused by activating mutations in GNAS. There are no effective therapies for FD. We previously showed that ColI(2.3)
+ /Rs1+ mice, in which Gs -GPCR signaling was hyper-activated in osteoblastic cell lineages using an engineered receptor strategy, developed a fibrotic bone phenotype with trabecularization that could be reversed by normalizing Gs -GPCR signaling, suggesting that targeting the Gs -GPCR or components of the downstream signaling pathway could serve as a promising therapeutic strategy for FD. The Wnt signaling pathway has been implicated in the pathogenesis of FD-like bone, but the specific Wnts and which cells produce them remain largely unknown. Single-cell RNA sequencing on long-bone stromal cells of 9-wk-old male ColI(2.3)+ /Rs1+ mice and littermate controls showed that fibroblastic stromal cells in ColI(2.3)+ /Rs1+ mice were expanded. Multiple Wnt ligands were up- or downregulated in different cellular populations, including in non-osteoblastic cells. Treatment with the porcupine inhibitor LGK974, which blocks Wnt signaling broadly, induced partial resorption of the trabecular bone in the femurs of ColI(2.3)+ /Rs1+ mice, but no significant changes in the craniofacial skeleton. Bone fibrosis remained evident after treatment. Notably, LGK974 caused significant bone loss in control mice. These results provide new insights into the role of Wnt and Gs -signaling in fibrosis and bone formation in a mouse model of Gs -GPCR pathway overactivation. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
26. DNA methylation may affect beef tenderness through signal transduction in Bos indicus
- Author
-
de Souza, Marcela Maria, Niciura, Simone Cristina Méo, Rocha, Marina Ibelli Pereira, Pan, Zhangyuan, Zhou, Huaijun, Bruscadin, Jennifer Jessica, da Silva Diniz, Wellison Jarles, Afonso, Juliana, de Oliveira, Priscila Silva Neubern, Mourão, Gerson B, Zerlotini, Adhemar, Coutinho, Luiz Lehmann, Koltes, James E, and de Almeida Regitano, Luciana Correia
- Subjects
Biological Sciences ,Genetics ,Biotechnology ,1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Cattle ,CpG Islands ,DNA Methylation ,Meat ,Muscle ,Skeletal ,Signal Transduction ,Nelore ,RRBS ,GNAS ,EBF3 ,Shear force ,Epigenome ,Muscle ,Methylation - Abstract
BackgroundBeef tenderness is a complex trait of economic importance for the beef industry. Understanding the epigenetic mechanisms underlying this trait may help improve the accuracy of breeding programs. However, little is known about epigenetic effects on Bos taurus muscle and their implications in tenderness, and no studies have been conducted in Bos indicus.ResultsComparing methylation profile of Bos indicus skeletal muscle with contrasting beef tenderness at 14 days after slaughter, we identified differentially methylated cytosines and regions associated with this trait. Interestingly, muscle that became tender beef had higher levels of hypermethylation compared to the tough group. Enrichment analysis of predicted target genes suggested that differences in methylation between tender and tough beef may affect signal transduction pathways, among which G protein signaling was a key pathway. In addition, different methylation levels were found associated with expression levels of GNAS, PDE4B, EPCAM and EBF3 genes. The differentially methylated elements correlated with EBF3 and GNAS genes overlapped CpG islands and regulatory elements. GNAS, a complex imprinted gene, has a key role on G protein signaling pathways. Moreover, both G protein signaling pathway and the EBF3 gene regulate muscle homeostasis, relaxation, and muscle cell-specificity.ConclusionsWe present differentially methylated loci that may be of interest to decipher the epigenetic mechanisms affecting tenderness. Supported by the previous knowledge about regulatory elements and gene function, the methylation data suggests EBF3 and GNAS as potential candidate genes and G protein signaling as potential candidate pathway associated with beef tenderness via methylation.
- Published
- 2022
27. Targeted silencing of GNAS in a human model of osteoprogenitor cells results in the deregulation of the osteogenic differentiation program
- Author
-
Francesca Marta Elli, Deborah Mattinzoli, Masami Ikehata, Francesca Bagnaresi, Maria A. Maffini, Giulia Del Sindaco, Angela Pagnano, Camilla Lucca, Piergiorgio Messa, Maura Arosio, Giuseppe Castellano, Carlo M. Alfieri, and Giovanna Mantovani
- Subjects
GNAS ,ectopic bone ,mesenchymal stem cells ,osteogenesis ,human cell model ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
IntroductionThe dysregulation of cell fate toward osteoprecursor cells associated with most GNAS-based disorders may lead to episodic de novo extraskeletal or ectopic bone formation in subcutaneous tissues. The bony lesion distribution suggests the involvement of abnormal differentiation of mesenchymal stem cells (MSCs) and/or more committed precursor cells. Data from transgenic mice support the concept that GNAS is a crucial factor in regulating lineage switching between osteoblasts (OBs) and adipocyte fates. The mosaic nature of heterotopic bone lesions suggests that GNAS genetic defects provide a sensitized background for ectopic osteodifferentiation, but the underlying molecular mechanism remains largely unknown.MethodsThe effect of GNAS silencing in the presence and/or absence of osteoblastic stimuli was evaluated in the human L88/5 MSC line during osteodifferentiation. A comparison of the data obtained with data coming from a bony lesion from a GNAS-mutated patient was also provided.ResultsOur study adds some dowels to the current fragmented notions about the role of GNAS during osteoblastic differentiation, such as the premature transition of immature OBs into osteocytes and the characterization of the differences in the deposed bone matrix.ConclusionWe demonstrated that our cell model partially replicates the in vivo behavior results, resulting in an applicable human model to elucidate the pathophysiology of ectopic bone formation in GNAS-based disorders.
- Published
- 2024
- Full Text
- View/download PDF
28. DNA methylation landscape reveals GNAS as a decitabine-responsive marker in patients with acute myeloid leukemia
- Author
-
Shujiao He, Yan Li, Lei Wang, Yisheng Li, Lu Xu, Diya Cai, Jingfeng Zhou, and Li Yu
- Subjects
Acute myeloid leukemia ,Prognostic biomarker ,Decitabine ,Differentially methylated region ,DNA methylation sequencing ,GNAS ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: The demethylation agent decitabine (DAC) is a pivotal non-intensive alternative treatment for acute myeloid leukemia (AML). However, patient responses to DAC are highly variable, and predictive biomarkers are warranted. Herein, the DNA methylation landscape of patients treated with a DAC-based combination regimen was compared with that of patients treated with standard chemotherapy to develop a molecular approach for predicting clinical response to DAC. Methods: Twenty-five non-M3 AML patients were enrolled and subjected to DNA methylation sequencing and profiling to identify differentially methylated regions (DMRs) and genes of interest. Moreover, the effects of a DAC-based regimen on apoptosis and gene expression were explored using Kasumi-1 and K562 cells. Results: Overall, we identified 541 DMRs that were specifically responsive to DAC, among which 172 DMRs showed hypomethylation patterns upon treatment and were aligned with the promoter regions of 182 genes. In particular, GNAS was identified as a critical DAC-responsive gene, with in vitro GNAS downregulation leading to reduced cell apoptosis induced by DAC and cytarabine combo treatment. Conclusions: We found that GNAS is a DAC-sensitive gene in AML and may serve as a prognostic biomarker to assess the responsiveness of patients with AML to DAC-based therapy.
- Published
- 2024
- Full Text
- View/download PDF
29. Maternal GNAS Contributes to the Extra-Large G Protein α-Subunit (XLαs) Expression in a Cell Type-Specific Manner.
- Author
-
Qiuxia Cui, Aksu, Cagri, Ay, Birol, Remillard, Claire E., Plagge, Antonius, Gardezi, Mina, Dunlap, Margaret, Gerstenfeld, Louis C., Qing He, and Bastepe, Murat
- Subjects
G proteins ,MESENCHYMAL stem cells ,FIBROUS dysplasia of bone ,G protein coupled receptors ,BONE regeneration - Abstract
NAS encodes the stimulatory G protein alpha-subunit (Gsα) and its large variant XLαs. Studies have suggested that XLαs is expressed exclusively paternally. Thus, XLαs deficiency is considered to be responsible for certain findings in patients with paternal GNAS mutations, such as pseudo-pseudohypoparathyroidism, and the phenotypes associated with maternal uniparental disomy of chromosome 20, which comprises GNAS. However, a study of bone marrow stromal cells (BMSC) suggested that XLαs could be biallelically expressed. Aberrant BMSC differentiation due to constitutively activating GNAS mutations affecting both Gsα and XLαs is the underlying pathology in fibrous dysplasia of bone. To investigate allelic XLαs expression, we employed next-generation sequencing and a polymorphism common to XLαs and Gsα, as well as A/B, another paternally expressed GNAS transcript. In mouse BMSCs, Gsα transcripts were 48.4 ± 0.3% paternal, while A/B was 99.8 ± 0.2% paternal. In contrast, XLαs expression varied among different samples, paternal contribution ranging from 43.0 to 99.9%. Sample-to-sample variation in paternal XLαs expression was also detected in bone (83.7-99.6%) and cerebellum (83.8 to 100%) but not in cultured calvarial osteoblasts (99.1 ± 0.1%). Osteoblastic differentiation of BMSCs shifted the paternal XLαs expression from 83.9 ± 1.5% at baseline to 97.2 ± 1.1%. In two human BMSC samples grown under osteoinductive conditions, XLαs expression was also predominantly monoallelic (91.3 or 99.6%). Thus, the maternal GNAS contributes significantly to XLαs expression in BMSCs but not osteoblasts. Altered XLαs activity may thus occur in certain cell types irrespective of the parental origin of a GNAS defect. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. The mystery of transient pregnancy-induced cushing's syndrome: a case report and literature review highlighting GNAS somatic mutations and LHCGR overexpression.
- Author
-
Li, Yufei, Lin, Jianfan, Fu, Shien, Li, Li, Huang, Zhenxing, Yang, Haiyan, Liang, Xinghuan, Qin, Yingfen, Zhou, Jia, Liu, Deyun, and Luo, Zuojie
- Abstract
Purpose: Transient pregnancy-induced Cushing's syndrome is a rare condition characterized by the manifestation of symptoms solely during pregnancy, which typically resolve spontaneously following delivery or miscarriage. While it has been established that GNAS is associated with adrenal tumors, its specific role in the pathogenesis of pregnancy-induced Cushing's syndrome remains uncertain.This work aims to examine the association between GNAS mutation and pregnancy-induced Cushing's syndrome. Methods: DNA was extracted from patients' peripheral blood and tumor tissues for whole-exome sequencing (WES) and Sanger sequencing. We used AlphaFold to predict the protein structure of wild-type and mutant GNAS and to make functional predictions, and immunohistochemistry was used to detect disease-associated protein expression. A review and summary of reported cases of transient pregnancy-induced Cushing's syndrome induced by pregnancy was conducted. Results: Using WES, we identified a somatic mutation in GNAS (NM_000516, c.C601T, p.R201C) that was predicted to have a deleterious effect using computational methods, such as AlphaFold. Human chorionic gonadotropin (hCG) stimulation tests had weakly positive results, and immunohistochemical staining of adrenal adenoma tissue also revealed positivity for luteinizing hormone/chorionic gonadotropin receptor (LHCGR) and cytochrome P450 family 11 subfamily B member 1 (CYP11B1). We reviewed 15 published cases of transient Cushing's syndrome induced by pregnancy. Among these cases, immunohistochemical staining of the adrenal gland showed positive LHCGR expression in 3 case reports, similar to our findings. Conclusion: Transient pregnancy-induced Cushing's syndrome may be associated with somatic GNAS mutations and altered adrenal pathology due to abnormal activation of LHCGR. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Histopathological Spectrum and Molecular Characterization of Liver Tumors in the Setting of Fontan-Associated Liver Disease.
- Author
-
Francalanci, Paola, Giovannoni, Isabella, Tancredi, Chantal, Gagliardi, Maria Giulia, Palmieri, Rosalinda, Brancaccio, Gianluca, Spada, Marco, Maggiore, Giuseppe, Pietrobattista, Andrea, Monti, Lidia, Castellano, Aurora, Giustiniani, Maria Cristina, Onetti Muda, Andrea, and Alaggio, Rita
- Subjects
- *
LIVER tumors , *BIOPSY , *GENETIC mutation , *IMMUNOHISTOCHEMISTRY , *CHOLANGIOCARCINOMA , *MOLECULAR biology , *LIVER diseases , *RISK assessment , *HISTOLOGICAL techniques , *RESEARCH funding , *CARDIOPULMONARY bypass , *HEPATOCELLULAR carcinoma , *DISEASE risk factors - Abstract
Simple Summary: Fontan-associated liver disease (FALD) is a late sequela of single ventricle palliation. The long-term consequences of Fontan's physiology have a spectrum of outcomes resulting in chronic hepatic venous congestion, liver cirrhosis, hyperplastic nodules, and liver neoplasms that are both benign, such as hepatic adenoma (HA), and malignant, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC). To date, genetic and molecular studies of HCC/CC contextual to FALD are lacking. We report the molecular profile of the heterogeneous group of tumors observed in the FALDs (HA, HCC, and CC) we studied with the aim of comparing any differences with the same tumors without FALD and identifying possible recurrent variables, such as molecular alterations, that could be predictive of clinical outcome. Purpose: Univentricular heart is corrected with the Fontan procedure (FP). In the long term, so-called Fontan-associated liver diseases (FALDs) can develop. The aim of this study is to analyze the molecular profile of FALDs. Methods: FALDs between January 1990 and December 2022 were reviewed for histology and immunohistochemistry, laboratory data, and images. Targeted next generation sequencing (NGS), performed on the DNA and RNA of both neoplastic and non-lesional liver tissue, was applied. Results: A total of 31/208 nodules > 1 cm in diameter were identified on imaging, but a liver biopsy was available for five patient demonstrating the following: one hepatocellular adenoma (HA), two hepatocellular carcinomas (HCCs), one fibrolamellar carcinoma (FLC), and one intrahepatic cholangiocarcinoma (ICC). Molecular analysis showed a copy number alteration involving FGFR3 in three cases (two HCCs and one ICC) as well as one HCC with a hotspot mutation on the CTNNB1 and NRAS genes. Tumor mutational burden ranged from low to intermediate. A variant of uncertain significance in GNAS was present in two HCCs and in one ICC. The same molecular profile was observed in a non-lesional liver. A DNAJB1-PRKACA fusion was detected only in one FLC. Conclusions: Neoplastic FALDs show some unusual molecular profiles compared with non-Fontan ones. The presence of the same alterations in non-lesional cardiac cirrhosis could contribute to the development of FALD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Choosing the Best Tissue and Technique to Detect Mosaicism in Fibrous Dysplasia/McCune–Albright Syndrome (FD/MAS).
- Author
-
Vado, Yerai, Manero-Azua, Africa, Pereda, Arrate, and Perez de Nanclares, Guiomar
- Subjects
- *
MOSAICISM , *SOMATIC mutation , *CHOICE (Psychology) , *PRECOCIOUS puberty , *NUCLEOTIDE sequencing , *MACULES - Abstract
GNAS-activating somatic mutations give rise to Fibrous Dysplasia/McCune–Albright syndrome (FD/MAS). The low specificity of extra-skeletal signs of MAS and the mosaic status of the mutations generate some difficulties for a proper diagnosis. We studied the clinical and molecular statuses of 40 patients referred with a clinical suspicion of FD/MAS to provide some clues. GNAS was sequenced using both Sanger and Next-Generation Sequencing (NGS). We were able to identify the pathogenic variants in 25% of the patients. Most of them were identified in the affected tissue, but not in blood. Additionally, NGS demonstrated the ability to detect more patients with mosaicism (8/34) than Sanger sequencing (4/39). Even if in some cases, the clinical information was not complete, we confirmed that, as in previous works, when the patients were young children with a single manifestation, such as hyperpigmented skin macules or precocious puberty, the molecular diagnosis was usually negative. In conclusion, as FD/MAS is caused by mosaic variants, it is essential to use sensitive techniques that allow for the detection of low percentages and to choose the right tissue to study. When not possible, and due to the low positive genetic rate, patients with FD/MAS should only be genetically tested when the clinical diagnosis is really uncertain. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Pseudohypoparathyroidism: complex disease variants with unfortunate names.
- Author
-
Jüppner, Harald
- Subjects
- *
GENE expression , *G protein coupled receptors , *G proteins , *CHROMOSOMES , *EPIGENETICS , *PARATHYROID hormone - Abstract
Several human disorders are caused by genetic or epigenetic changes involving the GNAS locus on chromosome 20q13.3 that encodes the alpha-subunit of the stimulatory G protein (Gsa) and several splice variants thereof. Thus, pseudohypoparathyroidism type Ia (PHP1A) is caused by heterozygous inactivating mutations involving the maternal GNAS exons 1-13 resulting in characteristic abnormalities referred to as Albright's hereditary osteodystrophy (AHO) that are associated with resistance to several agonist ligands, particularly to parathyroid hormone (PTH), thereby leading to hypocalcemia and hyperphosphatemia. GNAS mutations involving the paternal Gsa exons also cause most of these AHO features, but without evidence for hormonal resistance, hence the term pseudopseudohypoparathyroidism (PPHP). Autosomal dominant pseudohypoparathyroidism type Ib (PHP1B) due to maternal GNAS or STX16 mutations (deletions, duplications, insertions, and inversions) is associated with epigenetic changes at one or several differentially methylated regions (DMRs) within GNAS. Unlike the inactivating Gsa mutations that cause PHP1A and PPHP, hormonal resistance is caused in all PHP1B variants by impaired Gsa expression due to loss of methylation at GNAS exon A/B, which can be associated in some familial cases with epigenetic changes at the other maternal GNAS DMRs. The genetic defect(s) responsible for sporadic PHP1B, the most frequent variant of this disorder, remain(s) unknown for the majority of patients. However, characteristic epigenetic GNAS changes can be readily detected that include a gain of methylation at the neuroendocrine secretory protein (NESP) DMR. Multiple genetic or epigenetic GNAS abnormalities can thus impair Gsa function or expression, consequently leading to inadequate cAMP-dependent signaling events downstream of various Gsa-coupled receptors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Intrafamilial phenotypic heterogeneity in siblings with pseudohypoparathyroidism 1B due to maternal STX16 deletion.
- Author
-
Odom, John, Bacino, Carlos A., Karaviti, Lefkothea P., Bi, Weimin, and Hoyos-Martinez, Alfonso
- Abstract
Pseudohypoparathyroidism (PHP1B) is most commonly caused by epigenetic defects resulting in loss of methylation at the GNAS locus, although deletions of STX16 leading to GNAS methylation abnormalities have been previously reported. The phenotype of this disorder is variable and can include hormonal resistances and severe infantile obesity with hyperphagia. A possible time relationship between the onset of obesity and endocrinopathies has been previously reported but remains unclear. Understanding of the condition's natural history is limited, partly due to a scarcity of literature, especially in children. We report three siblings with autosomal dominant PHP1B caused by a deletion in STX16 who presented with early childhood onset PTH-resistance with normocalcemia with a progressive nature, accompanied by TSH-resistance and severe infantile obesity with hyperphagia in some, not all of the affected individuals. PHP1B from a STX16 deletion displays intrafamilial phenotypic variation. It is a novel cause of severe infantile obesity, which is not typically included in commercially available gene panels but must be considered in the genetic work-up. Finally, it does not seem to have a clear time relationship between the onset of obesity and hormonal resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. (Epi)genetic and clinical characteristics in 84 patients with pseudohypoparathyroidism type 1B.
- Author
-
Tatsuki Urakawa, Shinichiro Sano, Sayaka Kawashima, Akie Nakamura, Hirohito Shima, Motoki Ohta, Yuki Yamada, Ai Nishida, Hiromune Narusawa, Yoshiaki Ohtsu, Keiko Matsubara, Sumito Dateki, Yoshihiro Maruo, Maki Fukami, Tsutomu Ogata, and Masayo Kagami
- Subjects
- *
EPIGENETICS , *HYPOPARATHYROIDISM , *MICROSATELLITE repeats - Abstract
Objective: Pseudohypoparathyroidism type 1B (PHP1B) caused by methylation defects of differentially methylated regions (DMRs) on the GNAS locus can be categorized into groups according to etiologies and methylation defect patterns of the DMRs. The aim of this study was to clarify the clinical characteristics of each group. Design: Comprehensive molecular analyses consisting of methylation, copy number, and microsatellite analyses. Methods: Eighty-four patients with PHP1B were included in this study. We classified them into 5 groups, namely, autosomal dominant inheritance-PHP1B (Group 1, G1), sporadic-PHP1B (G2), and atypical-PHP1B (G3-G5), based on the methylation defect patterns in 4 DMRs on the GNAS locus and etiologies and evaluated the clinical findings in each group and compared them among the groups. Results: G2 had the youngest age and the highest serum intact parathyroid hormone levels among the 5 groups at the time of diagnosis. The most common symptoms at the time of diagnosis were tetany in G1, and seizures or loss of consciousness in G2. Albright's hereditary osteodystrophy and PHP-suggestive features were most frequently observed in the G2 proband. Nine patients had neurodevelopmental disorders (NDs) consisting of mild to borderline intellectual disability and/or developmental delay. There were no significant correlations between the average methylation ratios of 7 CpG sites in the GNAS-A/B:TSS-DMR and hormonal and biochemical findings. Conclusion: This study revealed the differences in some clinical characteristics, particularly clinical features, and ages at the time of diagnosis between G2 and other groups and detailed NDs observed in some patients with PHP1B. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
36. Editorial: Parathyroid disorders: updates of PTH/serum Ca2+ regulation and therapeutic prospects
- Author
-
Fan Zhang, Yinde Huang, Jiongyu Hu, and Supeng Yin
- Subjects
parathyroid ,parathyroid hormone ,hypoparathyroidism ,hyperparathyroidism ,GNAS ,blastocyst complementation ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Published
- 2024
- Full Text
- View/download PDF
37. Erratum: Maternal GNAS contributes to the extra-large G protein α-subunit (XLαs) expression in a cell type-specific manner
- Author
-
Frontiers Production Office
- Subjects
GNAS ,stimulatory G protein ,imprinting ,osteoblasts ,bone marrow stromal cells ,fibrous dysplasia of bone ,Genetics ,QH426-470 - Published
- 2024
- Full Text
- View/download PDF
38. Genetic characterization of intramuscular myxomas
- Author
-
William John Hatchett, Marta Brunetti, Kristin Andersen, Maren Randi Tandsæther, Ingvild Lobmaier, Marius Lund-Iversen, Thomas Lien-Dahl, Francesca Micci, and Ioannis Panagopoulos
- Subjects
intramuscular myxoma ,chromosomal aberrations ,GNAS ,codon 201 ,codon 227 ,Sanger sequencing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Pathology ,RB1-214 - Abstract
Introduction: Intramuscular myxomas are benign tumors that are challenging to diagnose, especially on core needle biopsies. Acquired chromosomal aberrations and pathogenic variants in codon 201 or codon 227 in GNAS complex locus gene (GNAS) have been reported in these tumors. Here we present our genetic findings in a series of 22 intramuscular myxomas.Materials and methods: The tumors were investigated for the presence of acquired chromosomal aberrations using G-banding and karyotyping. Pathogenic variants in codon 201 or codon 227 of GNAS were assessed using direct cycle Sanger sequencing and Ion AmpliSeq Cancer Hotspot Panel v2 methodologies.Results: Eleven tumors carried chromosomal abnormalities. Six tumors had numerical, four had structural, and one had both numerical and structural chromosomal aberrations. Gains of chromosomes 7 and 8 were the most common abnormalities being found in five and four tumors respectively. Pathogenic variants in GNAS were detected in 19 myxomas (86%) with both methodologies. The detected pathogenic variants were p.R201H in nine cases (seven with abnormal and two with normal karyotypes), p.R201C in five cases, all with normal karyotypes, p.R201S in three cases (two with abnormal and one with normal karyotype), p.R201G in one case with a normal karyotype, and p.Q227E in one case with a normal karyotype.Conclusion: Firstly, our data indicate a possible association between chromosomal abnormalities and GNAS pathogenic variants in intramuscular myxomas. Secondly, the presence of the rare pathogenic variants R201S, p.R201G and p.Q227E in 26% (5 out of 19) of myxomas with GNAS pathogenic variants shows that methodologies designed to detect only the common “hotspot” of p.R201C and p.R201H will give false negative results. Finally, a comparison between Ion AmpliSeq Cancer Hotspot Panel v2 and direct cycle Sanger sequencing showed that direct cycle Sanger sequencing provides a quick, reliable, and relatively cheap method to detect GNAS pathogenic variants, matching even the most cutting-edge sequencing methods.
- Published
- 2024
- Full Text
- View/download PDF
39. Genotype-phenotype correlations in pseudohypoparathyroidism type 1a patients: a systemic review.
- Author
-
Siqi Jiang, Yi Yang, An Song, Yue Jiang, Yan Jiang, Mei Li, Weibo Xia, Min Nie, Ou Wang, and Xiaoping Xing
- Subjects
- *
GENOTYPES , *PSEUDOHYPOPARATHYROIDISM - Abstract
Background: Pseudohypoparathyroidism type 1a (PHP1a) is a rare endocrine disease caused by partial defects of the α subunit of the stimulatory Guanosin triphosphate (GTP) binding protein (Gsα) resulting from maternal GNAS gene variation. The clinical manifestations are related to PTH resistance (hypocalcemia, hyperphosphatemia, and elevated serum intact PTH) in the presence or absence of multihormone resistance, and Albright’s hereditary osteodystrophy (AHO). Objectives: To summarize the molecular genetics results and clinical characteristics as well as to explore the correlations between them. Methods: Articles pertaining to PHP1a until May, 31, 2021 were reviewed and 527 patients with genetic diagnosis were included in the data analysis. The clinical characteristics and molecular genetics results of these patients were analyzed and compared to explore the correlations between them. Results: A total of 258 GNAS rare variants (RVs) were identified in 527 patients. The RVs were most commonly found in exons 1 and 7 (17.6% each), with frameshift (36.8%), and missense (31.3%) being the main types of RVs. The median age of onset was 5.0 years old. The most common clinical manifestations were elevation of PTH (86.7%) and AHO (87.5%). Thyroid stimulating hormone resistance was the most common hormone resistance (75.5%) other than PTH resistance. Patients with missense and in-frame RVs had lower incidence rates of the round face (P = .001) and subcutaneous ossifications (P < .001) than those with loss-of-function (non-sense, frameshift, splicing site variants, and large deletions) variants. Conclusions: This study revealed the correlation between loss-of-function RVs with round faces and subcutaneous ossifications in PHP 1a patients. Further exploration of genotype-phenotype correlations through more standardized and prospective studies with long-term follow-up is necessary. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. The role and mechanism of circ‐BNC2 on the malignant progression of oral squamous cell carcinoma.
- Author
-
Xia, Yingjie, Hei, Naiheng, Peng, Shixiong, and Cui, Zifeng
- Subjects
GENE expression ,SQUAMOUS cell carcinoma ,LIPID peroxidation (Biology) ,CELL death ,CIRCULAR RNA ,REACTIVE oxygen species ,POLYMERASE chain reaction ,SUPEROXIDES - Abstract
Background: Circular RNAs (circRNAs) play a key part in the progression of oral squamous cell carcinoma (OSCC). However, the role of circ‐BNC2 (circRNA ID hsa_circ_0086414) in OSCC progression is still unclear. Methods: Plasmid transfection was used to induce overexpression of circ‐BNC2. RNA expression of circ‐BNC2, microRNA‐142‐3p (miR‐142‐3p) and GNAS complex locus (GNAS) was detected by quantitative real‐time polymerase chain reaction. Protein expression was assessed by western blot assay or immunohistochemistry assay. Cell proliferation was investigated by 3‐(4,5‐dimethylthazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide (MTT) assay, colony formation assay and flow cytometry analysis. Cell migratory and invasive abilities and cell apoptosis were assessed by transwell assay and flow cytometry analysis, respectively. Oxidative stress was evaluated by superoxide dismutase activity detection assay, lipid peroxidation malondialdehyde assay and cellular reactive oxygen species assay. The binding relationship between miR‐142‐3p and circ‐BNC2 or GNAS was proved by dual‐luciferase reporter assay and RNA immunoprecipitation assay. The impacts of circ‐BNC2 overexpression on tumor growth in vivo were unveiled by a xenograft mouse model assay. Results: Circ‐BNC2 expression was downregulated in OSCC tissues and cells when compared with adjacent healthy tissues and normal human oral keratinocytes. Circ‐BNC2 overexpression repressed the proliferation, migration and invasion of OSCC cells but induced cell apoptosis and oxidative stress. Additionally, circ‐BNC2 overexpression inhibited tumor growth in vivo. Furthermore, circ‐BNC2 bound to miR‐142‐3p, and miR‐142‐3p targeted GNAS. MiR‐142‐3p mimic attenuated circ‐BNC2 overexpression‐mediated effects on the proliferation, migration, invasion, apoptosis and oxidative stress of OSCC cells. The regulation of miR‐142‐3p in OSCC cell tumor properties involved GNAS. Further, circ‐BNC2 introduction promoted GNAS expression by inhibiting miR‐142‐3p. Conclusion: Circ‐BNC2 suppressed OSCC malignant progression by upregulating GNAS expression in a miR‐142‐3p‐dependent manner, which suggested that circ‐BNC2 might be a novel target for OSCC therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. Unbiased Proteomic Profiling Uncovers a Targetable GNAS/PKA/PP2A Axis in Small Cell Lung Cancer Stem Cells
- Author
-
Coles, Garry L, Cristea, Sandra, Webber, James T, Levin, Rebecca S, Moss, Steven M, He, Andy, Sangodkar, Jaya, Hwang, Yeonjoo C, Arand, Julia, Drainas, Alexandros P, Mooney, Nancie A, Demeter, Janos, Spradlin, Jessica N, Mauch, Brandon, Le, Vicky, Shue, Yan Ting, Ko, Julie H, Lee, Myung Chang, Kong, Christina, Nomura, Daniel K, Ohlmeyer, Michael, Swaney, Danielle L, Krogan, Nevan J, Jackson, Peter K, Narla, Goutham, Gordan, John D, Shokat, Kevan M, and Sage, Julien
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Stem Cell Research - Nonembryonic - Human ,Cancer ,Stem Cell Research - Nonembryonic - Non-Human ,Lung Cancer ,Rare Diseases ,Lung ,Biotechnology ,Stem Cell Research ,Women's Health ,2.1 Biological and endogenous factors ,A549 Cells ,Animals ,Antineoplastic Agents ,Cell Line ,Tumor ,Chromogranins ,Cisplatin ,Cyclic AMP-Dependent Protein Kinases ,GTP-Binding Protein alpha Subunits ,Gs ,Humans ,Lung Neoplasms ,Mice ,Inbred NOD ,Mice ,Knockout ,Mice ,SCID ,Neoplastic Stem Cells ,Protein Phosphatase 2 ,Proteomics ,Signal Transduction ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,GNAS ,PKA ,PP2A ,SCLC ,cancer ,cancer stem cells ,kinase ,lung ,neuroendocrine ,phosphatase ,proteomics ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Using unbiased kinase profiling, we identified protein kinase A (PKA) as an active kinase in small cell lung cancer (SCLC). Inhibition of PKA activity genetically, or pharmacologically by activation of the PP2A phosphatase, suppresses SCLC expansion in culture and in vivo. Conversely, GNAS (G-protein α subunit), a PKA activator that is genetically activated in a small subset of human SCLC, promotes SCLC development. Phosphoproteomic analyses identified many PKA substrates and mechanisms of action. In particular, PKA activity is required for the propagation of SCLC stem cells in transplantation studies. Broad proteomic analysis of recalcitrant cancers has the potential to uncover targetable signaling networks, such as the GNAS/PKA/PP2A axis in SCLC.
- Published
- 2020
42. Adenosquamous carcinoma coexisting with intraductal papillary mucinous neoplasm of the pancreas: a case report
- Author
-
Hirozumi Sawai, Yuka Kiriyama, Hiromasa Kuzuya, Yoshiaki Fujii, Shuhei Ueno, Shuji Koide, Masaaki Kurimoto, Kenji Yamao, Yoichi Matsuo, Mamoru Morimoto, Hajime Koide, and Atsushi Kamiya
- Subjects
Adenosquamous carcinoma ,Intraductal papillary mucinous neoplasm ,Pancreas ,KRAS ,GNAS ,Medicine - Abstract
Abstract Background Adenosquamous carcinoma of the pancreas is a rare variant, with a worse prognosis than pancreatic ductal adenocarcinoma; moreover, it has characteristic clinical and histopathological features. Studies have mentioned the differentiation of intraductal papillary mucinous neoplasms into mucinous/tubular adenocarcinomas; however, their transdifferentiation into adenosquamous carcinoma remains unclear. Case presentation An 80-year-old Japanese woman was referred to our hospital for further examination of multiple pancreatic cysts. Enhanced computed tomography after close follow-up for 6 years revealed a new nodule with poor enhancement on the pancreatic body. Distal pancreatectomy and splenectomy were performed. Histopathological examination revealed an adenosquamous carcinoma with coexisting intraductal papillary mucinous neoplasms; moreover, the intraductal papillary mucinous neoplasms lacked continuity with the adenosquamous carcinoma. Immunohistochemical analysis revealed squamous cell carcinoma and differentiation from adenocarcinoma to squamous cell carcinoma. Gene mutation analysis revealed KRAS G12D and KRAS G12R mutations in adenosquamous carcinoma components and intraductal papillary mucinous neoplasm lesions, respectively, with none showing the mutation of GNAS codon 201. The final histopathological diagnosis was adenosquamous carcinoma with coexisting intraductal papillary mucinous neoplasms of the pancreas. Conclusions This is the rare case of adenosquamous carcinoma with coexisting intraductal papillary mucinous neoplasms of the pancreas. To investigate the underlying transdifferentiation pathway of intraductal papillary mucinous neoplasms into this rare subtype of pancreatic cancer, we explored gene mutation differences as a clinicopathological parameter.
- Published
- 2023
- Full Text
- View/download PDF
43. Intraductal oncocytic papillary neoplasm arising in Peutz-Jeghers Syndrome bile duct: a unique case report
- Author
-
Qingyue Liu, Zhiyu Wang, Chaoran Yu, Jianping Zhu, Chengli Liu, Xiangsheng Li, Li Ren, and Teng Li
- Subjects
Peutz-Jeghers syndrome ,Intraductal oncocytic papillary neoplasm ,GNAS ,STK11 ,EGFR ,Pathology ,RB1-214 - Abstract
Abstract Background Peutz-Jeghers syndrome (PJS) is a rare, autosomal dominant disorder caused by germline mutations of STK11/LKB1, with an increased risk of tumors at multiple sites. Intraductal oncocytic papillary neoplasm (IOPN) is a unique subtype of intraductal papillary neoplasm of the bile duct (IPNB) defined by a premalignant neoplasm with intraductal papillary or villous growth of biliary-type epithelium. IOPN has a distinct mutation profile compared with both IPNB and intraductal papillary mucinous neoplasm (IPMN). Case presentation We herein describe the case of a 44-year-old woman who presented as polyps in the intestinal lumen of sigmoid colon and a 3.1 × 2.1 cm mass in the left lobe of liver. Gross feature revealed a cystic papillary mass and the neoplasm had a clear boundary with the surrounding liver tissue. Histology revealed complex papillary structures, a small amount of fine fibrovascular cores and immunohistochemistry showed extensive positive for MUC5AC, MUC6, CD117. Therefore, histological and immunohistochemical examination of the liver tumor suggested the diagnosis of IOPN. Next-generation sequencing (NGS) revealed other than STK11 germline mutation, the tumor also harbors GNAS somatic mutation at codon 478 and EGFR amplification. Conclusion To our knowledge, this is the first report of IOPN arising in PJS. This case enlarges the spectrum of PJS related tumors and genetic rearrangements in IOPN.
- Published
- 2022
- Full Text
- View/download PDF
44. Neonatal cholestasis as the onset symptom of McCune–Albright syndrome: case reports and a literature review
- Author
-
Weiyuan Fang, Yanhui Zhang, Lian Chen, and Xinbao Xie
- Subjects
McCune–Albright syndrome (MAS) ,neonatal cholestasis ,GNAS ,liver histology ,liver ,Pediatrics ,RJ1-570 - Abstract
AimThis study aimed to summarize and show the characteristics and evolutionary process of neonatal cholestasis caused by McCune–Albright syndrome (MAS), as neonatal cholestasis may be the initial manifestation of MAS before other classic clinical features appear.MethodsThe clinical characteristics, treatment methods, and outcomes of three neonatal cholestasis cases caused by MAS in our center were retrospectively studied. In addition, all the reported cases of MAS combined with cholestasis were reviewed and summarized to show the cholestatic features in them.ResultsWe have confirmed three MAS cases in our center, presenting onset symptoms of jaundice, pale stool, and neonatal cholestasis soon after birth. The cholestasis subsided spontaneously at around the sixth month. The literature review showed that the levels of total bilirubin, conjugated bilirubin, ALT, AST, and GGT in neonatal MAS cholestasis cases were 207 μmol/L (range 65–445 μmol/L), 162 μmol/L (range 46–412 μmol/L), 821 U/L (range 85–3,597 U/L), 532 U/L (range 127–3,633 U/L), and 244 U/L (range 79–3,800 U/L), respectively. Liver histology showed canalicular and hepatocellular cholestasis, giant hepatic cell transformation, and bile paucity. Extrahepatic manifestations such as café-au-lait pigmented skin lesions, Cushing's syndrome, hyperthyroidism, renal tubular dysfunction, and skeletal abnormalities could occur simultaneously when jaundice occurred. GNAS mutations had a high positive rate (83.3%–100%) in liver tissue with cholestasis. Neonatal cholestasis caused by MAS could be self-resolved, but hepatic lesions persist and have malignant potential.ConclusionMAS can be one of the causes of neonatal cholestasis, which may be the first manifestation of the disease. Extrahepatic coexisting symptoms of MAS and liver histology can help to distinguish MAS from other etiology of cholestasis. Detecting GNAS mutations in liver tissue may shorten diagnostic time and is of particular interest in the partial and atypical forms of MAS with neonatal cholestasis. Neonatal cholestasis in children with MAS can self-resolve, but liver dysfunction and malignant lesions persist.
- Published
- 2023
- Full Text
- View/download PDF
45. Brain and eye involvement in McCune-Albright Syndrome: clinical and translational insights.
- Author
-
Mascioli, Ilaria, Iapadre, Giulia, Ingrosso, Diletta, Di Donato, Giulio, Giannini, Cosimo, Salpietro, Vincenzo, Chiarelli, Francesco, and Farello, Giovanni
- Subjects
FIBROUS dysplasia of bone ,CYCLIC adenylic acid ,ARNOLD-Chiari deformity ,CENTRAL nervous system ,GENETIC disorders ,PRECOCIOUS puberty - Abstract
McCune-Albright Syndrome (MAS) is a rare mosaic (post-zygotic) genetic disorder presenting with a broad continuum clinical spectrum. MAS arises from somatic, activating mutations in the GNAS gene, which induces a dysregulated Gsa-protein signaling in several tissues and an increased production of intracellular cyclic adenosine monophosphate (cAMP). Overall, MAS is a rare disorder affecting less than 1/100,000 children and, for this reason, data establishing genotype-phenotype correlations remain limited. Affected individuals clinically present with a variable combination of fibrous dysplasia of bone (FD), extra-skeletal manifestations (including cafeı-' au-lait spots) and precocious puberty which might also be associated to broad hyperfunctioning endocrinopathies, and also gastrointestinal and cardiological involvement. Central nervous system (CNS) and eye involvement in MAS are among the less frequently described complications and remain largely uncharacterized. These rare complications mainly include neurodevelopmental abnormalities (e.g., delayed motor development, cognitive and language impairment), CNS anomalies (e.g., Chiari malformation type I) and a wide array of ophthalmological abnormalities often associated with vision loss. The pathophysiological mechanisms underlying abnormal neurological development have not been yet fully elucidated. The proposed mechanisms include a deleterious impact of chronically dysregulated Gsa-protein signaling on neurological function, or a secondary (damaging) effect of (antenatal and/or early postnatal) hypercortisolism on early pre- and post-natal CNS development. In this Review, we summarize the main neurological and ophthalmological features eventually associated with the MAS spectrum, also providing a detailed overview of the potential pathophysiological mechanisms underlying these clinical complications. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. Intraductal Neoplasms of the Pancreas
- Author
-
Furukawa, Toru, Makuuchi, Masatoshi, editor, Kokudo, Norihiro, editor, Popescu, Irinel, editor, Belghiti, Jacques, editor, Han, Ho-Seong, editor, Takaori, Kyoichi, editor, and Duda, Dan G., editor
- Published
- 2022
- Full Text
- View/download PDF
47. Pathogenesis of Pituitary Adenomas
- Author
-
Tang, Sicheng, Albani, Adriana, Theodoropoulou, Marily, Tamagno, Gianluca, editor, and Gahete, Manuel D., editor
- Published
- 2022
- Full Text
- View/download PDF
48. A recurrent somatic missense mutation in GNAS gene identified in familial thyroid follicular cell carcinomas in German longhaired pointer dogs
- Author
-
Yun Yu, Freek Manders, Guy C. M. Grinwis, Martien A. M. Groenen, and Richard P. M. A. Crooijmans
- Subjects
Familial cancer ,Thyroid carcinoma ,Driver mutation ,Dog ,GNAS ,Mutational signature ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background We previously reported a familial thyroid follicular cell carcinoma (FCC) in a large number of Dutch German longhaired pointers and identified two deleterious germline mutations in the TPO gene associated with disease predisposition. However, the somatic mutation profile of the FCC in dogs has not been investigated at a genome-wide scale. Results Herein, we comprehensively investigated the somatic mutations that potentially contribute to the inherited tumor formation and progression using high depth whole-genome sequencing. A GNAS p.A204D missense mutation was identified in 4 out of 7 FCC tumors by whole-genome sequencing and in 20 out of 32 dogs’ tumors by targeted sequencing. In contrast to this, in the human TC, mutations in GNAS gene have lower prevalence. Meanwhile, the homologous somatic mutation in humans has not been reported. These findings suggest a difference in the somatic mutation landscape between TC in these dogs and human TC. Moreover, tumors with the GNAS p.A204D mutation had a significantly lower somatic mutation burden in these dogs. Somatic structural variant and copy number alterations were also investigated, but no potential driver event was identified. Conclusion This study provides novel insight in the molecular mechanism of thyroid carcinoma development in dogs. German longhaired pointers carrying GNAS mutations in the tumor may be used as a disease model for the development and testing of novel therapies to kill the tumor with somatic mutations in the GNAS gene.
- Published
- 2022
- Full Text
- View/download PDF
49. Progressive osseous heteroplasia: A case report with an unexpected trigger
- Author
-
Alessandra Boncompagni, Angela K. Lucas-Herald, Paula Beattie, Helen McDevitt, Lorenzo Iughetti, Panayiotis Constantinou, Esther Kinning, S. Faisal Ahmed, and Avril Mason
- Subjects
Ossification ,Calcium ,GNAS ,Inflammation ,Extravasation ,Rare ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Progressive osseous heteroplasia (POH) is a rare genetic disorder characterised by progressive heterotopic ossification (HO) within the skin and subcutaneous tissues. The condition is caused by heterozygous inactivating mutations of the GNAS gene and usually presents in infancy. We describe the case of a white male ex-preterm who was first referred because of subcutaneous calcium deposits along the right arm after extravasation of parenteral nutrition. As these lesions progressed, a skin biopsy was undertaken which revealed intramembranous ossification. Genetic testing revealed a constitutional, de novo, heterozygous, nonsense variant in the GNAS gene that has not previously been described, but which is consistent with patient's clinical diagnosis of POH. No endocrine abnormalities or other signs congruent with overlapping conditions were detected. To the best of our knowledge, this is the first case describing an inflammatory trigger in POH. Trials with intravenous bisphosphonate and glucocorticoid as well as with topical sodium thiosulphate were attempted without clinical improvement. Excision of the calcifications and physiotherapy seem to have provided a partial improvement on mobility of the elbow. This case widens the spectrum of phenotypes seen in GNAS mutation disorders and suggests that alternative anti-inflammatory treatments may be effective. Mutations in GNAS should be considered in cases of significant progressive calcium deposition after extravasation injury.
- Published
- 2023
- Full Text
- View/download PDF
50. Whole‐genome sequencing revealed a novel long‐range deletion mutation spanning GNAS in familial pseudohypoparathyroidism.
- Author
-
Fei, Yangfan, Liu, Lv, Wu, Lixia, Wang, Ou, Xing, Xiaoping, Li, Aiping, and Huang, Lingyi
- Subjects
- *
DELETION mutation , *PATHOLOGICAL physiology , *CHROMOSOMES , *CELLULAR signal transduction , *MOTHERS , *NUCLEOTIDE sequencing - Abstract
Background: Pseudohypoparathyroidism (PHP) is a series of diseases related to pathological changes and neurocognitive and endocrine abnormalities, mainly due to the GNAS mutation on chromosome 20q13.2, which weakens receptor‐mediated hormone signal transduction. Considering its complex genetic and epigenetic characteristics, GNAS may produce complex clinical phenotypes in families or sporadic cases. This study presented a case of familial PHP caused by a deletion mutation in the 20q13.2 region. Methods and Results: The proband and her second daughter had PHP, and the proband's mother had pseudo‐PHP. Whole‐genome sequencing revealed that the proband had an 849.81 kb deletion spanning GNAS near the maternal 20q13.2 chromosome. Multiplex ligation‐dependent probe amplification methylation analysis indicated that the proband as well as her mother and second daughter had seemingly abnormal GNAS methylation. This is different from the phenotype (feeding difficulty, slow growth, and special facial features) of previously reported cases with the deletion of fragments near the 20q13.2 chromosome. Conclusions: This report demonstrated the variability of 20q13.2 deletion phenotypes and the clinical importance of using multiple molecular genetic detection methods. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.