Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 410
Filter
Add more filters

Publication year range
1.
Blood ; 141(1): 72-89, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36130301

ABSTRACT

Immune aplastic anemia (AA) is a severe blood disease characterized by T-lymphocyte- mediated stem cell destruction. Hematopoietic stem cell transplantation and immunosuppression are effective, but they entail costs and risks, and are not always successful. The Janus kinase (JAK) 1/2 inhibitor ruxolitinib (RUX) suppresses cytotoxic T-cell activation and inhibits cytokine production in models of graft-versus-host disease. We tested RUX in murine immune AA for potential therapeutic benefit. After infusion of lymph node (LN) cells mismatched at the major histocompatibility complex [C67BL/6 (B6)⇒CByB6F1], RUX, administered as a food additive (Rux-chow), attenuated bone marrow hypoplasia, ameliorated peripheral blood pancytopenia, preserved hematopoietic progenitors, and prevented mortality, when used either prophylactically or therapeutically. RUX suppressed the infiltration, proliferation, and activation of effector T cells in the bone marrow and mitigated Fas-mediated apoptotic destruction of target hematopoietic cells. Similar effects were obtained when Rux-chow was fed to C.B10 mice in a minor histocompatibility antigen mismatched (B6⇒C.B10) AA model. RUX only modestly suppressed lymphoid and erythroid hematopoiesis in normal and irradiated CByB6F1 mice. Our data support clinical trials of JAK/STAT inhibitors in human AA and other immune bone marrow failure syndromes.


Subject(s)
Anemia, Aplastic , Bone Marrow Diseases , Pancytopenia , Mice , Humans , Animals , Pancytopenia/pathology , Anemia, Aplastic/pathology , Bone Marrow Failure Disorders/pathology , Bone Marrow/pathology , Bone Marrow Diseases/pathology , Janus Kinase 1
2.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Article in English | MEDLINE | ID: mdl-34417303

ABSTRACT

Sterile α motif domain-containing protein 9-like (SAMD9L) is encoded by a hallmark interferon-induced gene with a role in controlling virus replication that is not well understood. Here, we analyze SAMD9L function from the perspective of human mutations causing neonatal-onset severe autoinflammatory disease. Whole-genome sequencing of two children with leukocytoclastic panniculitis, basal ganglia calcifications, raised blood inflammatory markers, neutrophilia, anemia, thrombocytopaenia, and almost no B cells revealed heterozygous de novo SAMD9L mutations, p.Asn885Thrfs*6 and p.Lys878Serfs*13. These frameshift mutations truncate the SAMD9L protein within a domain a region of homology to the nucleotide-binding and oligomerization domain (NOD) of APAF1, ∼80 amino acids C-terminal to the Walker B motif. Single-cell analysis of human cells expressing green fluorescent protein (GFP)-SAMD9L fusion proteins revealed that enforced expression of wild-type SAMD9L repressed translation of red fluorescent protein messenger RNA and globally repressed endogenous protein translation, cell autonomously and in proportion to the level of GFP-SAMD9L in each cell. The children's truncating mutations dramatically exaggerated translational repression even at low levels of GFP-SAMD9L per cell, as did a missense Arg986Cys mutation reported recurrently as causing ataxia pancytopenia syndrome. Autoinflammatory disease associated with SAMD9L truncating mutations appears to result from an interferon-induced translational repressor whose activity goes unchecked by the loss of C-terminal domains that may normally sense virus infection.


Subject(s)
Ataxia/pathology , Gene Expression Regulation , Mutation, Missense , Myelodysplastic Syndromes/pathology , Pancytopenia/pathology , Protein Biosynthesis , Tumor Suppressor Proteins/genetics , Ataxia/genetics , Child , Female , Heterozygote , Humans , Infant, Newborn , Male , Myelodysplastic Syndromes/genetics , Pancytopenia/genetics
3.
Blood ; 137(18): 2450-2462, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33512449

ABSTRACT

Inborn errors of immunity (IEI) are a genetically heterogeneous group of disorders with a broad clinical spectrum. Identification of molecular and functional bases of these disorders is important for diagnosis, treatment, and an understanding of the human immune response. We identified 6 unrelated males with neutropenia, infections, lymphoproliferation, humoral immune defects, and in some cases bone marrow failure associated with 3 different variants in the X-linked gene TLR8, encoding the endosomal Toll-like receptor 8 (TLR8). Interestingly, 5 patients had somatic variants in TLR8 with <30% mosaicism, suggesting a dominant mechanism responsible for the clinical phenotype. Mosaicism was also detected in skin-derived fibroblasts in 3 patients, demonstrating that mutations were not limited to the hematopoietic compartment. All patients had refractory chronic neutropenia, and 3 patients underwent allogeneic hematopoietic cell transplantation. All variants conferred gain of function to TLR8 protein, and immune phenotyping demonstrated a proinflammatory phenotype with activated T cells and elevated serum cytokines associated with impaired B-cell maturation. Differentiation of myeloid cells from patient-derived induced pluripotent stem cells demonstrated increased responsiveness to TLR8. Together, these findings demonstrate that gain-of-function variants in TLR8 lead to a novel childhood-onset IEI with lymphoproliferation, neutropenia, infectious susceptibility, B- and T-cell defects, and in some cases, bone marrow failure. Somatic mosaicism is a prominent molecular mechanism of this new disease.


Subject(s)
Bone Marrow Failure Disorders/pathology , Gain of Function Mutation , Immunologic Deficiency Syndromes/pathology , Inflammation/pathology , Mosaicism , Pancytopenia/pathology , Toll-Like Receptor 8/genetics , Adolescent , Adult , B-Lymphocytes/pathology , Bone Marrow Failure Disorders/etiology , Bone Marrow Failure Disorders/metabolism , Cell Differentiation , Child , Child, Preschool , Cytokines/metabolism , Female , Follow-Up Studies , Humans , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/metabolism , Infant , Inflammation/etiology , Inflammation/metabolism , Lymphocyte Activation , Male , Pancytopenia/etiology , Pancytopenia/metabolism , Pedigree , Prognosis , T-Lymphocytes/immunology , Young Adult
4.
Eur J Haematol ; 107(3): 354-363, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34076909

ABSTRACT

INTRODUCTION: Allogeneic hematopoietic cell transplantation (HCT) can be curative for acute myeloid leukemia (AML). Novel therapies may render patients' bone marrow hypocellularity and lead to prolonged post-therapy pancytopenia. Patients' bone marrow cellularity (BMC) at pretransplant assessment and post-treatment pancytopenia (classification CR-incomplete [CRi]) may manifest AML persistence. METHODOLOGY: We retrospectively examined the impact of BMC and ELN response (ELNr) on a single-center cohort of 337 patients who underwent allogeneic HCT for AML in CR1. RESULTS: Median follow-up was 33 months. Overall survival (OS) for the whole cohort was 55.8% at 2 years, while cumulative incidence of relapse (CIR) was 20.8%, and non-relapse mortality was 27.5%. OS and CIR were not significantly different between BMC groups; and neither was ELNr. ELNr CRi was associated with BMC aplastic and hypocellular marrow states (P < 2.6e-8). Multivariate analysis confirmed neither BMC nor attainment of ELNr CR vs CRi affected OS or relapse. Significant factors for survival included age at transplant, cytogenetic risk, development of acute Gr II-IV GvHD, and moderate-severe chronic GvHD, while cytogenetic risk and chronic GvHD affected relapse. CONCLUSION: Neither ELNr status nor pretransplant BMC influenced relapse post-HCT or OS. Hypocellularity and CRi are not negative prognostic factors for post-HCT outcomes of AML.


Subject(s)
Bone Marrow Cells/pathology , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/therapy , Pancytopenia/pathology , Transplantation, Homologous/methods , Adolescent , Adult , Age Factors , Aged , Bone Marrow Cells/immunology , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Multivariate Analysis , Myeloablative Agonists/administration & dosage , Myeloablative Agonists/adverse effects , Pancytopenia/etiology , Pancytopenia/immunology , Pancytopenia/mortality , Recurrence , Retrospective Studies , Survival Analysis , Transplantation Conditioning
5.
Pediatr Blood Cancer ; 68(4): e28893, 2021 04.
Article in English | MEDLINE | ID: mdl-33484072

ABSTRACT

Cytopenias are common among neonates in neonatal intensive care units (NICU). Although, bone marrow aspirations (BMAs) are often performed as part of diagnostic workup, but trephine bone marrow biopsies (BMBs) have not been reported from living neonates. BMB is indispensable to accurately assess the cellularity and architecture. There is paucity of literature regarding the technique of BMB in neonates. In this report, for the first time, we describe trephine BMB from posterior superior iliac crest (PSIC) using 18-gauge BMA needle in six living neonates admitted to NICU where BMB findings helped in understanding the underlying mechanism and diagnosis of cytopenias.


Subject(s)
Biopsy/methods , Bone Marrow/pathology , Hematologic Diseases/diagnosis , Anemia/diagnosis , Anemia/pathology , Anemia, Diamond-Blackfan/diagnosis , Anemia, Diamond-Blackfan/pathology , Hematologic Diseases/pathology , Humans , Infant , Pancytopenia/diagnosis , Pancytopenia/pathology , Thrombocytopenia/diagnosis , Thrombocytopenia/pathology
6.
Pediatr Blood Cancer ; 68(2): e28799, 2021 02.
Article in English | MEDLINE | ID: mdl-33200495

ABSTRACT

Pearson syndrome (PS) is a very rare and often fatal multisystem disease caused by deletions in mitochondrial DNA that result in sideroblastic anemia, vacuolization of marrow precursors, and pancreatic dysfunction. Spontaneous recovery from anemia is often observed within several years of diagnosis. We present the case of a 4-month-old male diagnosed with PS who experienced prolonged severe pancytopenia preceding the emergence of monosomy 7. Whole-exome sequencing identified two somatic mutations, including RUNX1 p.S100F that was previously reported as associated with myeloid malignancies. The molecular defects associated with PS may have the potential to progress to advanced myelodysplastic syndrome .


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , Congenital Bone Marrow Failure Syndromes/therapy , Core Binding Factor Alpha 2 Subunit/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/therapy , Membrane Proteins/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/therapy , Muscular Diseases/genetics , Muscular Diseases/therapy , Nerve Tissue Proteins/genetics , Blood Transfusion , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , DNA, Mitochondrial/genetics , Genetic Predisposition to Disease/genetics , Humans , Infant , Male , Pancytopenia/genetics , Pancytopenia/pathology , Exome Sequencing
7.
Blood Cells Mol Dis ; 80: 102373, 2020 02.
Article in English | MEDLINE | ID: mdl-31718920

ABSTRACT

Gaucher disease is an inherited lysosomal storage disease commonly associated with hepatosplenomegaly and cytopenias. Progressive cytopenias may be interpreted as an indication of advanced disease and suggest the need to start Gaucher disease specific treatment. As bone marrow evaluation is not routinely performed in Gaucher disease, other causes of cytopenias such as myelodysplastic syndrome a stem cell disorder may be missed. Six patients are described who suffered simultaneously from Gaucher disease and myelodysplastic syndrome, with a discussion of the diagnostic challenges.


Subject(s)
Gaucher Disease/complications , Gaucher Disease/diagnosis , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/diagnosis , Adult , Aged , Aged, 80 and over , Bone Marrow/pathology , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Pancytopenia/pathology
8.
Int J Med Sci ; 17(12): 1665-1672, 2020.
Article in English | MEDLINE | ID: mdl-32714069

ABSTRACT

Aplastic anemia (AA) is a rare disorder characterized by the suppression of bone marrow function resulting in progressive pancytopenia. The pathogenesis of AA is complex and involves an abnormal hematopoietic microenvironment, hematopoietic stem cell/progenitor cell deficiencies, and immunity disorders. However, the underlying mechanism of the disease is still not fully uncovered. In this research, we collected both donor and patient samples and found suppressed proliferation, abnormal differentiation as well as increased apoptosis of patient mesenchymal stem cells (MSCs). Considering the close relationship of parathyroid hormone (PTH) and MSCs differentiation, further studies showed that although patients maintained normal serum PTH level, their CD8+ T cells possessed lower PTH receptors. The insensitive to PTH of patients' CD8+ T cells finally lead to reduced expression of key Wnt factors. In all, bone marrow CD8+ T cells may play an important role in inducing MSCs adipogenesis and osteogenesis imbalancement.


Subject(s)
Anemia, Aplastic/genetics , Mesenchymal Stem Cells/metabolism , Pancytopenia/genetics , Parathyroid Hormone/genetics , Adipogenesis/genetics , Adolescent , Anemia, Aplastic/pathology , Apoptosis/genetics , Bone Marrow/immunology , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cellular Microenvironment/genetics , Child , Female , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Male , Mesenchymal Stem Cells/immunology , Osteogenesis/genetics , Pancytopenia/immunology , Pancytopenia/pathology , Parathyroid Hormone/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Am J Hum Genet ; 98(6): 1146-1158, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27259050

ABSTRACT

Ataxia-pancytopenia (AP) syndrome is characterized by cerebellar ataxia, variable hematologic cytopenias, and predisposition to marrow failure and myeloid leukemia, sometimes associated with monosomy 7. Here, in the four-generation family UW-AP, linkage analysis revealed four regions that provided the maximal LOD scores possible, one of which was in a commonly microdeleted chromosome 7q region. Exome sequencing identified a missense mutation (c.2640C>A, p.His880Gln) in the sterile alpha motif domain containing 9-like gene (SAMD9L) that completely cosegregated with disease. By targeted sequencing of SAMD9L, we subsequently identified a different missense mutation (c.3587G>C, p.Cys1196Ser) in affected members of the first described family with AP syndrome, Li-AP. Neither variant is reported in the public databases, both affect highly conserved amino acid residues, and both are predicted to be damaging. With time in culture, lymphoblastic cell lines (LCLs) from two affected individuals in family UW-AP exhibited copy-neutral loss of heterozygosity for large portions of the long arm of chromosome 7, resulting in retention of only the wild-type SAMD9L allele. Newly established LCLs from both individuals demonstrated the same phenomenon. In addition, targeted capture and sequencing of SAMD9L in uncultured blood DNA from both individuals showed bias toward the wild-type allele. These observations indicate in vivo hematopoietic mosaicism. The hematopoietic cytopenias that characterize AP syndrome and the selective advantage for clones that have lost the mutant allele support the postulated role of SAMD9L in the regulation of cell proliferation. Furthermore, we show that AP syndrome is distinct from the dyskeratoses congenita telomeropathies, with which it shares some clinical characteristics.


Subject(s)
Cerebellar Ataxia/genetics , Chromosome Aberrations , Mutation, Missense/genetics , Pancytopenia/genetics , Proteins/genetics , Adolescent , Adult , Cerebellar Ataxia/pathology , Child , Chromosomes, Human, Pair 7/genetics , Exome/genetics , Female , Genetic Linkage , Genotype , High-Throughput Nucleotide Sequencing/methods , Humans , Intracellular Signaling Peptides and Proteins , Loss of Heterozygosity , Male , Middle Aged , Pancytopenia/pathology , Pedigree , Tumor Suppressor Proteins/genetics , Young Adult
10.
Am J Pathol ; 188(6): 1457-1468, 2018 06.
Article in English | MEDLINE | ID: mdl-29574182

ABSTRACT

The fundamental structure of eukaryotic cell plasma membrane is the phospholipid bilayer, which contains four major phospholipids. These phospholipids are asymmetrically distributed between the outer and inner leaflets. P4-ATPase flippase complexes play essential roles in ensuring this asymmetry. We found that conditional deletion of Tmem30a, the ß subunit of P4-ATPase flippase complex, caused pancytopenia in mice. Tmem30a deficiency resulted in depletion of lineage-committed blood cells in the peripheral blood, spleen, and bone marrow. Ablation of Tmem30a also caused the depletion of hematopoietic stem cells (HSCs). HSC RNA sequencing results revealed that multiple biological processes and signal pathways were involved in the event, including mammalian target of rapamycin signaling, genes for HSC stemness, and genes responding to interferons. Our results also revealed that targeting Tmem30a signaling had therapeutic utility in BCR/ABL1-induced chronic myeloid leukemia.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Proteins/physiology , Pancytopenia/pathology , Proto-Oncogene Proteins c-bcr/metabolism , Animals , Cells, Cultured , Hematopoietic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Knockout , Pancytopenia/etiology , Pancytopenia/metabolism , Signal Transduction
11.
Haematologica ; 104(11): 2164-2177, 2019 11.
Article in English | MEDLINE | ID: mdl-30923091

ABSTRACT

Loss-of-function studies have determined that Notch signaling is essential for hematopoietic and endothelial development. By deleting a single allele of the Notch1 transcriptional activation domain we generated viable, post-natal mice exhibiting hypomorphic Notch signaling. These heterozygous mice, which lack only one copy of the transcriptional activation domain, appear normal and have no endothelial or hematopoietic phenotype, apart from an inherent, cell-autonomous defect in T-cell lineage development. Following chemotherapy, these hypomorphs exhibited severe pancytopenia, weight loss and morbidity. This phenotype was confirmed in an endothelial-specific, loss-of-function Notch1 model system. Ang1, secreted by hematopoietic progenitors after damage, activated endothelial Tie2 signaling, which in turn enhanced expression of Notch ligands and potentiated Notch1 receptor activation. In our heterozygous, hypomorphic model system, the mutant protein that lacks the Notch1 transcriptional activation domain accumulated in endothelial cells and interfered with optimal activity of the wildtype Notch1 transcriptional complex. Failure of the hypomorphic mutant to efficiently drive transcription of key gene targets such as Hes1 and Myc prolonged apoptosis and limited regeneration of the bone marrow niche. Thus, basal Notch1 signaling is sufficient for niche development, but robust Notch activity is required for regeneration of the bone marrow endothelial niche and hematopoietic recovery.


Subject(s)
Cellular Microenvironment , Endothelial Cells/physiology , Receptor, Notch1/metabolism , Receptor, TIE-2/metabolism , Regeneration , Signal Transduction , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cellular Microenvironment/drug effects , Endothelial Cells/drug effects , Fluorouracil/pharmacology , Gamma Rays/adverse effects , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Pancytopenia/etiology , Pancytopenia/metabolism , Pancytopenia/pathology , Signal Transduction/drug effects
12.
Curr Oncol Rep ; 21(10): 87, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31414187

ABSTRACT

PURPOSE OF REVIEW: Discuss the pathophysiology, clinical presentation, diagnosis, and treatment of immune-mediated cytopenias (IMC) after hematopoietic cell transplantation (HCT). RECENT FINDINGS: Key risk factors for post-HCT IMC include younger age, non-malignant disease, and umbilical cord blood stem cell source. While anemia predominates, any or all three hematopoietic cell lines can be affected. In rare cases, IMC can cause graft failure or death. IMC is hypothesized to result from immune dysregulation upon reconstitution of donor hematopoietic cells (i.e., dysfunctional regulatory T cells). Aside from blood product transfusions, IMC treatment includes immune-suppressive or ablative agents. First-line therapies, including corticosteroids and intravenous immunoglobulin, are often inadequate, prompting use of additional agents aimed at antibody production/T cell dysfunction or direct antibody removal via plasmapheresis. IMC occurs in up to 20% of high-risk HCT populations. Morbidity and mortality from IMC post-HCT have been reduced by improved recognition and aggressive early interventions.


Subject(s)
Anemia, Hemolytic, Autoimmune/therapy , Graft vs Host Disease/therapy , Hematologic Diseases/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Pancytopenia/therapy , Thrombocytopenia/therapy , Anemia, Hemolytic, Autoimmune/etiology , Anemia, Hemolytic, Autoimmune/pathology , Graft vs Host Disease/etiology , Graft vs Host Disease/pathology , Hematologic Diseases/immunology , Hematologic Diseases/pathology , Hematopoietic Stem Cell Transplantation/methods , Humans , Pancytopenia/etiology , Pancytopenia/pathology , Prognosis , Thrombocytopenia/etiology , Thrombocytopenia/pathology
13.
J Gastroenterol Hepatol ; 34(8): 1417-1423, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30462857

ABSTRACT

BACKGROUND AND AIM: Idiopathic portal hypertension (IPH) refers to a relatively rare condition characterized by intrahepatic portal hypertension in the absence of underlying disease such as liver cirrhosis. METHODS: We retrospectively reviewed 338 patients with IPH that were diagnosed at the pathological consultation center of our hospital. RESULTS: The ratio of male to female patients was 1:1. Mean age at onset was 35.1 ± 16.5 years; male patients on average were 12 years younger than female patients at onset. The median duration from onset to IPH diagnosis was 12 months. In 50 patients, medication use may have been an etiological factor. The most common clinical manifestations were splenomegaly (91.3%) and hypersplenism (68.9%); 57.0% patients presented varicosis, while 25.1% patients had a history of variceal bleeding. Nodular regenerative hyperplasia was found in 22.2% liver biopsies. Among patients for whom laboratory data were available, 65.0%, 50.3%, and 71.4% patients presented leukopenia, anemia, and thrombocytopenia due to hypersplenism. Liver function was mostly in the compensated stage. Female patients showed worse leukopenia and anemia, while male patients were more likely to have abnormal serum transaminase and bilirubin levels. Sixty-seven patients received surgical or interventional treatment. CONCLUSIONS: High-quality liver biopsy, detailed clinical information, and expert pathologist are necessary for diagnosis of IPH. IPH can occur concurrently with other liver disease such as hepatitis and drug-induced liver injury. Medication appears to be an important etiological factor for IPH in China. Management approach was largely focused on treatment of portal hypertension and its complications.


Subject(s)
Hypertension, Portal/pathology , Liver Cirrhosis/pathology , Liver/pathology , Pancytopenia/pathology , Splenomegaly/pathology , Adolescent , Adult , Biopsy , China/epidemiology , Female , Humans , Hypertension, Portal/epidemiology , Hypertension, Portal/physiopathology , Hypertension, Portal/therapy , Liver Cirrhosis/epidemiology , Liver Cirrhosis/physiopathology , Liver Cirrhosis/therapy , Liver Function Tests , Male , Middle Aged , Pancytopenia/epidemiology , Pancytopenia/physiopathology , Pancytopenia/therapy , Portal Pressure , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Factors , Splenomegaly/epidemiology , Splenomegaly/physiopathology , Splenomegaly/therapy , Young Adult , Idiopathic Noncirrhotic Portal Hypertension
14.
J Pediatr Hematol Oncol ; 41(6): 473-477, 2019 08.
Article in English | MEDLINE | ID: mdl-31233462

ABSTRACT

Griscelli syndrome type 2 (GS2) is an autosomal recessive condition associated with the development of hemophagocytic lymphohistiocytosis. GS2 is caused by a gene mutation involving RAB27A, which affects a melanosome anchoring complex in melanocytes and releases cytolytic granules from T cells and natural killer cells. GS2 is known to have immunologic compromise and oculocutaneous albinism. We present the case of 2 sisters who had vastly different phenotypic presentations despite having the same genetic frameshift mutation in the RAB27A gene. Patient 1 presented with seizures and neurological compromise, whereas patient 2 presented with pancytopenia and diarrhea. Both patients developed hemophagocytic lymphohistiocytosis.


Subject(s)
Diarrhea/pathology , Frameshift Mutation , Lymphohistiocytosis, Hemophagocytic/pathology , Pancytopenia/pathology , Piebaldism/pathology , Primary Immunodeficiency Diseases/pathology , Seizures/pathology , rab27 GTP-Binding Proteins/genetics , Child, Preschool , Diarrhea/complications , Diarrhea/genetics , Female , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/genetics , Pancytopenia/complications , Pancytopenia/genetics , Piebaldism/complications , Piebaldism/genetics , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/genetics , Prognosis , Seizures/complications , Seizures/genetics , Siblings
15.
Blood ; 127(3): 310-3, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26644450

ABSTRACT

BRCA1 is critical for maintenance of genomic stability and interacts directly with several proteins that regulate hematopoietic stem cell function and are part of the Fanconi anemia (FA) double-strand break DNA repair pathway. The effects of complete BRCA1 deficiency on bone marrow (BM) function are unknown. To test the hypothesis that Brca1 is essential in hematopoiesis, we developed a conditional mouse model with Mx1-Cre-mediated Brca1 deletion. Mice lacking Brca1 in the BM have baseline cytopenias and develop spontaneous bone marrow failure or diverse hematologic malignancies by 6 months of age. Brca1(-/-) BM cells have a reduced capacity to form hematopoietic colonies in vitro and to reconstitute hematopoiesis in irradiated recipients, consistent with a hematopoietic progenitor functional defect. Brca1(-/-) BM cells also show FA-like hypersensitivity to the DNA crosslinking agent mitomycin C, and karyotypes feature genomic instability. Taken together, our results show that loss of Brca1 in murine BM causes hematopoietic defects similar to those seen in people with FA, which provides strong evidence that Brca1 is critical for normal hematopoiesis and that Brca1 is a bona fide FA-like gene.


Subject(s)
BRCA1 Protein/deficiency , BRCA1 Protein/genetics , Hematologic Neoplasms/genetics , Hemoglobinuria, Paroxysmal/genetics , Anemia, Aplastic , Animals , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Diseases , Bone Marrow Failure Disorders , Disease Models, Animal , Genomic Instability , Hematologic Neoplasms/diagnosis , Hematopoiesis/genetics , Hemoglobinuria, Paroxysmal/diagnosis , Humans , Leukemic Infiltration , Leukocyte Count , Mice , Mice, Knockout , Pancytopenia/genetics , Pancytopenia/pathology , Phenotype
16.
Haematologica ; 103(6): 931-938, 2018 06.
Article in English | MEDLINE | ID: mdl-29748447

ABSTRACT

Chronic lymphocytic leukemia is an extremely heterogeneous disease and prognostic factors such as chromosomal abnormalities are important predictors of time to first treatment and survival. Trisomy 12 is the second most frequent aberration detected by fluorescence in situ hybridization at the time of diagnosis (10-25%), and it confers an intermediate prognostic risk, with a median time to first treatment of 33 months and a median overall survival of 114 months. Here, we review the unique morphological, immunophenotypic, and genetic characteristics of patients with chronic lymphocytic leukemia and trisomy 12. These patients carry a significantly higher expression of CD19, CD22, CD20, CD79b, CD24, CD27, CD38, CD49d, sIgM, sIgk, and sIgλ and lower expression of CD43 compared with patients with normal karyotype. Circulating cells show increased expression of the integrins CD11b, CD18, CD29, and ITGB7, and of the adhesion molecule CD323. Patients with chronic lymphocytic leukemia and trisomy 12 frequently have unmutated IGHV, ZAP-70 positivity, and closely homologous stereotyped B-cell receptors. They rarely show TP53 mutations but frequently have NOTCH1 mutations, which can be identified in up to 40% of those with a rapidly progressive clinical course.


Subject(s)
Chromosomes, Human, Pair 12 , Immunophenotyping , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Trisomy , Biomarkers , Bone Marrow/pathology , Chromosome Aberrations , Combined Modality Therapy , Genetic Association Studies , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunophenotyping/methods , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Pancytopenia/pathology , Phenotype , Prognosis , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Treatment Outcome
18.
Pediatr Blood Cancer ; 65(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-28868793

ABSTRACT

Diagnosis of bone marrow failure (BMF) disorders is challenging but essential for optimal patient management. Here, we report a young adult from nonconsanguineous parents with progressive pancytopenia since childhood, bone pain, increased bone density, and haphazard ossification replacing hematopoiesis within the bone marrow. Sequencing revealed two novel biallelic variants of unknown significance within the thromboxane A synthase gene, TBXAS1 (c.266T > C; c.989T > C), bioinformatically predicted to disrupt the protein. TBXAS1 mutations result in Ghosal hematodiaphyseal dysplasia (OMIM 231095), the autosomal recessive syndrome associated with abnormal bone structure and BMF. Identification of the genetic defect prompted steroid therapy leading to resolution of symptoms.


Subject(s)
Anemia, Refractory , Bone Density/genetics , Osteochondrodysplasias , Pancytopenia , Point Mutation , Thromboxane-A Synthase/deficiency , Anemia, Refractory/enzymology , Anemia, Refractory/genetics , Anemia, Refractory/pathology , Chronic Disease , Female , Humans , Infant , Osteochondrodysplasias/enzymology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Pancytopenia/enzymology , Pancytopenia/genetics , Pancytopenia/pathology
19.
Haematologica ; 102(10): 1691-1703, 2017 10.
Article in English | MEDLINE | ID: mdl-28729300

ABSTRACT

Acquired aplastic anemia, the prototypical bone marrow failure disease, is characterized by pancytopenia and marrow hypoplasia. Most aplastic anemia patients respond to immunosuppressive therapy, usually with anti-thymocyte globulin and cyclosporine, but some relapse on cyclosporine withdrawal or require long-term administration of cyclosporine to maintain blood counts. In this study, we tested efficacy of rapamycin as a new or alternative treatment in mouse models of immune-mediated bone marrow failure. Rapamycin ameliorated pancytopenia, improved bone marrow cellularity, and extended animal survival in a manner comparable to the standard dose of cyclosporine. Rapamycin effectively reduced Th1 inflammatory cytokines interferon-γ and tumor necrosis factor-α, increased the Th2 cytokine interleukin-10, stimulated expansion of functional regulatory T cells, eliminated effector CD8+ T cells (notably T cells specific to target cells bearing minor histocompatibility antigen H60), and preserved hematopoietic stem and progenitor cells. Rapamycin, but not cyclosporine, reduced the proportion of memory and effector T cells and maintained a pool of naïve T cells. Cyclosporine increased cytoplasmic nuclear factor of activated T-cells-1 following T-cell receptor stimulation, whereas rapamycin suppressed phosphorylation of two key signaling molecules in the mammalian target of rapamycin pathway, S6 kinase and protein kinase B. In summary, rapamycin was an effective therapy in mouse models of immune-mediated bone marrow failure, acting through different mechanisms to cyclosporine. Its specific expansion of regulatory T cells and elimination of clonogenic CD8+ effectors support its potential clinical utility in the treatment of aplastic anemia.


Subject(s)
Anemia, Aplastic/immunology , Anemia, Aplastic/pathology , Bone Marrow Diseases/immunology , Bone Marrow Diseases/pathology , Bone Marrow/immunology , Bone Marrow/pathology , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/pathology , Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , Anemia, Aplastic/drug therapy , Anemia, Aplastic/metabolism , Anemia, Aplastic/mortality , Animals , Bone Marrow/drug effects , Bone Marrow Diseases/drug therapy , Bone Marrow Diseases/mortality , Bone Marrow Failure Disorders , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/mortality , Immunologic Memory , Mice , Pancytopenia/immunology , Pancytopenia/pathology , Signal Transduction , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Treatment Outcome
20.
Ann Hematol ; 96(8): 1287-1295, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28601896

ABSTRACT

Myelodysplastic syndromes (MDS) represent a heterogeneous group of hematologic disorders characterized by cytopenia(s) and predisposition to leukemic progression. An immune dysregulation and an aberrant bone marrow microenvironment seem to be key elements in the physiopathological process of MDS. In order to evaluate a possible association between susceptibility and clinic-pathologic features, we genotyped 153 MDS patients for functional cytokine polymorphisms: TNF (-308 G/A), IFNG (+874 A/T and +875 CAn), IL6 (-174 G/C), and TGFB1 (+869 C/T and +915 G/C). The frequency of TNF and IL6 polymorphisms was different between patients and healthy controls (n = 131), suggesting a relatedness to MDS susceptibility in our population. Furthermore, the presence of each or both high-producing genotypes [TNF: p = 0.048, odds ratio (OR): 3.979; IL6: p = 0.001, OR: 6.835; both: p = 0.010, OR: 6.068] and thrombocytopenia at platelet counts of <50,000/µL (p = 0.004, OR: 4.857) were independently associated with an increased risk of manifesting a hemoglobin level of <8 g/dL at diagnosis. In particular, a severe bicytopenia was more frequently observed in patients with the TNF (high)_IL6 (high) combined genotype (p = 0.004, OR: 8.357), who consistently became transfusion dependent earlier (2.9 vs. 34.6 months; p = 0.001); and this likelihood was more evident in patients with lower bone marrow blast counts. The contribution of the remaining functional polymorphisms to the disease phenotype was less relevant. Our results demonstrate that TNF and IL6 gene polymorphisms, as underlying host features, are likely to play a key role in influencing the severity of the cytopenias in MDS and they may be instrumental for tailoring cytokine-target therapies.


Subject(s)
Genetic Predisposition to Disease/genetics , Interleukin-6/genetics , Myelodysplastic Syndromes/genetics , Pancytopenia/complications , Polymorphism, Single Nucleotide , Tumor Necrosis Factor-alpha/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Argentina , Female , Gene Frequency , Genotype , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Myelodysplastic Syndromes/complications , Pancytopenia/pathology , Prospective Studies , Retrospective Studies , Severity of Illness Index , Thrombocytopenia/complications , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL