Try a new search

Format these results:

Searched for:

person:db1510

in-biosketch:true

Total Results:

60


Use of flow cytometric light scattering to recognize the characteristic vacuolated marrow cells in VEXAS syndrome

Ding, Yanna; Dulau-Florea, Alina E; Groarke, Emma M; Patel, Bhavisha A; Beck, David B; Grayson, Peter C; Ferrada, Marcela A; Young, Neal S; Calvo, Katherine R; Braylan, Raul C
PMID: 37477595
ISSN: 2473-9537
CID: 5536162

Early activation of inflammatory pathways in UBA1-mutated hematopoietic stem and progenitor cells in VEXAS

Wu, Zhijie; Gao, Shouguo; Gao, Qingyan; Patel, Bhavisha A; Groarke, Emma M; Feng, Xingmin; Manley, Ash Lee; Li, Haoran; Ospina Cardona, Daniela; Kajigaya, Sachiko; Alemu, Lemlem; Quinones Raffo, Diego; Ombrello, Amanda K; Ferrada, Marcela A; Grayson, Peter C; Calvo, Katherine R; Kastner, Daniel L; Beck, David B; Young, Neal S
VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome is a pleiotropic, severe autoinflammatory disease caused by somatic mutations in the ubiquitin-like modifier activating enzyme 1 (UBA1) gene. To elucidate VEXAS pathophysiology, we performed transcriptome sequencing of single bone marrow mononuclear cells and hematopoietic stem and progenitor cells (HSPCs) from VEXAS patients. HSPCs are biased toward myeloid (granulocytic) differentiation, and against lymphoid differentiation in VEXAS. Activation of multiple inflammatory pathways (interferons and tumor necrosis factor alpha) occurs ontogenically early in primitive hematopoietic cells and particularly in the myeloid lineage in VEXAS, and inflammation is prominent in UBA1-mutated cells. Dysregulation in protein degradation likely leads to higher stress response in VEXAS HSPCs, which positively correlates with inflammation. TCR usage is restricted and there are increased cytotoxicity and IFN-γ signaling in T cells. In VEXAS syndrome, both aberrant inflammation and myeloid predominance appear intrinsic to hematopoietic stem cells mutated in UBA1.
PMCID:10439277
PMID: 37586319
ISSN: 2666-3791
CID: 5595732

A ubiquitin-based effector-to-inhibitor switch coordinates early brain, craniofacial, and skin development

Asmar, Anthony J; Abrams, Shaun R; Hsin, Jenny; Collins, Jason C; Yazejian, Rita M; Wu, Youmei; Cho, Jean; Doyle, Andrew D; Cinthala, Samhitha; Simon, Marleen; van Jaarsveld, Richard H; Beck, David B; Kerosuo, Laura; Werner, Achim
The molecular mechanisms that coordinate patterning of the embryonic ectoderm into spatially distinct lineages to form the nervous system, epidermis, and neural crest-derived craniofacial structures are unclear. Here, biochemical disease-variant profiling reveals a posttranslational pathway that drives early ectodermal differentiation in the vertebrate head. The anteriorly expressed ubiquitin ligase CRL3-KLHL4 restricts signaling of the ubiquitous cytoskeletal regulator CDC42. This regulation relies on the CDC42-activating complex GIT1-βPIX, which CRL3-KLHL4 exploits as a substrate-specific co-adaptor to recognize and monoubiquitylate PAK1. Surprisingly, we find that ubiquitylation converts the canonical CDC42 effector PAK1 into a CDC42 inhibitor. Loss of CRL3-KLHL4 or a disease-associated KLHL4 variant reduce PAK1 ubiquitylation causing overactivation of CDC42 signaling and defective ectodermal patterning and neurulation. Thus, tissue-specific restriction of CDC42 signaling by a ubiquitin-based effector-to-inhibitor is essential for early face, brain, and skin formation, revealing how cell-fate and morphometric changes are coordinated to ensure faithful organ development.
PMCID:10371987
PMID: 37495603
ISSN: 2041-1723
CID: 5592202

Spectrum of clonal hematopoiesis in VEXAS syndrome

Gutierrez-Rodrigues, Fernanda; Kusne, Yael; Fernandez, Jenna; Lasho, Terra L; Shalhoub, Ruba N; Ma, Xiaoyang; Alessi, Hugh; Finke, Christy M; Koster, Matthew J; Mangaonkar, Abhishek A; Warrington, Kenneth J; Begna, Kebede; Xie, Zhuoer; Ombrello, Amanda K; Viswanatha, David S; Ferrada, Marcela A; Wilson, Lorena; Go, Ronald S; Kourelis, Taxiarchis V; Reichard, Kaaren K; Olteanu, Horatiu; Darden, Ivana; Hironaka, Dalton; Alemu, Lemlem; Kajigaya, Sachiko; Calado, Rodrigo T; Groarke, Emma M; Rosenzweig, Sofia; Kastner, Daniel L; Calvo, Katherine R; Wu, Colin O; Grayson, Peter C; Young, Neal S; Beck, David B; Patel, Bhavisha A; Patnaik, Mrinal M
VEXAS is caused by somatic mutations in UBA1 (UBA1mut) and characterized by heterogenous systemic auto-inflammation and progressive hematologic manifestations, meeting criteria for myelodysplastic syndrome (MDS) and plasma cell dyscrasias. The landscape of myeloid-related gene mutations leading to typical clonal hematopoiesis (CH) in these patients is unknown. Retrospectively, we screened 80 VEXAS patients for CH in their peripheral blood (PB) and correlated findings with clinical outcomes in 77. UBA1mutwere most common at hotspot p.M41 (median variant allele frequency/VAF = 75%). Typical CH mutations co-occurred with UBA1mut in 60% of patients, mostly in DNMT3A and TET2, and were not associated with inflammatory or hematologic manifestations. In prospective single-cell proteogenomic sequencing (scDNA), UBA1mutwas the dominant clone, present mostly in branched clonal trajectories. Based on integrated bulk and scDNA analyses, clonality in VEXAS followed two major patterns: with either typical CH preceding UBA1mutselection in a clone (Pattern 1), or occurring as an UBA1mutsubclone or in independent clones (Pattern 2). VAF in PB differed markedly between DNMT3A and TET2 clones (median VAF of 25% vs 1%). DNMT3A and TET2 clones associated with hierarchies representing patterns 1 and 2, respectively. Overall survival for all patients was 60% at 10 years. Transfusion-dependent anemia, moderate thrombocytopenia, and typical CH mutations, each correlated with poor outcome. In VEXAS, UBA1mut cells are the primary cause of systemic inflammation and marrow failure, being a new molecularly defined somatic entity associated with MDS. VEXAS-associated MDS is distinct from classical MDS in its presentation and clinical course.
PMID: 37084382
ISSN: 1528-0020
CID: 5466382

Novel somatic UBA1 variant in a patient with VEXAS syndrome

Stiburkova, Blanka; Pavelcova, Katerina; Belickova, Monika; Magaziner, Samuel J; Collins, Jason C; Werner, Achim; Beck, David B; Balajkova, Veronika; Salek, Cyril; Vostry, Martin; Mann, Herman; Vencovsky, Jiri
OBJECTIVE:Somatic mutations in UBA1 have recently been causally linked to a severe adult-onset inflammatory condition referred to as VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome. UBA1 is of fundamental importance to the modulation of ubiquitin homeostasis and to the majority of downstream ubiquitylation-dependent cellular processes. Direct sequencing analysis of exon 3 containing prevalent variants p.Met41Leu, p.Met41Val, and/or p.Met41Thr is usually used to confirm the disease associated mutations. METHODS:We studied clinical, biochemical and molecular genetic characteristics of a fifty-nine-year-old male with two-year history of arthritis, fever, night sweats, nonspecific skin rash, lymphadenopathy, and myelodysplastic syndrome with multilineage dysplasia. RESULTS:The mutational analysis revealed a hitherto undescribed sequence variant c.1430G>C in exon 14 (p.Gly477Ala) in UBA1 gene. In vitro enzymatic analyses showed that p.Gly477Ala led to both decreased E1 ubiquitin thioester formation and E2 enzyme charging. CONCLUSION/CONCLUSIONS:We herein report a case of a patient of European ancestry with clinical manifestations of VEXAS syndrome associated with a newly identified dysfunctional variant UBA1 enzyme. Due to insufficient response to various immunosuppressive treatments, allogeneic hematopoietic stem cell transplantation was performed, which resulted in significant improvement of clinical and laboratory manifestations of the disease.
PMID: 36762418
ISSN: 2326-5205
CID: 5420952

VEXAS syndrome: A review of bone marrow aspirate and biopsies reporting myeloid and erythroid precursor vacuolation

Cherniawsky, Hannah; Friedmann, Jordan; Nicolson, Hamish; Dehghan, Natasha; Stubbins, Ryan J; Foltz, Lynda M; Leitch, Heather A; Sreenivasan, Gayatri M; Ambler, Kimberley L S; Nevill, Thomas J; McGinnis, Eric; Wilson, Lorena; Beck, David B; Chen, Luke Y C; Marcon, Krista M
Myeloid and erythroid precursor vacuolation is a common dysplastic finding associated with myeloid malignancies, toxins, drug, and nutritional deficiencies. It has been described as a core morphologic feature in VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome. We sought to determine the number of cases attributable to VEXAS syndrome in bone marrow biopsies and aspirates (BAMB) reporting myeloid precursor vacuolation. We reviewed 1318 individual BAMB reports from January 2020 to July 2021 where "vacuole(s)," "vacuolation," or "vacuolated" was reported. Bone marrow biopsies with vacuolation confined to blasts or those completed as routine workup prior to stem cell transplant or post induction chemotherapy for AML (acute myeloid leukemia) were excluded. Myeloid and erythroid precursor vacuolation was noted in 219 reports representing 210 patients. The most common etiology was myelodysplastic syndrome (MDS) (38.6%), AML (16.7%), lymphoproliferative disorders and multiple myeloma (7.6%), drug or toxin exposure (5.2%) myeloproliferative neoplasm (MPN) or MPN/MDS overlap syndrome (4.3%). VEXAS syndrome was determined to be the etiology in 2.9% of patients. Two additional cases of VEXAS syndrome with bone marrow biopsies reported in the specified time frame did not explicitly report myeloid or erythroid precursor vacuolation but were identified based on clinical suspicion and repeat BAMB review. Myeloid and erythroid precursor vacuolation is a dysplastic feature attributable to VEXAS syndrome in at least 2.9% of cases. Standardized reporting of vacuolization, triaging of molecular sequencing and optimal treatment of this disorder are critical issues facing those seeing patients with suspected VEXAS syndrome.
PMID: 36788756
ISSN: 1600-0609
CID: 5448052

Estimated Prevalence and Clinical Manifestations of UBA1 Variants Associated With VEXAS Syndrome in a Clinical Population

Beck, David B; Bodian, Dale L; Shah, Vandan; Mirshahi, Uyenlinh L; Kim, Jung; Ding, Yi; Magaziner, Samuel J; Strande, Natasha T; Cantor, Anna; Haley, Jeremy S; Cook, Adam; Hill, Wesley; Schwartz, Alan L; Grayson, Peter C; Ferrada, Marcela A; Kastner, Daniel L; Carey, David J; Stewart, Douglas R
IMPORTANCE:VEXAS (vacuoles, E1-ubiquitin-activating enzyme, X-linked, autoinflammatory, somatic) syndrome is a disease with rheumatologic and hematologic features caused by somatic variants in UBA1. Pathogenic variants are associated with a broad spectrum of clinical manifestations. Knowledge of prevalence, penetrance, and clinical characteristics of this disease have been limited by ascertainment biases based on known phenotypes. OBJECTIVE:To determine the prevalence of pathogenic variants in UBA1 and associated clinical manifestations in an unselected population using a genomic ascertainment approach. DESIGN, SETTING, AND PARTICIPANTS:This retrospective observational study evaluated UBA1 variants in exome data from 163 096 participants within the Geisinger MyCode Community Health Initiative. Clinical phenotypes were determined from Geisinger electronic health record data from January 1, 1996, to January 1, 2022. EXPOSURES:Exome sequencing was performed. MAIN OUTCOMES AND MEASURES:Outcome measures included prevalence of somatic UBA1 variation; presence of rheumatologic, hematologic, pulmonary, dermatologic, and other findings in individuals with somatic UBA1 variation on review of the electronic health record; review of laboratory data; bone marrow biopsy pathology analysis; and in vitro enzymatic assays. RESULTS:In 163 096 participants (mean age, 52.8 years; 94% White; 61% women), 11 individuals harbored likely somatic variants at known pathogenic UBA1 positions, with 11 of 11 (100%) having clinical manifestations consistent with VEXAS syndrome (9 male, 2 female). A total of 5 of 11 individuals (45%) did not meet criteria for rheumatologic and/or hematologic diagnoses previously associated with VEXAS syndrome; however, all individuals had anemia (hemoglobin: mean, 7.8 g/dL; median, 7.5 g/dL), which was mostly macrocytic (10/11 [91%]) with concomitant thrombocytopenia (10/11 [91%]). Among the 11 patients identified, there was a pathogenic variant in 1 male participant prior to onset of VEXAS-related signs or symptoms and 2 female participants had disease with heterozygous variants. A previously unreported UBA1 variant (c.1861A>T; p.Ser621Cys) was found in a symptomatic patient, with in vitro data supporting a catalytic defect and pathogenicity. Together, disease-causing UBA1 variants were found in 1 in 13 591 unrelated individuals (95% CI, 1:7775-1:23 758), 1 in 4269 men older than 50 years (95% CI, 1:2319-1:7859), and 1 in 26 238 women older than 50 years (95% CI, 1:7196-1:147 669). CONCLUSIONS AND RELEVANCE:This study provides an estimate of the prevalence and a description of the clinical manifestations of UBA1 variants associated with VEXAS syndrome within a single regional health system in the US. Additional studies are needed in unselected and genetically diverse populations to better define general population prevalence and phenotypic spectrum.
PMID: 36692560
ISSN: 1538-3598
CID: 5415112

Interleukin-1 receptor antagonist gene ( IL1RN ) variants modulate the cytokine release syndrome and mortality of SARS-CoV-2

Attur, Mukundan; Petrilli, Christopher; Adhikari, Samrachana; Iturrate, Eduardo; Li, Xiyue; Tuminello, Stephanie; Hu, Nan; Chakravarti, Aravinda; Beck, David; Abramson, Steven B
OBJECTIVE/UNASSIGNED:, the gene encoding the anti-inflammatory IL-1 receptor antagonist (IL-1Ra), on the cytokine release syndrome and mortality. METHODS/UNASSIGNED:gene were assessed for association with laboratory markers of the cytokine release syndrome (CRS) and mortality. RESULTS/UNASSIGNED:rs419598 CC SNV exhibited lower inflammatory biomarker levels, and was associated with reduced mortality compared to the CT/TT genotype in men (OR 0.49 (0.23 - 1.00); 0.052), with the most pronounced effect observed between the ages of 55-74 [5.5% vs. 18.4%, p<0.001]. CONCLUSION/UNASSIGNED:modulates the COVID-19 cytokine release syndrome via endogenous " anti-inflammatory" mechanisms. SIGNIFICANCE STATEMENT/UNASSIGNED:merits further evaluation in severe SARS-CoV-2 infection.
PMCID:9882468
PMID: 36711766
CID: 5602052

Novel genetic mutation in myositis-variant of VEXAS syndrome

Topilow, James S; Ospina Cardona, Daniela; Beck, David B; Ferrada, Marcela A; McMahan, Zsuzsanna H; Paik, Julie J
PMID: 35713495
ISSN: 1462-0332
CID: 5282802

Notes from the Field: VEXAS Syndrome and Disease Taxonomy in Rheumatology

Grayson, Peter C; Beck, David B; Ferrada, Marcela A; Nigrovic, Peter A; Kastner, Daniel L
PMID: 35696333
ISSN: 2326-5205
CID: 5282512