scholarly journals Somatic uniparental disomy mitigates the most damaging EFL1 allele combination in Shwachman-Diamond syndrome

Blood ◽  
2021 ◽  
Author(s):  
Sangmoon Lee ◽  
Chang Hoon Shin ◽  
Jawon Lee ◽  
Seong Dong Jeong ◽  
Che Ry Hong ◽  
...  

Shwachman-Diamond syndrome (SDS; OMIM: #260400) is caused by variants in SBDS (Shwachman-Bodian-Diamond syndrome gene), which encodes a protein that plays an important role in ribosome assembly. Recent reports suggest that recessive variants in EFL1 are also responsible for SDS. However, the precise genetic mechanism that leads to EFL1-induced SDS remains incompletely understood. Here we present three unrelated Korean SDS patients that carry biallelic pathogenic variants in EFL1 with biased allele frequencies, resulting from a bone marrow-specific somatic uniparental disomy (UPD) in chromosome 15. The recombination events generated cells that were homozygous for the relatively milder variant, allowing for the evasion of catastrophic physiological consequences. Still, the milder EFL1 variant was solely able to impair 80S ribosome assembly and induce SDS features in cell line and animal models. The loss of EFL1 resulted in a pronounced inhibition of terminal oligo-pyrimidine element-containing ribosomal protein transcript 80S assembly. Therefore, we propose a more accurate pathogenesis mechanism of EFL1 dysfunction that eventually leads to aberrant translational control and ribosomopathy.

2018 ◽  
Author(s):  
Sangmoon Lee ◽  
Chang Hoon Shin ◽  
Che Ry Hong ◽  
Jun-Dae Kim ◽  
Ah-Ra Kim ◽  
...  

We present three unrelated Korean Shwachman-Diamond syndrome (SDS) patients that carry an incomplete but identical homozygous EFL1 p.Thr1069Ala variant due to a bone marrow-specific mosaic uniparental disomy (UPD) in chromosome 15. This rare variant is found in 0.017% of East Asians and is asymptomatic in a heterozygous status, but harbors a hypomorphic effect, leading to 80S assembly of ribosomal protein (RP) transcripts. We propose a novel somatically-induced pathogenesis mechanism and EFL1 dysfunction that eventually leads to aberrant translational control and ribosomopathy.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3618-3618
Author(s):  
Nimrat Chatterjee ◽  
Christopher Lee Williams ◽  
Saleh Bhar ◽  
Alison A Bertuch

Abstract Shwachman-Diamond syndrome (SDS), an autosomal recessive disorder, is characterized by bone marrow dysfunction, exocrine pancreatic insufficiency, congenital abnormalities, and leukemia predisposition (Myers et al., 2012). Most patients with SDS harbor biallelic mutations in the Shwachman-Bodian-Diamond syndrome (SBDS) gene. SBDS is known to play a role in ribosome biogenesis by enabling eviction of the ribosome anti-association factor eIF6 from the 60S ribosomal subunit, to allow formation of the 80S ribosome (Wong et al., 2011). SBDS-depleted cells are, therefore, defective in ribosome assembly. In addition, absence of SBDS sensitizes cells to ultraviolet irradiation, translation inhibitors, and endoplasmic reticulum (ER) stressors, such as tunicamycin (Ball et al., 2009). A recent report indicated that lymphoblastoid cell lines (LCLs) derived from two SDS patients accumulated more DNA damage after being exposed to ionizing radiation (IR) (Morini et al., 2015). A deficiency in DNA repair was alluded to as a possible cause, however, the mechanism underlying this previously unreported phenotype was not determined. In this study, we investigated LCLs derived from five SDS patients with biallelic SBDS mutations and found all to be hypersensitive to IR in a colony survival assay. In this assay, increasing doses of IR resulted in a significantly lower survival fraction in SDS-compared to control-LCLs. We found SBDS expression to increase in control-cells when stressed with IR, suggesting that SBDS is a stress response protein and its absence in SDS-LCLs induces hypersensitivity to IR. Because knockdown of SBDS in HEK293 cells induces an ER stress response (Ball et al., 2009), we examined the expression of the ER stress response factor phospho-eIF2α in untreated and IR exposed SDS-LCLs and found phospho-eIF2α expression to be markedly increased compared to controls. This result indicated that SDS-LCLs may have an activated ER stress response, as was further confirmed by exposing these cells to additional ER stressors, tunicamycin and H2O2, and observing a similar upregulation of phospho-eIF2α. Because ER stress is known to suppress DNA double strand break (DSBR) (Yamamori et al., 2013), we examined the expression of Rad51 and Ku70, which are required for the homology-directed and nonhomologous end-joining pathways of DSBR, respectively. Surprisingly, we found Rad51 and Ku70 protein levels to be repressed in SDS-LCLs compared to controls, both with and without exposure to IR. Collectively, these data support the hypothesis that, in addition to its role in ribosome biogenesis, SBDS is a stress response protein that plays an important role in regulating the ER stress response. In SDS-cells, where SBDS is lacking, activated ER stress represses DNA repair proteins rendering cells hypersensitive to IR and other stresses. This novel pathway to ER stress induction may contribute to the bone marrow failure and cancer predisposition seen in SDS patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2586-2586
Author(s):  
Lisa J. McReynolds ◽  
Kristine Jones ◽  
Kedest Teshome ◽  
Alyssa Kennedy ◽  
Akiko Shimamura ◽  
...  

Abstract Shwachman-Diamond syndrome (SDS) is an inherited bone marrow failure syndrome (IBMFS) with gastrointestinal manifestations (pancreatic insufficiency) and cytopenias, primarily neutropenia. Skeletal dysplasias and short stature are frequent. Patients with SDS are at significantly increased risk of myelodysplastic syndrome and acute myeloid leukemia. More than 90% of patients have autosomal recessive inheritance of germline pathogenic variants in SBDS, a ribosome biogenesis gene. The most common variants are c.258+2T>C (disrupts the donor splice site of intron 2, leading to an 8bp deletion and a premature protein truncation due to a frameshift); and c.183_184TA>CT (introduces an in-frame stop codon). To date, the vast majority of pathogenic alleles reported have been single nucleotide variants (SNVs) or small insertions/deletions. Rare cases have been reported with SBDS exon 3 deletions. The National Cancer Institute's IBMFS study is a longitudinal cohort study with 521 families enrolled, including 54 SDS or SDS-like families. Through clinical testing or whole exome sequencing all but nine families have had their disease-causing alleles identified. Three of the nine families had a known single pathogenic variant in SBDS. Array comparative genomic hybridization (aCGH) was uninformative in all but one. We initially focused on the family with a known SBDS c.258+2T>C and potential deletion on aCGH. The 27 year-old (yo) male proband was diagnosed with SDS at 5yo due to a history of malabsorption, requiring pancreatic enzyme supplementation, failure to thrive (FTT), short stature and neutropenia. He also has metaphyseal dysplasia, scoliosis, psychomotor retardation and learning disability. A bone marrow (BM) biopsy at 21yo showed hypoplasia with mild dysplastic changes and a del20q clone. aCGH showed a possible large deletion, but the resolution was too low to determine exact breakpoints. Therefore, long-range Single Molecule, Real-Time (SMRT) Sequencing (PacBio Systems) was undertaken. This identified a read spanning a 19kb deletion with exact coordinates (hg19: chr7:66,436,397-66,455,294), and was confirmed with polymerase chain reaction and Sanger sequencing. This deletion removes part of intron 4, all of exon 5 and the 3'UTR of SBDS. Western blotting for SBDS showed significant decrease in protein production from cultured fibroblasts. This deletion in combination with the known SBDS variant and western blot is consistent with the SDS phenotype in this patient. We then designed a targeted SMRT sequencing panel consisting of the four published SDS genes: SBDS, EFL1, DNAJC21 and SRP54 to evaluate the remaining families. The second proband evaluated was a 19yo female heterozygous for the SBDS c.258+2T>C variant. She had a history of malabsorption requiring enzyme supplementation and FTT in childhood. She has short stature, deformity of the distal metaphysis of the ulnae, and ongoing neutropenia with a hypocellular BM but no cytogenetic clones. Targeted SMRT sequencing identified a deletion in SBDS which deletes all of exon 3 and part of the surrounding introns (872bp, hg19: chr7: 66,457,992-66,458,863). Western blotting for SBDS showed a decrease in protein production in cultured fibroblasts consistent with a diagnosis of SDS. Sequencing of the family members indicated the deletion was paternal in origin, while the SNV was maternal. A deletion of SRP54 was identified in two affected siblings from a third family; both siblings have a history of neutropenia and mild BM dysplasia. The deletion encompasses exon 8 of SRP54, which encodes part of the G-domain, critical for SRP54 cotranslational function, and is near other reported autosomal dominant pathogenic variants. One affected individual in this family also has a single SBDS c.183_184TA>CT variant, but had normal SBDS protein levels by western blot. In this family, the SBDS variant is likely not disease causing as individuals with heterozygous pathogenic variants of SBDS are generally asymptomatic, but rather the dominant SRP54 deletion is the disease causing genetic alteration in this SDS-like family. No large deletions or insertions were identified in DNAJC21. Analysis of EFL1 is on-going. These cases illustrate the advantages of using long-read sequencing methodologies to identify large deletions which are not readily found using short-read sequencing, and identify large deletions as a novel mechanism of SDS inheritance. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 451-451
Author(s):  
Valentino Bezzerri ◽  
Antonio Vella ◽  
Elisabetta D'Aversa ◽  
Martina Api ◽  
Marisole Allegri ◽  
...  

Shwachman-Diamond syndrome (SDS) is one of the more common inherited bone marrow failure syndromes (IBMFS). Almost 90% of patients with SDS present mutations in the Shwachman-Bodian-Diamond syndrome gene (SBDS) which encodes for the homonymous small protein involved in ribogenesis. SDS is a multiple-organ disease mostly characterized by exocrine pancreas insufficiency, bone malformations, and more importantly bone marrow failure. Most patients with SDS present severe neutropenia, whereas thrombocytopenia and anemia are less frequent. Furthermore, 15-20% of patients develop myelodysplastic syndrome with high risk of acute myeloid leukemia (AML). STAT3 pathway is upregulated both in primary SDS leukocytes and immortalized B cells. Being STAT3 a key regulator of interleukin-6 (IL-6), we postulated that STAT3 hyper-activation could lead to a dysregulation of the IL-6 signaling cascade. Increased levels of IL-6 have been found in pediatric patients with AML and it has been associated with poorer outcomes in these patients, highlighting IL-6 as a cytokine potentially involved in the development of AML. Thus, our hypothesis is that STAT3-IL6 axis may contribute to leukemogenesis in SDS. Almost 55% of patients with SDS carry a specific nonsense mutations, namely the c.183-184TA>CT, which cause a premature termination codon (PTC). Ataluren (PTC124, PTC Therapeutics Inc, NJ) is a small PTC suppressor molecule already approved by the European Medicines Agency as a therapeutic option for Duchenne muscular dystrophy. Interestingly, we recently reported that ataluren can restore SBDS expression in bone marrow progenitors and in peripheral blood mononuclear cells isolated from patients with SDS. Moreover, we have shown that ataluren can reduce mTOR hyper-phosphorylation and excessive apoptotic rate observed in SDS leukocytes. More importantly, we reported that ataluren can improve myeloid differentiation in a small cohort of patients (Bezzerri et al, Am J Hematol 2018). In this further analysis considering an enlarged cohort of 20 SDS patients carrying nonsense mutations we found the following: Ataluren can significantly improve both myeloid colony-forming unit-granulocyte/macrophage (CFU-GM) and colony-forming unit granulocyte, erythrocyte, monocyte, megakaryocyte (CFU-GEMM) generation from bone marrow mononuclear stem cells obtained from an enlarged cohort of 20 patients with SDS carrying nonsense mutations. Ataluren indeed almost doubled the number of CFU-GM and CFU-GEMM after 7 and 14 days of treatment.Colony-forming unit erythroid (CFU-E) generation was not affected by the treatment.Ataluren induces neutrophil maturation in SDS bone marrow mononuclear stem cells (mean increase of 61% CD16+ CD11b+ cells over untreated controls) after 24-48 hours of treatment.Consistently with STAT3 hyper-activation observed in SDS cells, here we show that patients with SDS present a significantly increased level of IL-6 in plasma (4.3-fold higher expression than the healthy control group). Also lymphoblastoid cell lines (LCL) and primary bone marrow mesenchymal stromal cells (MSC) obtained from patients with SDS show increased IL-6 release in culture supernatants compared to healthy controls (2.5-fold and 6.8-fold higher levels, respectively).Of note, ataluren can reduce IL-6 expression in SDS cells restoring normal levels both in LCL and MSC. In conclusion, these new data support the enrollment of patients for the first clinical trial for this drug in SDS. Furthermore, this study could pave the way for the use of ataluren for other nonsense mutation-mediated IBMFS where STAT3-IL6 axis and similar pro-leukemic pathways are involved. Disclosures Bezzerri: Marco Cipolli, Valentino Bezzerri, Baroukh Maurice Assael: Patents & Royalties: WO2018/050706 A1 "Method of treatment of Shwachman-Diamond syndrome". Cipolli:Marco Cipolli, Valentino Bezzerri, Baroukh Maurice Assael: Patents & Royalties: WO2018/050706 A1 "Method of treatment of Shwachman-Diamond syndrome".


2018 ◽  
Vol 184 (6) ◽  
pp. 974-981 ◽  
Author(s):  
Roberto Valli ◽  
Antonella Minelli ◽  
Marta Galbiati ◽  
Giovanna D'Amico ◽  
Annalisa Frattini ◽  
...  

2012 ◽  
Vol 4 (3) ◽  
pp. 30 ◽  
Author(s):  
Shin-ichiro Hagiwara ◽  
Arata Watanabe

Shwachman-Diamond syndrome (SDS) is a rare, inherited, autosomal recessive disease characterized by exocrine pancreatic dysfunction, skeletal problems and varying degrees of cytopenias resulting in bone marrow dysfunction. We report the first case of SDS that was difficult to distinguish from celiac disease because this is a valuable example of the variety in SDS presentation.


2019 ◽  
Vol 32 (8) ◽  
pp. 879-884 ◽  
Author(s):  
Raquel Corripio ◽  
Carla Tubau ◽  
Laura Calvo ◽  
Carme Brun ◽  
Núria Capdevila ◽  
...  

Abstract Background There is little evidence of the effects of early treatment with growth hormone (GH) in infants with Prader-Willi syndrome (PWS). A prospective study was conducted to assess the safety of GH therapy in infants younger than 2 years of age with PWS. Methods A total of 14 patients with PWS started treatment with GH under the age of 2 years and were followed over a 2-year period. A deletion of chromosome 15 was present in nine infants (64.3%) and maternal uniparental disomy 15 in five infants (35.7%). The median age at start of GH treatment was 9.6 months (interquartile range [IQR] 9.0–18.3 months). Changes in height standard deviation score (SDS), body mass index (BMI) SDS and subcapsular and tricipital skinfolds in the follow-up period were evaluated with a mixed-model regression analysis using the Package R. Results There were no fatal adverse events. A significant decrease (p < 0.001) in tricipital and subcapsular skinfold thickness, with an upward trend of height SDS and a downward trend of BMI SDS, was observed. Infants who started GH before 15 months of age started walking at a median of 18.0 [17.0–19.5] months vs. 36.6 [36.3–37.8] months for those who began treatment with GH after 15 months of age (p = 0.024). Conclusions GH treatment in infants with PWS less than 2 years of age is safe and improved body composition. Infants who received GH before the age of 15 months started to walk earlier.


2015 ◽  
Vol 05 (03) ◽  
pp. 087-092
Author(s):  
Anirban Chakraborty ◽  
Indrani Karunasagar

AbstractRibosomes are molecular machineries that decode the information within mRNAs and generate all the proteins required for cellular activities. Ribosomes are essential to every living organism. The synthesis of ribosome is an intricate process, which is carried out in multiple steps throughout the cell in a highly coordinated fashion. For many years, the general perception was that any defects in the “ribosome assembly line” would have fatal consequences on cell. However, it has now become clear that production of defective ribosomes does not lead to lethality in human embryos. Rather, it manifests as specific disease conditions called ribosomopathies, which are rare genetic disorders affecting the bone marrow. This group of diseases has received considerable attention in recent years because of the mystery associated with them i.e. the tissue-specific nature of the clinical phenotypes despite the fact that the genes mutated in patients code for proteins that are absolutely essential and are housekeeping in nature. Despite considerable progress in understanding these diseases, it still remains unclear why defects in the production of a macromolecule as indispensable and as ubiquitous as the ribosome go unnoticed and why the effects are not universal but rather are restricted to specific cell types. This review is aimed at introducing the readers to important ribosomopathies with a brief description about the clinical symptoms, molecular genetics, and the treatments strategies.


Blood ◽  
1986 ◽  
Vol 67 (2) ◽  
pp. 350-353 ◽  
Author(s):  
J Stamberg ◽  
A Shende ◽  
P Lanzkowsky

Abstract A 12-year-old girl with acute lymphoblastic leukemia (ALL) had two types of acquired cytogenetic abnormalities in her pretreatment peripheral blood and bone marrow: hyperdiploidy due to tetrasomy 8, 10, and 21; and, in the hyperdiploid cells, a shift from heterozygosity to homozygosity for a polymorphic variant on chromosome 15. Both abnormalities disappeared after chemotherapy, when the patient entered clinical remission. It has recently been found that shifts to homozygosity occur in retinoblastoma and Wilms' tumor. Our observation extends this finding to leukemia and indicates that such shifts may have general importance in tumorigenesis.


2018 ◽  
Vol 07 (03) ◽  
pp. 117-121
Author(s):  
Figen Özçay ◽  
Lale Olcay ◽  
Serdar Ceylaner ◽  
Taner Sezer ◽  
Zeren Barış

AbstractWe present a patient with failure to thrive and severe hypotonia, who was initially suspected of having a neurometabolic disease but later diagnosed as Shwachman–Diamond syndrome (SDS), which was genetically confirmed. SDS is a multisystemic disease, which is characterized by exocrine pancreatic deficiency, bone marrow dysfunction with increased risk for malignant transformation, and skeletal abnormalities. It should be included in differential diagnosis of patients with failure to thrive and unexplained neurodevelopmental delay with neutropenia.


Sign in / Sign up

Export Citation Format

Share Document