Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep Med ; 4(8): 101160, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37586319

ABSTRACT

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.


Subject(s)
Hematopoietic Stem Cells , Inflammation , Humans , Proteolysis , Cell Differentiation , Inflammation/genetics
2.
Blood ; 139(1): 34-43, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34525188

ABSTRACT

Patients with severe aplastic anemia (SAA) are either treated with bone marrow transplant (BMT) or immunosuppression (IST) depending on their age, comorbidities, and available donors. In 2017, our phase 2 trial reported improved hematologic responses with the addition of eltrombopag (EPAG) to standard IST for SAA when compared with a historical cohort treated with IST alone. However, the rates and characteristics of long-term complications, relapse, and clonal evolution, previously described in patients treated with IST alone, are not yet known with this new regimen, IST and EPAG. Patients were accrued from 2012 to 2020, with a total of 178 subjects included in this secondary endpoint analysis. With double the sample size and a much longer median follow-up (4 years) since the original publication in 2017, we report a cumulative relapse rate of 39% in responding patients who received cyclosporine (CSA) maintenance and clonal evolution of 15% in all treated patients at 4 years. Relapse occurred at distinct timepoints: after CSA dose reduction and EPAG discontinuation at 6 months, and after 2 years when CSA was discontinued. Most relapsed patients were retreated with therapeutic doses of CSA +/- EPAG, and two-thirds responded. Clonal evolution to a myeloid malignancy or chromosome 7 abnormality (high-risk) was noted in 5.7% of patients and conferred a poorer overall survival. Neither relapse nor high-risk evolution occurred at a higher rate than was observed in a historical comparator cohort, but the median time to both events was earlier in IST and EPAG treated patients. This trial was registered at www.clinicaltrials.gov as #NCT01623167.


Subject(s)
Anemia, Aplastic/drug therapy , Benzoates/therapeutic use , Cyclosporine/therapeutic use , Hydrazines/therapeutic use , Immunosuppressive Agents/therapeutic use , Pyrazoles/therapeutic use , Adult , Female , Humans , Male , Middle Aged , Prospective Studies , Survival Analysis , Treatment Outcome , Young Adult
3.
Blood ; 138(26): 2799-2809, 2021 12 30.
Article in English | MEDLINE | ID: mdl-34724566

ABSTRACT

Immune aplastic anemia (AA) features somatic loss of HLA class I allele expression on bone marrow cells, consistent with a mechanism of escape from T-cell-mediated destruction of hematopoietic stem and progenitor cells. The clinical significance of HLA abnormalities has not been well characterized. We examined the somatic loss of HLA class I alleles and correlated HLA loss and mutation-associated HLA genotypes with clinical presentation and outcomes after immunosuppressive therapy in 544 AA patients. HLA class I allele loss was detected in 92 (22%) of the 412 patients tested, in whom there were 393 somatic HLA gene mutations and 40 instances of loss of heterozygosity. Most frequently affected was HLA-B*14:02, followed by HLA-A*02:01, HLA-B*40:02, HLA-B*08:01, and HLA-B*07:02. HLA-B*14:02, HLA-B*40:02, and HLA-B*07:02 were also overrepresented in AA. High-risk clonal evolution was correlated with HLA loss, HLA-B*14:02 genotype, and older age, which yielded a valid prediction model. In 2 patients, we traced monosomy 7 clonal evolution from preexisting clones harboring somatic mutations in HLA-A*02:01 and HLA-B*40:02. Loss of HLA-B*40:02 correlated with higher blood counts. HLA-B*07:02 and HLA-B*40:01 genotypes and their loss correlated with late-onset of AA. Our results suggest the presence of specific immune mechanisms of molecular pathogenesis with clinical implications. HLA genotyping and screening for HLA loss may be of value in the management of immune AA. This study was registered at clinicaltrials.gov as NCT00001964, NCT00061360, NCT00195624, NCT00260689, NCT00944749, NCT01193283, and NCT01623167.


Subject(s)
Anemia, Aplastic/genetics , Genes, MHC Class I , HLA-A Antigens/genetics , HLA-B Antigens/genetics , Mutation , Adolescent , Adult , Alleles , Anemia, Aplastic/immunology , Clonal Evolution , Female , Gene Deletion , Gene Expression , HLA-A Antigens/immunology , HLA-B Antigens/immunology , Humans , Immunity , Loss of Heterozygosity , Male , Middle Aged , Young Adult
4.
Br J Haematol ; 190(4): 610-617, 2020 08.
Article in English | MEDLINE | ID: mdl-32311088

ABSTRACT

Immune aplastic anaemia (AA) is caused by cytotoxic T lymphocytes (CTLs) that destroy haematopoietic stem and progenitor cells. Enhanced type 1 T helper (Th1) responses and reduced regulatory T cells (Tregs) are involved in the immune pathophysiology. CD24hi CD38hi regulatory B cells (Bregs) suppress CTLs and Th1 responses, and induce Tregs via interleukin 10 (IL-10). We investigated circulating B-cell subpopulations, including CD24hi CD38hi Bregs, as well as total B cells, CD4+ T cells, CD8+ T cells and natural killer cells in 104 untreated patients with severe and very severe AA, aged ≥18 years. All patients were treated with standard immunosuppressive therapy (IST) plus eltrombopag. CD24hi CD38hi Bregs were markedly reduced in patients with AA compared to healthy individuals, especially in very severe AA, but residual Bregs remained functional, capable of producing IL-10; total B-cell counts and the other B-cell subpopulations were similar to those of healthy individuals. CD24hi CD38hi Bregs did not correlate with responses to IST, and they recovered to levels present in healthy individuals after therapy. Mature naïve B-cell counts were unexpectedly associated with IST response. Markedly reduced CD24hi CD38hi Bregs, especially in very severe AA, with recovery after IST suggest Breg deficits may contribute to the pathophysiology of immune AA.


Subject(s)
ADP-ribosyl Cyclase 1/analysis , Anemia, Aplastic/blood , Antigens, CD19/analysis , B-Lymphocyte Subsets/pathology , B-Lymphocytes, Regulatory/pathology , CD24 Antigen/analysis , Lymphopenia/etiology , Membrane Glycoproteins/analysis , Adolescent , Adult , Aged , Anemia, Aplastic/complications , Anemia, Aplastic/drug therapy , Anemia, Aplastic/pathology , Antilymphocyte Serum/therapeutic use , B-Lymphocyte Subsets/chemistry , B-Lymphocytes, Regulatory/chemistry , Benzoates/therapeutic use , Bone Marrow/pathology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cyclosporine/therapeutic use , Female , Humans , Hydrazines/therapeutic use , Immunosuppressive Agents/therapeutic use , Interleukin-10/biosynthesis , Interleukin-10/genetics , Killer Cells, Natural/pathology , Lymphocyte Count , Lymphopenia/blood , Lymphopenia/pathology , Male , Middle Aged , Pyrazoles/therapeutic use , Receptors, Thrombopoietin/agonists , Young Adult
5.
Int J Oncol ; 52(4): 1209-1223, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29532865

ABSTRACT

Additional sex combs-like 1 (ASXL1) is a well­known tumor suppressor gene and epigenetic modifier. ASXL1 mutations are frequent in myeloid malignances; these mutations are risk factors for the development of myelodysplasia and also appear as small clones during normal aging. ASXL1 appears to act as an epigenetic regulator of cell survival and myeloid differentiation; however, the molecular mechanisms underlying the malignant transformation of cells with ASXL1 mutations are not well defined. Using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein-9 nuclease (Cas9) genome editing, heterozygous and homozygous ASXL1 mutations were introduced into human U937 leukemic cells. Comparable cell growth and cell cycle progression were observed between wild-type (WT) and ASXL1-mutated U937 cells. Drug-induced cytotoxicity, as measured by growth inhibition and apoptosis in the presence of the cell-cycle active agent 5-fluorouracil, was variable among the mutated clones but was not significantly different from WT cells. In addition, ASXL1-mutated cells exhibited defects in monocyte/macrophage differentiation. Transcriptome analysis revealed that ASXL1 mutations altered differentiation of U937 cells by disturbing genes involved in myeloid differentiation, including cytochrome B-245 ß chain and C-type lectin domain family 5, member A. Dysregulation of numerous gene sets associated with cell death and survival were also observed in ASXL1-mutated cells. These data provide evidence regarding the underlying molecular mechanisms induced by mutated ASXL1 in leukemogenesis.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Repressor Proteins/genetics , CRISPR-Cas Systems , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/genetics , Fluorouracil/pharmacology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelomonocytic, Chronic/pathology , Tetradecanoylphorbol Acetate/pharmacology , Transcriptome , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...