Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Curr Opin Hematol ; 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39045882

ABSTRACT

PURPOSE OF REVIEW: Atypical chemokine receptor-1 (ACKR1)/Duffy antigen receptor of chemokines (DARC)-associated neutropenia (ADAN; OMIM 611862), previously named benign ethnic neutropenia, and present in two-thirds of individuals identifying as Black in the USA, is associated with mild to moderate decreases in peripheral neutrophil counts that nevertheless do not lead to increased infections. Consequently, recent initiatives have sought to establish normal neutrophil count reference ranges for ADAN, considering it a normal variant rather than a clinical disorder requiring medical intervention. RECENT FINDINGS: A limited number of studies elucidating the mechanism of neutropenia in ADAN has suggested that neutrophils may redistribute from peripheral blood to the tissues including the spleen: this might explain why ADAN is not associated with increased risks of infection since the total number of neutrophils in the body remains normal. In this review, we critically examine the research underlying the molecular basis of ADAN. SUMMARY: Insights into the biology of neutrophils and their trafficking may inform the clinical interpretation of neutropenia in ADAN. The bulk of research suggests that ADAN does not lead to a diminished host defense as do other forms of neutropenia. However, ADAN may lead to increased proinflammatory signaling, with possible implications for senescence of the immune system and predisposition to autoimmunity and cancer.

3.
Lancet Haematol ; 11(5): e368-e382, 2024 May.
Article in English | MEDLINE | ID: mdl-38697731

ABSTRACT

Diamond-Blackfan anaemia (DBA), first described over 80 years ago, is a congenital disorder of erythropoiesis with a predilection for birth defects and cancer. Despite scientific advances, this chronic, debilitating, and life-limiting disorder continues to cause a substantial physical, psychological, and financial toll on patients and their families. The highly complex medical needs of affected patients require specialised expertise and multidisciplinary care. However, gaps remain in effectively bridging scientific discoveries to clinical practice and disseminating the latest knowledge and best practices to providers. Following the publication of the first international consensus in 2008, advances in our understanding of the genetics, natural history, and clinical management of DBA have strongly supported the need for new consensus recommendations. In 2014 in Freiburg, Germany, a panel of 53 experts including clinicians, diagnosticians, and researchers from 27 countries convened. With support from patient advocates, the panel met repeatedly over subsequent years, engaging in ongoing discussions. These meetings led to the development of new consensus recommendations in 2024, replacing the previous guidelines. To account for the diverse phenotypes including presentation without anaemia, the panel agreed to adopt the term DBA syndrome. We propose new simplified diagnostic criteria, describe the genetics of DBA syndrome and its phenocopies, and introduce major changes in therapeutic standards. These changes include lowering the prednisone maintenance dose to maximum 0·3 mg/kg per day, raising the pre-transfusion haemoglobin to 9-10 g/dL independent of age, recommending early aggressive chelation, broadening indications for haematopoietic stem-cell transplantation, and recommending systematic clinical surveillance including early colorectal cancer screening. In summary, the current practice guidelines standardise the diagnostics, treatment, and long-term surveillance of patients with DBA syndrome of all ages worldwide.


Subject(s)
Anemia, Diamond-Blackfan , Consensus , Humans , Anemia, Diamond-Blackfan/diagnosis , Anemia, Diamond-Blackfan/therapy , Anemia, Diamond-Blackfan/genetics , Disease Management , Hematopoietic Stem Cell Transplantation
4.
Semin Hematol ; 59(1): 30-37, 2022 01.
Article in English | MEDLINE | ID: mdl-35491056

ABSTRACT

Fanconi anemia, telomeropathies and ribosomopathies are members of the inherited bone marrow failure syndromes, rare genetic disorders that lead to failure of hematopoiesis, developmental abnormalities, and cancer predisposition. While each disorder is caused by different genetic defects in seemingly disparate processes of DNA repair, telomere maintenance, or ribosome biogenesis, they appear to lead to a common pathway characterized by premature senescence of hematopoietic stem cells. Here we review the experimental data on senescence and inflammation underlying marrow failure and malignant transformation. We conclude with a critical assessment of current and future therapies targeting these pathways in inherited bone marrow failure syndromes patients.


Subject(s)
Anemia, Aplastic , Bone Marrow Diseases , Fanconi Anemia , Anemia, Aplastic/therapy , Bone Marrow Diseases/genetics , Bone Marrow Diseases/therapy , Cellular Senescence/genetics , Congenital Bone Marrow Failure Syndromes , Fanconi Anemia/genetics , Fanconi Anemia/therapy , Humans
5.
Exp Biol Med (Maywood) ; 247(4): 330-337, 2022 02.
Article in English | MEDLINE | ID: mdl-35068219

ABSTRACT

Cytokine storm is an umbrella term that describes an inflammatory syndrome characterized by elevated levels of circulating cytokines and hyperactivation of innate and/or adaptive immune cells. One type of cytokine storm is hemophagocytic lymphohistiocytosis (HLH), which can be either primary or secondary. Severe COVID-19-associated pneumonia and acute respiratory distress syndrome (ARDS) can also lead to cytokine storm/cytokine release syndrome (CS/CRS) and, more rarely, meet criteria for the diagnosis of secondary HLH. Here, we review the immunobiology of primary and secondary HLH and examine whether COVID-19-associated CS/CRS can be discriminated from non-COVID-19 secondary HLH. Finally, we review differences in immunobiology between these different entities, which may inform both clinical diagnosis and treatment of patients.


Subject(s)
COVID-19/complications , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/therapy , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/etiology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Cytokine Release Syndrome/virology , Humans , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/therapy
6.
Exp Biol Med (Maywood) ; 246(1): 5-9, 2021 01.
Article in English | MEDLINE | ID: mdl-32972235

ABSTRACT

IMPACT STATEMENT: Severe COVID-19 associated pneumonia and acute respiratory distress syndrome has recently been described with life-threatening features of cytokine storm and loosely referred to as hemophagocytic lymphohistiocytosis (HLH) or macrophage activation syndrome (MAS). Although a recent report indicated favorable responses to the interleukin-1 receptor antagonist, anakinra in eight patients with COVID-19 secondary HLH diagnosed using the HScore calculation, others have suggested that the diagnosis of secondary HLH is uncommon and that the use of the HScore has limited value in guiding immunomodulatory therapy for COVID-19. Here, we provide additional perspective on this important controversy based upon comparisons between 14 COVID-19 cytokine storm patients and 10 secondary HLH patients seen immediately prior to the pandemic. We hypothesize that identification of HLH may relate to the severity or timing of cytokine release and suggest distinguishing between cytokine release syndrome and secondary HLH, reserving the latter term for cases fulfilling diagnostic criteria.


Subject(s)
COVID-19/complications , Cytokine Release Syndrome/etiology , Lymphohistiocytosis, Hemophagocytic/etiology , Adult , Aged , Aged, 80 and over , Female , Humans , Lymphohistiocytosis, Hemophagocytic/virology , Male , Middle Aged , Splenomegaly/etiology
7.
BMJ Case Rep ; 20182018 Feb 05.
Article in English | MEDLINE | ID: mdl-29437728

ABSTRACT

A 67-year-old right-handed woman presented with dysarthria, left upper extremity weakness and right-sided neglect of 3 hours duration. Imaging of the brain revealed acute right middle cerebral artery stroke; however, tissue plasminogen activator could not be administered due to severe thrombocytopenia. A peripheral smear revealed schistocytes and the patient was treated empirically for thrombotic thrombocytopenic purpura (TTP) with therapeutic plasma exchange. An extensive workup revealed no embolic source or other cause for stroke, and a diagnosis of large vessel infarct secondary to TTP was made. After a prolonged hospital course, the patient had partial neurological recovery and was discharged to a rehabilitation facility. Although transient neurologic deficits due to small vessel occlusions are well described in TTP, large vessel infarct can occur as well. This diagnosis should be considered in patients presenting with concomitant stroke and thrombocytopenia, as untreated TTP is nearly always fatal.


Subject(s)
Dysarthria/diagnosis , Purpura, Thrombotic Thrombocytopenic/diagnosis , Stroke/diagnosis , Adrenal Cortex Hormones/therapeutic use , Aged , Dysarthria/etiology , Dysarthria/physiopathology , Female , Humans , Plasma Exchange/methods , Purpura, Thrombotic Thrombocytopenic/physiopathology , Purpura, Thrombotic Thrombocytopenic/therapy , Stroke/physiopathology , Stroke/therapy , Treatment Outcome
8.
Hum Gene Ther ; 27(10): 792-801, 2016 10.
Article in English | MEDLINE | ID: mdl-27550323

ABSTRACT

Diamond blackfan anemia (DBA) is a well-known inherited bone marrow failure syndrome mostly caused by mutations in ribosomal protein (RP) genes but also rarely in the hematopoietic transcription factor gene, GATA1, or TSR2, a ribosomal protein (Rps26) chaperone gene. About 25% of patients have heterozygous mutations in the RPS19 gene, which leads to haploinsufficiency of Rps19 protein in most cases. However, some RPS19 missense mutations appear to act in a dominant negative fashion. DBA typically leads to a hypoplastic anemia that becomes apparent during the first year of life, and standard treatment includes steroids or red blood cell transfusions, each modality having attendant side effects. The only curative therapy is allogeneic stem-cell transplantation, but this option is limited to patients with a histocompatible donor. DBA-mutant embryonic, induced pluripotent, and hematopoietic stem cells all exhibit growth abnormalities that can be corrected by DNA gene transfer, suggesting the possibility of ex vivo autologous gene therapy. The authors have been interested in the application of spliceosome-mediated mRNA trans-splicing (SMaRT) technology to RNA repair of DBA stem cells. Compared with gene replacement or other RNA re-programming approaches, SMaRT has several potential advantages. First, delivery of the entire normal cDNA is unnecessary, thus minimizing the overall size of the construct for packaging into a viral delivery vector. Second, RNA transcription of the corrected gene relies on the cell's endogenous transcriptional, processing, and regulatory machinery, thereby ensuring faithful and contextual expression. Third, RNA trans-splicing employs the endogenous spliceosome enzymatic machinery present in nearly all cells. Fourth, RNA trans-splicing converts mutant transcripts into therapeutically useful mRNA, and thus may be capable of treating disorders caused by dominant negative mutations. This review critically assesses prospects for both gene and RNA repair in DBA stem cells.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Genetic Therapy/methods , Hematopoietic Stem Cells , Trans-Splicing/genetics , Anemia, Diamond-Blackfan/pathology , Anemia, Diamond-Blackfan/therapy , GATA1 Transcription Factor/genetics , Gene Transfer Techniques , Haploinsufficiency/genetics , Mutation, Missense/genetics , RNA Editing/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/therapeutic use
9.
Blood ; 127(11): 1481-92, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26679864

ABSTRACT

Current therapeutic strategies for sickle cell anemia are aimed at reactivating fetal hemoglobin. Pomalidomide, a third-generation immunomodulatory drug, was proposed to induce fetal hemoglobin production by an unknown mechanism. Here, we report that pomalidomide induced a fetal-like erythroid differentiation program, leading to a reversion of γ-globin silencing in adult human erythroblasts. Pomalidomide acted early by transiently delaying erythropoiesis at the burst-forming unit-erythroid/colony-forming unit-erythroid transition, but without affecting terminal differentiation. Further, the transcription networks involved in γ-globin repression were selectively and differentially affected by pomalidomide including BCL11A, SOX6, IKZF1, KLF1, and LSD1. IKAROS (IKZF1), a known target of pomalidomide, was degraded by the proteasome, but was not the key effector of this program, because genetic ablation of IKZF1 did not phenocopy pomalidomide treatment. Notably, the pomalidomide-induced reprogramming was conserved in hematopoietic progenitors from individuals with sickle cell anemia. Moreover, multiple myeloma patients treated with pomalidomide demonstrated increased in vivo γ-globin levels in their erythrocytes. Together, these data reveal the molecular mechanisms by which pomalidomide reactivates fetal hemoglobin, reinforcing its potential as a treatment for patients with ß-hemoglobinopathies.


Subject(s)
Hematopoietic Stem Cells/drug effects , Thalidomide/analogs & derivatives , Transcription, Genetic/drug effects , gamma-Globins/genetics , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Carrier Proteins/blood , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Fetal Hemoglobin/biosynthesis , Gene Expression Regulation, Developmental , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Histone Demethylases/blood , Humans , Ikaros Transcription Factor/blood , Ikaros Transcription Factor/drug effects , Kruppel-Like Transcription Factors/blood , Lentivirus/genetics , Multiple Myeloma/blood , Multiple Myeloma/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nuclear Proteins/blood , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins , SOXD Transcription Factors/blood , Thalidomide/pharmacology , beta-Globins/biosynthesis , beta-Globins/genetics , gamma-Globins/biosynthesis
10.
PLoS One ; 9(2): e89098, 2014.
Article in English | MEDLINE | ID: mdl-24558476

ABSTRACT

Diamond Blackfan anemia (DBA) is a rare inherited bone marrow failure syndrome caused by ribosomal protein haploinsufficiency. DBA exhibits marked phenotypic variability, commonly presenting with erythroid hypoplasia, less consistently with non-erythroid features. The p53 pathway, activated by abortive ribosome assembly, is hypothesized to contribute to the erythroid failure of DBA. We studied murine embryonic stem (ES) cell lines harboring a gene trap mutation in a ribosomal protein gene, either Rps19 or Rpl5. Both mutants exhibited ribosomal protein haploinsufficiency and polysome defects. Rps19 mutant ES cells showed significant increase in p53 protein expression, however, there was no similar increase in the Rpl5 mutant cells. Embryoid body formation was diminished in both mutants but nonspecifically rescued by knockdown of p53. When embryoid bodies were further differentiated to primitive erythroid colonies, both mutants exhibited a marked reduction in colony formation, which was again nonspecifically rescued by p53 inhibition. Cell cycle analyses were normal in Rps19 mutant ES cells, but there was a significant delay in the G2/M phase in the Rpl5 mutant cells, which was unaffected by p53 knockdown. Concordantly, Rpl5 mutant ES cells had a more pronounced growth defect in liquid culture compared to the Rps19 mutant cells. We conclude that the defects in our RPS19 and RPL5 haploinsufficient mouse ES cells are not adequately explained by p53 stabilization, as p53 knockdown appears to increase the growth and differentiation potential of both parental and mutant cells. Our studies demonstrate that gene trap mouse ES cells are useful tools to study the pathogenesis of DBA.


Subject(s)
Anemia, Diamond-Blackfan/metabolism , Cell Differentiation/physiology , Disease Models, Animal , Embryonic Stem Cells/physiology , Erythroid Cells/cytology , Ribosomal Proteins/genetics , Animals , Blotting, Western , Cell Cycle/physiology , DNA Primers/genetics , Haploinsufficiency , Mice , Real-Time Polymerase Chain Reaction , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
11.
Blood ; 122(14): 2487-90, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23943650

ABSTRACT

Classical 5q- syndrome is an acquired macrocytic anemia of the elderly. Similar to Diamond Blackfan anemia (DBA), an inherited red cell aplasia, the bone marrow is characterized by a paucity of erythroid precursors. RPS14 deletions in combination with other deletions in the region have been implicated as causative of the 5q- syndrome phenotype. We asked whether smaller, less easily detectable deletions could account for a syndrome with a modified phenotype. We employed single-nucleotide polymorphism array genotyping to identify small deletions in patients diagnosed with DBA and other anemias lacking molecular diagnoses. Diminutive mosaic deletions involving RPS14 were identified in a 5-year-old patient with nonclassical DBA and in a 17-year-old patient with myelodysplastic syndrome. Patients with nonclassical DBA and other hypoproliferative anemias may have somatically acquired 5q deletions with RPS14 haploinsufficiency not identified by fluorescence in situ hybridization or cytogenetic testing, thus refining the spectrum of disorders with 5q- deletions.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Macrocytic/genetics , Cytogenetic Analysis/methods , Ribosomal Proteins/genetics , Adolescent , Anemia, Diamond-Blackfan/diagnosis , Anemia, Macrocytic/diagnosis , Anemia, Macrocytic/drug therapy , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Female , Genotype , Humans , Immunologic Factors/therapeutic use , Lenalidomide , Phenotype , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
12.
Biochem Biophys Res Commun ; 437(1): 29-34, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23792098

ABSTRACT

Shwachman Diamond syndrome (SDS) is an inherited bone marrow failure syndrome typically characterized by neutropenia, exocrine pancreas dysfunction, metaphyseal chondrodysplasia, and predisposition to myelodysplastic syndrome and leukemia. SBDS, the gene affected in most cases of SDS, encodes a protein known to influence many cellular processes including ribosome biogenesis, mitotic spindle assembly, chemotaxis, and the regulation of reactive oxygen species production. The best characterized role for the SBDS protein is in the production of functional 60S ribosomal subunits. Given that a reduction in functional 60S subunits could impact on the translational output of cells depleted of SBDS we analyzed protein synthesis in yeast cells lacking SDO1, the ortholog of SBDS. Cells lacking SDO1 selectively increased the synthesis of POR1, the ortholog of mammalian VDAC1 a major anion channel of the mitochondrial outer membrane. Further studies revealed the cells lacking SDO1 were compromised in growth on non-fermentable carbon sources suggesting mitochondrial function was impaired. These observations prompted us to examine mitochondrial function in human cells where SBDS expression was reduced. Our studies indicate that reduced expression of SBDS decreases mitochondrial membrane potential and oxygen consumption and increases the production of reactive oxygen species. These studies indicate that mitochondrial function is also perturbed in cells expressing reduced amounts of SBDS and indicate that disruption of mitochondrial function may also contribute to SDS pathophysiology.


Subject(s)
Bone Marrow Diseases/metabolism , Bone Marrow Diseases/pathology , Exocrine Pancreatic Insufficiency/metabolism , Exocrine Pancreatic Insufficiency/pathology , Lipomatosis/metabolism , Lipomatosis/pathology , Mitochondria/metabolism , Models, Biological , Saccharomyces cerevisiae/metabolism , Carbon/pharmacology , Cell Line , Fermentation/drug effects , Gene Knockdown Techniques , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Proteins/metabolism , Proteomics , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Shwachman-Diamond Syndrome
13.
Pediatr Blood Cancer ; 60(2): 281-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22997148

ABSTRACT

BACKGROUND: Shwachman-Diamond syndrome (SDS), associated with SBDS mutations, is characterized by pancreatic exocrine dysfunction and marrow failure. Sdo1, the yeast ortholog of SBDS, is implicated in maturation of the 60S ribosomal subunit, with delayed export of 60S-like particles from the nucleoplasm when depleted. Sdo1 is needed for release of the anti-subunit association factor Tif6 from 60S subunits, and Tif6 may not be recycled to the nucleus when Sdo1 is absent. METHODS: To clarify the role of SBDS in human ribosome function, TF-1 erythroleukemia and A549 lung carcinoma cells were transfected with vectors expressing RNAi against SBDS. RESULTS: Growth and hematopoietic colony forming potential of TF-1 knockdown cells were markedly hindered when compared to controls. To analyze the effect of SBDS on 60S subunit maturation in A549 cells, subunit localization was assessed by transfection with a vector expressing a fusion between human RPL29 and GFP: we found a higher percentage of SBDS-depleted cells with nuclear localization of 60S subunits. Polysome analysis of TF-1 knockdown cells showed a decrease in free 60S and 80S subunits. We also analyzed the levels of eIF6 (human ortholog of Tif6) following near-complete knockdown of SBDS in TF-1 cells and found an approximately 20% increase in the amount of eIF6 associated with the 60S subunit. CONCLUSIONS: We conclude that knockdown of SBDS leads to growth inhibition and defects in ribosome maturation, suggesting a role for wild-type SBDS in nuclear export of pre-60S subunits. Furthermore, knockdown of SBDS may interfere with eIF6 recycling.


Subject(s)
Hematopoiesis/physiology , Proteins/metabolism , Ribosomes/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Humans , Proteins/genetics , RNA, Small Interfering , Transfection
14.
Expert Rev Hematol ; 5(4): 373-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22992231

ABSTRACT

Shwachman-Diamond syndrome (SDS) is an autosomal recessive disease caused by mutations in the SBDS gene in approximately 90% of cases. SDS is characterized by exocrine pancreatic insufficiency and bone marrow failure, which predisposes to the development of myelodysplastic syndrome and/or acute myeloid leukemia. In a new report, the French national cohort studied 102 SDS patients with a median follow-up of 11.6 years, focusing on the natural history of severe cytopenias. The authors concluded that SDS patients with a young age (<3 months) at first symptomatic presentation or cytopenia at diagnosis were at a high risk of subsequent severe hematological complications (either malignant or nonmalignant). Their findings raise the possibility that a clinical algorithm may predict the subsequent development of hematological complications in SDS.

16.
Ann N Y Acad Sci ; 1242: 40-55, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22191555

ABSTRACT

Shwachman-Diamond syndrome (SDS) is an autosomal recessive disorder characterized by pancreatic exocrine insufficiency and bone marrow failure, often associated with neurodevelopmental and skeletal abnormalities. Mutations in the SBDS gene have been shown to cause SDS. The purpose of this document is to provide draft guidelines for diagnosis, evaluation of organ and system abnormalities, and treatment of hematologic, pancreatic, dietary, dental, skeletal, and neurodevelopmental complications. New recommendations regarding diagnosis and management are presented, reflecting advances in understanding the genetic basis and clinical manifestations of the disease based on the consensus of experienced clinicians from Canada, Europe, and the United States. Whenever possible, evidence-based conclusions are made, but as with other rare diseases, the data on SDS are often anecdotal. The authors welcome comments from readers.


Subject(s)
Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/therapy , Exocrine Pancreatic Insufficiency/diagnosis , Exocrine Pancreatic Insufficiency/therapy , Lipomatosis/diagnosis , Lipomatosis/therapy , Evidence-Based Medicine , Humans , Phenotype , Practice Guidelines as Topic , Shwachman-Diamond Syndrome
17.
Semin Hematol ; 48(2): 136-43, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435510

ABSTRACT

A number of human disorders, dubbed ribosomopathies, are linked to impaired ribosome biogenesis or function. These include but are not limited to Diamond Blackfan anemia (DBA), Shwachman Diamond syndrome (SDS), and the 5q- myelodysplastic syndrome (MDS). This review focuses on the latter two non-DBA disorders of ribosome function. Both SDS and 5q- syndrome lead to impaired hematopoiesis and a predisposition to leukemia. SDS, due to bi-allelic mutations of the SBDS gene, is a multi-system disorder that also includes bony abnormalities, and pancreatic and neurocognitive dysfunction. SBDS associates with the 60S subunit in human cells and has a role in subunit joining and translational activation in yeast models. In contrast, 5q- syndrome is associated with acquired haplo-insufficiency of RPS14, a component of the small 40S subunit. RPS14 is critical for 40S assembly in yeast models, and depletion of RPS14 in human CD34(+) cells is sufficient to recapitulate the 5q- erythroid defect. Both SDS and the 5q- syndrome represent important models of ribosome function and may inform future treatment strategies for the ribosomopathies.


Subject(s)
Anemia, Macrocytic/metabolism , Anemia, Macrocytic/pathology , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/pathology , Exocrine Pancreatic Insufficiency/metabolism , Exocrine Pancreatic Insufficiency/pathology , Ribosomes/metabolism , Ribosomes/pathology , Anemia, Diamond-Blackfan , Anemia, Macrocytic/genetics , Bone Marrow Diseases/genetics , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , Exocrine Pancreatic Insufficiency/genetics , Humans , Lipomatosis , Ribosomes/genetics , Shwachman-Diamond Syndrome
18.
Tissue Eng Part C Methods ; 16(6): 1523-31, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20586611

ABSTRACT

Human mesenchymal stem cells form the supportive structure in which the functional cells of a differentiated tissue reside. We describe the creation of ectopic niches within polyurethane scaffolds coated with human mesenchymal stem cells. When implanted subcutaneously in NOD/SCID mice, these niches supported engraftment of primary human acute myeloid leukemia cells. The scaffolds showed vascularization and presence of osteoclasts and adipocytes, suggestive of an organizing human bone marrow microenvironment in the murine host. The chemokine stromal-derived factor-1 (SDF-1 or CXCL12) and its receptor CXCR4 are critical for homing and migration of acute myeloid leukemia. We found that a CXCR4 antagonist could disrupt homing to the ectopic niches, possibly by modulation of the mesenchymal stroma. We believe that these scaffold niches provide a new and powerful tool to study the leukemia stem cell microenvironment and may be useful for identification of novel drug targets.


Subject(s)
Choristoma/pathology , Leukemia/pathology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/pathology , Tissue Scaffolds , Animals , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Disease Models, Animal , Humans , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Stem Cell Niche/cytology , Tissue Scaffolds/chemistry , Transplantation, Heterologous , Tumor Microenvironment/physiology
19.
Blood ; 115(17): 3421-2, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20430961
20.
Haematologica ; 95(1): 57-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19713223

ABSTRACT

BACKGROUND: Diamond-Blackfan anemia and Shwachman-Diamond syndrome are inherited bone marrow failure syndromes linked to defects in ribosome synthesis. The purpose of this study was to determine whether yeast models for Diamond-Blackfan anemia and Shwachman-Diamond syndrome differed in the mechanism by which ribosome synthesis was affected. DESIGN AND METHODS: Northern blotting, pulse-chase analysis, and polysome profiling were used to study ribosome synthesis in yeast models. Localization of 60S ribosomal subunits was assessed using RPL25eGFP. RESULTS: Relative to wild-type controls, each disease model showed defects in 60S subunit maturation, but with distinct underlying mechanisms. In the model of Diamond-Blackfan anemia, 60S subunit maturation was disrupted at a relatively early stage with abortive complexes subject to rapid degradation. 5S ribosomal RNA, unlike other large subunit ribosomal RNA in this model, accumulated as an extra-ribosomal species. In contrast, subunit maturation in the Shwachman-Diamond syndrome model was affected at a later step, giving rise to relatively stable pre-60S particles with associated 5S ribosomal RNA retained in the nucleus. Conclusions These differences between the yeast Diamond-Blackfan anemia and Shwachman-Diamond syndrome models have implications for signaling mechanisms linking abortive ribosome assembly to cell fate decisions and may contribute to the divergent clinical presentations of Diamond-Blackfan anemia and Shwachman-Diamond syndrome.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Models, Biological , Ribosomes/genetics , Saccharomyces cerevisiae Proteins/genetics , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/pathology , Animals , Disease Models, Animal , Humans , Mutation , Ribosomal Proteins/genetics , Ribosome Subunits, Large, Eukaryotic/genetics , Ribosomes/pathology , Saccharomyces cerevisiae Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...