15 results on '"Diego Quinones Raffo"'
Search Results
2. Utility of plasma cell-free DNA for de novo detection and quantification of clonal hematopoiesis
- Author
-
Fernanda Gutierrez-Rodrigues, Isabel Beerman, Emma M. Groarke, Bhavisha A. Patel, Nina Spitofsky, Laura W. Dillon, Diego Quinones Raffo, Christopher S. Hourigan, Sachiko Kajigaya, Luigi Ferrucci, and Neal S. Young
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Although cell-free DNA (cfDNA) tests have emerged as a potential non-invasive alternative to bone marrow biopsies for monitoring clonal hematopoiesis in hematologic diseases, whether commercial cfDNA assays can be implemented for the detection and quantification of de novo clonal hematopoiesis in place of blood cells is uncertain. In this study, peripheral plasma cfDNA samples available from patients with aplastic anemia (n=25) or myelodysplastic syndromes (n=27) and a healthy cohort (n=107) were screened for somatic variants in genes related to hematologic malignancies using a Clinical Laboratory Improvement Amendments-certified panel. Results were further compared to DNA sequencing of matched blood cells. In reported results, 85% of healthy subjects, 36% of patients with aplastic anemia and 74% of patients with myelodysplastic syndromes were found to have somatic cfDNA variants, most frequently in DNMT3A, TET2, ASXL1 and SF3B1. However, concordance between cfDNA and blood cell findings was poor for the detection of clonal hematopoiesis when the allele frequency of the variants was
- Published
- 2021
- Full Text
- View/download PDF
3. Detectable mutations precede late myeloid neoplasia in aplastic anemia
- Author
-
Bhavisha A. Patel, Jack Ghannam, Emma M. Groarke, Meghali Goswami, Laura Dillon, Fernanda Gutierrez-Rodrigues, Olga Rios, Diego Quinones Raffo, Jennifer Lotter, Neal S. Young, and Christopher S. Hourigan
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
4. Circulating exosomal microRNAs in acquired aplastic anemia and myelodysplastic syndromes
- Author
-
Valentina Giudice, Lauren G. Banaszak, Fernanda Gutierrez-Rodrigues, Sachiko Kajigaya, Reema Panjwani, Maria del Pilar Fernandez Ibanez, Olga Rios, Christopher K. Bleck, Erin S. Stempinski, Diego Quinones Raffo, Danielle M. Townsley, and Neal S. Young
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Exosomal microRNAs modulate cancer cell metabolism and the immune response. Specific exosomal microRNAs have been reported to be reliable biomarkers of several solid and hematologic malignancies. We examined the possible diagnostic and prognostic values of exosomal microRNAs in two human bone marrow failure diseases: aplastic anemia and myelodysplastic syndromes. After screening 372 microRNAs in a discovery set (n=42) of plasma exosome samples, we constructed a customized PCR plate, including 42 microRNAs, for validation in a larger cohort (n=99). We identified 25 differentially expressed exosomal microRNAs uniquely or frequently present in aplastic anemia and/or myelodysplastic syndromes. These microRNAs could be related to intracellular functions, such as metabolism, cell survival, and proliferation. Clinical parameters and progression-free survival were correlated to microRNA expression levels in aplastic anemia and myelodysplastic syndrome patients before and after six months of immunosuppressive therapy. One microRNA, mir-126-5p, was negatively correlated with a response to therapy in aplastic anemia: patients with higher relative expression of miR-126-5p at diagnosis had the shortest progression-free survival compared to those with lower or normal levels. Our findings suggest utility of exosomal microRNAs in the differential diagnosis of bone marrow failure syndromes. (Registered at clinicaltrials.gov identifiers: 00260689, 00604201, 00378534, 01623167, 00001620, 00001397, 00217594).
- Published
- 2018
- Full Text
- View/download PDF
5. Differential diagnosis of bone marrow failure syndromes guided by machine learning
- Author
-
Fernanda Gutierrez-Rodrigues, Eric Munger, Xiaoyang Ma, Emma M. Groarke, Youbao Tang, Bhavisha A Patel, Luiz Fernando B. Catto, Diego V Clé, Marena R. Niewisch, Raquel MA Paiva, Flavia Donaires, André Luiz Pinto, Gustavo Borges, Barbara A. Santana, Lisa J McReynolds, Neelam Giri, Burak Altintas, Xing Fan, Ruba N Shalhoub, Christopher Mark Siwy, Carrie Diamond, Diego Quinones Raffo, Kathleen Marie Craft, Sachiko Kajigaya, Ronald M Summers, Paul P. Liu, Lea Cunningham, Dennis D Hickstein, Cynthia E. Dunbar, Ricardo Pasquini, Michel Michels de Oliveira, Elvira D R P Velloso, Blanche P Alter, Sharon A Savage, Carmem M.S. Bonfim, Colin O. Wu, Rodrigo T. Calado, and Neal S Young
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
The choice to postpone treatment while awaiting genetic testing can result in significant delay in definitive therapies in severely pancytopenic patients. Conversely, inherited bone marrow failure (BMF) misdiagnosis can expose patients to ineffectual and expensive therapies, toxic transplant conditioning regimens, and inappropriate use of an affected family member as a stem cell donor. To predict the likelihood of patients having acquired or inherited BMF, we developed a two-step data-driven machine-learning model using 25 clinical and laboratory variables typically recorded at the initial clinical encounter. For model's development, patients were labeled as having acquired or inherited BMF depending on their genomic data. Datasets were unbiasedly clustered and an ensemble model was trained with cases from the largest cluster of the training cohort (n=359) and validated with an independent cohort (n=127). Cluster A, the largest group, was mostly immune or inherited aplastic anemia, whereas Cluster B was composed of underrepresented BMF phenotypes, and not included in the next step of data modeling due to small sample size. The ensemble model Cluster A-specific was accurate (89%) to predict BMF etiology, correctly predicting inherited and likely immune BMF in 79% and 92% of cases, respectively. Our model represents a practical guide for BMF diagnosis and highlights the importance of clinical and laboratory variables in the initial evaluation, particularly telomere length. Our tool can be potentially used by general hematologists and health care providers not specialized in BMF, and in under-resourced centers, to prioritize patients for genetic testing or for expeditious treatment.
- Published
- 2022
- Full Text
- View/download PDF
6. Analysis of deficiency of adenosine deaminase 2 pathogenesis based on single-cell RNA sequencing of monocytes
- Author
-
Patrycja Hoffmann, Neal S. Young, Amanda K. Ombrello, Shouguo Gao, Carrie Diamond, Naoki Watanabe, Zhijie Wu, Sai Batchu, Lemlem Alemu, Diego Quinones Raffo, Sachiko Kajigaya, and Deborah L. Stone
- Subjects
Adult ,Male ,0301 basic medicine ,Cell signaling ,Adolescent ,Adenosine Deaminase ,CD14 ,Immunology ,Inflammation ,Biology ,Monocytes ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Agammaglobulinemia ,Gene expression ,medicine ,Humans ,Receptors, Tumor Necrosis Factor, Type II ,Immunology and Allergy ,Macrophage ,RNA, Messenger ,Child ,Sequence Analysis, RNA ,Gene Expression Profiling ,Monocyte ,NF-kappa B ,Cell Biology ,Middle Aged ,Tissue Donors ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Child, Preschool ,030220 oncology & carcinogenesis ,Mutation ,Cytokines ,Intercellular Signaling Peptides and Proteins ,Female ,Severe Combined Immunodeficiency ,Tumor necrosis factor alpha ,Interferons ,Single-Cell Analysis ,medicine.symptom ,Signal Transduction - Abstract
Deficiency of adenosine deaminase 2 (DADA2) is a rare autosomal recessive disease caused by loss-of-function variants in the ADA2 gene. DADA2 typically presents in childhood and is characterized by vasculopathy, stroke, inflammation, immunodeficiency, as well as hematologic manifestations. ADA2 protein is predominantly present in stimulated monocytes, dendritic cells, and macrophages. To elucidate molecular mechanisms in DADA2, CD14+ monocytes from 14 patients and 6 healthy donors were analyzed using single-cell RNA sequencing (scRNA-seq). Monocytes were purified by positive selection based on CD14 expression. Subpopulations were imputed from their transcriptomes. Based on scRNA-seq, monocytes could be classified as classical, intermediate, and nonclassical. Further, we used gene pathway analytics to interpret patterns of up- and down-regulated gene transcription. In DADA2, the frequency of nonclassical monocytes was higher compared with that of healthy donors, and M1 macrophage markers were up-regulated in patients. By comparing gene expression of each monocyte subtype between patients and healthy donors, we identified upregulated immune response pathways, including IFNα/β and IFNγ signaling, in all monocyte subtypes. Distinctively, the TNFR2 noncanonical NF-κB pathway was up-regulated only in nonclassical monocytes. Patients’ plasma showed increased IFNγ and TNFα levels. Our results suggest that elevated IFNγ activates cell signaling, leading to differentiation into M1 macrophages from monocytes and release of TNFα. Immune responses and more general response to stimuli pathways were up-regulated in DADA2 monocytes, and protein synthesis pathways were down-regulated, perhaps as stress responses. Our identification of novel aberrant immune pathways has implications for therapeutic approaches in DADA2 (registered at clinicaltrials.gov NCT00071045).
- Published
- 2021
- Full Text
- View/download PDF
7. Detectable mutations precede late myeloid neoplasia in aplastic anemia
- Author
-
Diego Quinones Raffo, Neal S. Young, Emma M. Groarke, Laura W. Dillon, Bhavisha A Patel, Christopher S. Hourigan, Fernanda Gutierrez-Rodrigues, Jack Ghannam, Olga Rios, Meghali Goswami, and Jennifer Lotter
- Subjects
Myeloid neoplasia ,business.industry ,Immunology ,Medicine ,Case Report ,Hematology ,Aplastic anemia ,business ,medicine.disease - Published
- 2020
8. Single-cell profiling of T lymphocytes in deficiency of adenosine deaminase 2
- Author
-
Diego Quinones Raffo, Shouguo Gao, Xingmin Feng, Patrycja Hoffmann, Zhijie Wu, Carrie Diamond, Amanda K. Ombrello, Sai Batchu, Sachiko Kajigaya, Neal S. Young, Lemlem Alemu, Naoki Watanabe, and Deborah L. Stone
- Subjects
Adult ,Male ,Adolescent ,Adenosine Deaminase ,T cell ,T-Lymphocytes ,Immunology ,Cell ,Inflammation ,Biology ,Lymphocyte Activation ,Skin Diseases ,Young Adult ,Immune system ,medicine ,Immunology and Allergy ,Humans ,Vascular Diseases ,Child ,Immunodeficiency ,Cells, Cultured ,Gene Expression Profiling ,T-cell receptor ,Immunologic Deficiency Syndromes ,Cell Biology ,Middle Aged ,medicine.disease ,Prognosis ,medicine.anatomical_structure ,STAT1 Transcription Factor ,Gene Expression Regulation ,Monocyte differentiation ,Case-Control Studies ,Mutation ,Intercellular Signaling Peptides and Proteins ,Female ,medicine.symptom ,Single-Cell Analysis ,CD8 ,Biomarkers ,Follow-Up Studies - Abstract
Deficiency of adenosine deaminase 2 (DADA2) is a monogenic vasculitis syndrome caused by autosomal-recessive loss-of-function mutations in the ADA2 gene (previously known as CECR1). Vasculitis, vasculopathy, and inflammation are dominant clinical features of this disease; the spectrum of manifestations includes immunodeficiency and lymphoproliferation as well as hematologic manifestations. ADA2 is primarily secreted by stimulated monocytes and macrophages. Aberrant monocyte differentiation to macrophages and neutrophils are important in the pathogenesis of DADA2, but little is known about T lymphocytes in this disease. We performed combined single-cell RNA sequencing and single-cell TCR sequencing in order to profile T cell repertoires in 10 patients with DADA2. Although there were no significant alterations of T cell subsets, we observed activation of both CD8+ and CD4+ T cells. There was no clonal expansion of T cells: most TCRs were expressed at basal levels in patients and healthy donors. TCR usage was private to individual patients and not disease specific, indicating as unlikely a common pathogenic background or predisposition to a common pathogen. We recognized activation of IFN pathways as a signature of T cells and STAT1 as a hub gene in the gene network of T cell activation and cytotoxicity. Overall, T cells in DADA2 patients showed distinct cell–cell interactions with monocytes, as compared with healthy donors, and many of these ligand–receptor interactions likely drove up-regulation of STAT1 in both T cells and other immune cells in patients. Our analysis reveals previously undercharacterized cell characteristics in DADA2.
- Published
- 2021
9. Long-term outcomes in patients with severe aplastic anemia treated with immunosuppression and eltrombopag: a phase 2 study
- Author
-
Diego Quinones Raffo, Colin O. Wu, Fernanda Gutierrez-Rodrigues, Bhavisha A Patel, Ruba Shalhoub, Neal S. Young, Jennifer Lotter, Emma M Groarke, and Olga Rios
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Eltrombopag ,Phases of clinical research ,Biochemistry ,Somatic evolution in cancer ,Benzoates ,chemistry.chemical_compound ,Young Adult ,Internal medicine ,Medicine ,Humans ,Prospective Studies ,Chromosome 7 (human) ,business.industry ,Anemia, Aplastic ,Immunosuppression ,Cell Biology ,Hematology ,Middle Aged ,Survival Analysis ,Discontinuation ,Regimen ,Hydrazines ,Treatment Outcome ,chemistry ,Cohort ,Cyclosporine ,Pyrazoles ,Female ,business ,Immunosuppressive Agents - 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.
- Published
- 2021
10. Clinical Utility of Plasma Cell-Free DNA for Detection and Quantification of Clonal Hematopoiesis
- Author
-
Fernanda Gutierrez-Rodrigues, Sachiko Kajigaya, Emma M. Groarke, Neal S. Young, Isabel Beerman, Nina Spitofsky, Bhavisha A Patel, Diego Quinones Raffo, and Luigi Ferrucci
- Subjects
medicine.anatomical_structure ,Immunology ,Clonal hematopoiesis ,medicine ,Cell Biology ,Hematology ,Plasma cell ,Biology ,Biochemistry ,Molecular biology ,Free dna - Abstract
Cell-free DNA (cfDNA) extracted from peripheral blood plasma has been increasingly used as a non-invasive approach for diagnosis and surveillance of solid and hematologic malignancies. Somatic variants associated with clonal hematopoiesis of indeterminate potential (CHIP) are commonly detected in such liquid biopsies, suggesting that cfDNA may be useful for their detection. CHIP has emerged as a predictor of progression to hematological malignancies; however, clones are still largely detected using peripheral blood (PB) and bone marrow (BM) cells. In this study, we investigated the performance of cfDNA for detection and quantification of CHIP compared to matched PB/BM cells. cfDNA initially was collected from a healthy cohort (n=106), part of the Baltimore Longitudinal Study of Aging/NIH; later we expanded to screen patients with aplastic anemia (AA; n=53) and myelodysplastic syndrome (MDS; n=27) monitored at the National Heart, Lung, and Blood Institute. Samples were screened for somatic mutations in myeloid neoplasm-related genes using a commercial panel of 177 genes. In HC (median age:72 [range:24-96]), 78/106 subjects (73%) were found to have one (28%) or two and more (45%) cfDNA variants in CHIP-related genes, most frequently in DNMT3A,TET2, TP53, and ASXL1 (Figure 1A). CHIP was observed in more than 60% of individuals older than 40 years, higher than typically reported. In contrast, only 17/53 of AA patients (32%; median age:51 [range:13-82]) were found to have one (n=10) or two or more (n=7) cfDNA variants, most commonly DNMT3A and SF3B1 (Figure 1B). In MDS, 17/29 (58%; median age:63 [range:35-85]) had one (n=10) or two or more (n=7) variants, TET2 and SF3B1 being most frequent (Figure 1C). Median VAF of cfDNA variants was significantly different among cohorts (HC:2.5% [95CI%:2-4] vs AA:18% [95CI%:6-32] vs MDS:38.6% [95CI%:27-42]; t-test,p cfDNA results were validated against matched PB/BM cells collected from HC (n=25), AA (n=56), and MDS patients (n=54). cfDNA variants were classified as true, and false positives or negatives according to their presence in PB/BM (Figure 1E). In HC, cfDNA sensitivity was moderate (58%), but specificity and PPV were low compared to PB. In AA and MDS, sensitivity, specificity and PPV values were high compared with BM (Table 1). In all cohorts, the median VAF of discordant variants was significantly lower than VAF of true-positives (9.5% [95CI%:8-12] vs. 36% [95CI%:31-37]; t-test,p40yo; Figure 1A). Discordant pairs were found most often in ASXL1, KIT, TET2, and ZRSR2 (Figure 1E). Although linear regression showed high correlation between the VAF of paired samples (R2>0.7), agreement of VAF by Bland-Altman analysis was poor; this approach calculates the bias, the mean difference between the VAF of matched samples, and its standard deviation (SD) to more accurately evaluate agreement between paired values. Here, the bias and SD were both as high as 9% in HC and patients. These large variations translated to a wide range of upper and lower limits of agreement (mean ± 2*SD), which represent the limits of acceptable differences. In summary, the landscape of somatic cfDNA variants depended on variant VAF. Technical factors were an important source of assay discordance, and although cfDNA variants were reliably detected at higher VAFs, their quantification was not comparable to VAF detected in blood cells. A single cfDNA clone varied up to 9% in size and SD was unacceptably high in all cohorts. cfDNA and PB/BM may not be interchangeable, as cfDNA may either over- or under-estimate clone size, regardless of disease status. Since small changes in clone size and dynamics may influence clinical evaluation and decisions, the use of cfDNA for CHIP detection requires robust sequencing coverage and validation of variants at VAF Disclosures Young: Novartis: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
11. Deficit of circulating CD19
- Author
-
Xingmin Feng, Diego Quinones Raffo, Lemlem Alemu, Bhavisha A Patel, Neal S. Young, Emma M. Groarke, Yoshitaka Zaimoku, and Sachiko Kajigaya
- Subjects
CD4-Positive T-Lymphocytes ,Male ,CD38 ,CD8-Positive T-Lymphocytes ,Benzoates ,0302 clinical medicine ,Bone Marrow ,Cytotoxic T cell ,Medicine ,B-Lymphocytes, Regulatory ,Membrane Glycoproteins ,Anemia, Aplastic ,Hematology ,Middle Aged ,aplastic anaemia ,Interleukin-10 ,Killer Cells, Natural ,Interleukin 10 ,Haematopoiesis ,Hydrazines ,030220 oncology & carcinogenesis ,Cyclosporine ,Female ,Receptors, Thrombopoietin ,Immunosuppressive Agents ,Research Paper ,Adult ,Adolescent ,Regulatory B cells ,Antigens, CD19 ,immunosuppressive therapy ,B-Lymphocyte Subsets ,03 medical and health sciences ,Young Adult ,Immune system ,Lymphopenia ,Humans ,Red Cells and Iron ,Lymphocyte Count ,Progenitor cell ,Aged ,Antilymphocyte Serum ,lymphocyte subsets ,business.industry ,flow cytometry ,CD24 Antigen ,regulatory B cells ,ADP-ribosyl Cyclase 1 ,Immunology ,Pyrazoles ,business ,CD8 ,030215 immunology - 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. CD24hiCD38hi regulatory B cells (Bregs) suppress CTLs and Th1 responses, and induce Tregs via interleukin 10 (IL‐10). We investigated circulating B‐cell subpopulations, including CD24hiCD38hi 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. CD24hiCD38hi 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. CD24hiCD38hi 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 CD24hiCD38hi Bregs, especially in very severe AA, with recovery after IST suggest Breg deficits may contribute to the pathophysiology of immune AA.
- Published
- 2020
12. CRISPR/Cas9-mediated ASXL1 mutations in U937 cells disrupt myeloid differentiation
- Author
-
Fernanda Gutierrez-Rodrigues, Lauren G. Banaszak, Shouguo Gao, Sachiko Kajigaya, Diego Quinones Raffo, Keyvan Keyvanfar, Xin Zhao, Zhijie Wu, and Neal S. Young
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Tumor suppressor gene ,U937 cells ,Biology ,medicine.disease_cause ,Transcriptome ,03 medical and health sciences ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Humans ,Epigenetics ,CRISPR/Cas9 ,Gene ,Cell Proliferation ,Mutation ,U937 cell ,Cell Cycle ,RNA sequencing ,Cell Differentiation ,Leukemia, Myelomonocytic, Chronic ,Articles ,Cell cycle ,ASXL1 mutations ,Cell biology ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,myeloid differentiation ,Tetradecanoylphorbol Acetate ,Fluorouracil ,CRISPR-Cas Systems - Abstract
Additional sex combs-like 1 (ASXL1) is a well‑known 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 β 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.
- Published
- 2018
- Full Text
- View/download PDF
13. Circulating exosomal microRNAs in acquired aplastic anemia and myelodysplastic syndromes
- Author
-
Sachiko Kajigaya, Maria del Pilar Fernandez Ibanez, Danielle M. Townsley, Fernanda Gutierrez-Rodrigues, Reema Panjwani, Olga Rios, Christopher K. E. Bleck, Neal S. Young, Erin Stempinski, Lauren G. Banaszak, Diego Quinones Raffo, and Valentina Giudice
- Subjects
0301 basic medicine ,Anemia ,Exosomes ,Exosome ,Article ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,medicine ,Humans ,Circulating MicroRNA ,Aplastic anemia ,business.industry ,Myelodysplastic syndromes ,Gene Expression Profiling ,Case-control study ,Anemia, Aplastic ,Biomarkers ,Case-Control Studies ,Computational Biology ,MicroRNAs ,Myelodysplastic Syndromes ,Prognosis ,Reproducibility of Results ,Aplastic ,Hematology ,Bone Marrow Failure ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,business - Abstract
Exosomal microRNAs modulate cancer cell metabolism and the immune response. Specific exosomal microRNAs have been reported to be reliable biomarkers of several solid and hematologic malignancies. We examined the possible diagnostic and prognostic values of exosomal microRNAs in two human bone marrow failure diseases: aplastic anemia and myelodysplastic syndromes. After screening 372 microRNAs in a discovery set (n=42) of plasma exosome samples, we constructed a customized PCR plate, including 42 microRNAs, for validation in a larger cohort (n=99). We identified 25 differentially expressed exosomal microRNAs uniquely or frequently present in aplastic anemia and/or myelodysplastic syndromes. These microRNAs could be related to intracellular functions, such as metabolism, cell survival, and proliferation. Clinical parameters and progression-free survival were correlated to microRNA expression levels in aplastic anemia and myelodysplastic syndrome patients before and after six months of immunosuppressive therapy. One microRNA, mir-126-5p, was negatively correlated with a response to therapy in aplastic anemia: patients with higher relative expression of miR-126-5p at diagnosis had the shortest progression-free survival compared to those with lower or normal levels. Our findings suggest utility of exosomal microRNAs in the differential diagnosis of bone marrow failure syndromes. (Registered at clinicaltrials.gov identifiers: 00260689, 00604201, 00378534, 01623167, 00001620, 00001397, 00217594).
- Published
- 2018
14. Spectrum and Clinical Significance of HLA Class I Alleles and Their Somatic Mutations in Immune Aplastic Anemia
- Author
-
Holly DiFebo, Emma M. Groarke, Willy A. Flegel, Lemlem Alemu, Neal S. Young, Xingmin Feng, Olga Rios, Yoshitaka Zaimoku, Audrey Lee, Sharon Adams, Diego Quinones Raffo, Sachiko Kajigaya, and Bhavisha A Patel
- Subjects
Immunology ,Cell Biology ,Hematology ,Human leukocyte antigen ,Gene mutation ,Biology ,medicine.disease ,Biochemistry ,Somatic evolution in cancer ,Loss of heterozygosity ,HLA-B Antigens ,medicine ,Allele ,Aplastic anemia ,Allele frequency - Abstract
Clonal hematopoiesis associated with loss of HLA class I alleles due to somatic mutations and/or 6p loss of heterozygosity (LOH) is frequent in immune aplastic anemia (AA). HLA-B*40:02 is more likely to be involved in HLA loss in Japanese AA patients, suggesting a role for this allele in immune pathophysiology (Zaimoku Y et al, Blood 2017). Mutations in non-B*40:02 HLA class I alleles have been reported in a limited number of patients from the United States (Babushok D et al, Blood Adv 2017) and Japan (Mizumaki H et al, 60th ASH meeting), but their prevalence and clinical significance are not well characterized. We investigated somatic mutations of HLA class I alleles, HLA allele frequencies, and their correlations with outcomes of therapy in a total of 532 AA patients, aged 2 years or older, treated on various Hematology Branch protocols (clinicaltrials.gov NCTs 00001964, 00061360, 00195624, 00260689, 00944749, 01193283, and 01623167). HLA allele-lacking (HLA-) monocytes from cryopreserved peripheral blood mononuclear cells were screened by flow cytometry after staining with allele-specific monoclonal antibodies for HLA-A and/or HLA-B (HLA-flow) in 172 AA patients. HLA- monocytes accounting for 0.5% to 100% (median 9.5%) of total monocytes were detected in 49 (28%) of the 172 patients and in 59 (15%) of 382 alleles analyzed (Figure 1). Loss of cell surface expression was frequent for HLA-B14 (46%), B27 (33%), B49 (33%), A68 (26%), A2 (23%), B40 (21%), and B8 (21%). One percent to 60% (median, 8.9%) of glycosylphosphatidylinositol-linked protein-negative (GPI-) monocytes were also present in 43% (21 of 49) of the patients with HLA- monocytes, but GPI- clones had normal HLA cell surface expression. Deep sequencing of HLA-A, HLA-B and HLA-C on sorted HLA- and HLA+GPI+ monocytes was performed in 42 of the 48 patients from whom adequate cells were available. Somatic mutations and/or LOH corresponding to the lacking alleles were detected in all 42 cases (Figure 1): 9 had both somatic mutations and LOH, 20 had somatic mutations only, and 13 had LOH only. Among the 13 patients who showed only LOH in the absent allele, 6 had somatic mutations in other alleles of HLA+ monocytes that was not analyzable of HLA expression, and 2 had a breakpoint of LOH between HLA-A and HLA-C, leading to loss of a single HLA-A allele. Somatic mutations or LOH involving only one allele were present in 37 patients among 6 HLA-A alleles (in 02:01 [7 patients], 02:05 [1], 02:06 [3], 02:11 [1], 68:01 [2], 68:02 [2]) and 10 HLA-B alleles (07:02 [1], 08:01 [4], 14:01 [1], 14:02 [7], 27:05 [1], 35:02 [1], 35:05 [1], 40:01 [1], 40:02 [3], 45:01 [1]), but were not found in HLA-C alleles. HLA allele frequencies in AA patients, including 271 white Americans, 120 African-Americans, and 99 Hispanics and Latinos, were compared with ethnicity-matched individuals in bone marrow donor datasets of the National Marrow Donor Program, and underwent random-effects meta-analyses. HLA-B*07, B*14, and B*40 were overrepresented in AA, while A*02, A*68, and B*08 frequencies were similar to those of healthy donors (Figure 2). In 164 severe AA patients who were initially treated with horse antithymocyte globulin (hATG), cyclosporine, and eltrombopag between 2012 and 2018, 36 and 79 were positive and negative for HLA- monocytes, respectively, and 49 were not tested by HLA-flow. There was no significant difference in overall and complete response rates at six months among the three groups (Figure 3). Clonal evolution, defined as acquisition of abnormal bone marrow cytogenetics or morphology, especially high-risk evolution to chromosome 7 abnormalities, complex cytogenetics, or morphological MDS/AML, tended to be more frequent in patients with HLA- monocytes, compared to the other two groups, but the difference did not reach statistical significance. Clinical outcomes were also assessed according to the presence of specific HLA alleles in 400 severe AA patients who were treated with hATG-based initial immunosuppressive therapy from 2000 to 2018: there was no significant differences in probabilities of response and clonal evolution according to the alleles associated with somatic mutations. Our study revealed that somatic mutations in HLA genes in AA are broadly distributed, but some alleles are preferentially affected. Inconsistent with previous studies, we found that outcomes of therapy did not significantly correlate with HLA gene mutations or with distinct HLA alleles. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
15. Deficit of Circulating CD19+CD24hiCD38hi Regulatory B Cells in Severe Aplastic Anemia
- Author
-
Lemlem Alemu, Neal S. Young, Sachiko Kajigaya, Yoshitaka Zaimoku, Bhavisha A Patel, Diego Quinones Raffo, Emma M. Groarke, and Xingmin Feng
- Subjects
business.industry ,Regulatory B cells ,Immunology ,Naive B cell ,Cell Biology ,Hematology ,CD38 ,medicine.disease ,Biochemistry ,medicine.anatomical_structure ,Immune system ,Cytotoxic T cell ,Medicine ,Aplastic anemia ,business ,CD8 ,B cell - Abstract
Background: Immune aplastic anemia (AA) is caused by cytotoxic T cells (CTLs) that destroy hematopoietic stem and progenitor cells. Regulatory T cells (Tregs) are reduced in AA and increase in response to immunosuppressive therapy (IST; Solomou E et al, Blood 2007). Recent studies suggested an immune regulatory role of regulatory B cells (Bregs). Human CD19+CD24hiCD38hi Bregs suppress Th1 response of CD4+ T cells as well as IFN-γ production by CD8+ CTLs (Mauri C, Menon M, J Clin Invest 2017). The quantity and/or function of Bregs are impaired in autoimmune diseases, malignancies, chronic graft-versus-host disease, and during rejection of transplanted organs. Methods: We investigated B cell phenotypes including CD24hiCD38hi Bregs in previously untreated severe AA (SAA) and very severe AA (VSAA) patients, and healthy individuals aged 18 years and older, and tested their correlation with severity and response to IST. Absolute numbers of lymphocyte subsets, including CD19+ B cells, CD8+ T cells, CD4+ T cells, and NK cell (TBNK), were quantified in fresh blood. Percentages of B cell subsets among total CD19+ B cells, including CD24hiCD38hi Bregs, CD24loCD38lo mature naïve B cells, CD24hiCD38lo memory B cells and CD24loCD38hi plasma cells/plasmablasts, were analyzed using cryopreserved peripheral blood mononuclear cells (PBMCs). Blood samples were obtained from patients close to time of diagnosis and before institution of definitive therapy. All patients were treated with horse anti-thymocyte globulin, cyclosporine, and eltrombopag between 2012 and 2018 at the Hematology Branch, NHLBI (clinicaltrials.gov NCT01623167). Results: TBNK analysis revealed no significant difference in total B cell counts in 104 AA patients compared to 40 healthy individuals (median, 137/μl [IQR, 73-212] vs 163/μl [106-242], P=.11); NK cells were significantly decreased in patients with AA, as previously reported (Gascon P et al, Blood 1986). Total B cell count did not correlate with severity of AA (P=.89) nor with overall response at six months (P=.93). CD8+ T cells and NK cells were lower in VSAA patients compared to SAA patients. None of the TBNK subsets was predictive of overall response in six months after IST. When we assessed the phenotype of B cells among 60 AA patients whose cryopreserved PBMCs were available, CD24hiCD38hi Bregs were markedly decreased as compared to 29 healthy individuals (0.31% [0.14-0.85%] vs 1.9% [1.3-3.6%], P=3×10-7; Figure, Table), while there was no significant difference in other B cell phenotypes. Among these 60 patients, the percentage of CD24hiCD38hi Bregs was especially decreased in VSAA patients compared to SAA (0.18% [0.11-0.34%] vs 0.50% [0.17-1.4%], P=.017). In contrast, CD24loCD38lo mature naïve B cells were higher in VSAA than in SAA (69% [58-86%] vs 60% [42-70%], P=.024). CD24hiCD38hi Breg frequency was positively associated with neutrophil and reticulocyte counts (correlation coefficients [r], 0.34 and 0.26, respectively), while the frequency of CD24loCD38lo mature naïve B cells was negatively correlated (r, -0.34 and -0.40). CD24loCD38lo mature naïve B cells before IST were significantly lower in 47 patients who achieved overall responses at six months compared to 13 non-responders (64% [42-71%), vs 73% [58-88%], P=.014), but CD24hiCD38hi Breg frequency was not correlated with IST responses. At six months after IST, CD24hiCD38hi Bregs in AA patients had recovered to levels present in healthy individuals (2.3% [0.98-4.8%]), in both 34 responders and five non-responders; non-responders showed non-significant increased CD24loCD38lo mature naïve B cells at six months (P=.068). Discussion: A deficit of circulating CD24hiCD38hi Bregs in immune AA with recovery after IST, as occurs with Tregs, suggests Bregs may contribute to the immune pathophysiology in AA. We unexpectedly observed a higher percentage of CD24loCD38lo mature naïve B cells to be associated with more severe disease and a lower probability of responses to IST. B cell phenotype analysis may be beneficial for monitoring of AA and predicting outcomes of therapy. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.