Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Eur J Haematol ; 88(3): 195-209, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22023389

ABSTRACT

Barth syndrome (BTHS), a rare, X-linked, recessive disease, is characterized by neutropenia and cardiomyopathy. BTHS is caused by loss-of-function mutations of the tafazzin (TAZ) gene. We developed a model of BTHS by transfecting human HL60 myeloid progenitor cells with TAZ-specific shRNAs. Results demonstrate a significant downregulation in TAZ expression, mimicking the effects of naturally occurring truncation mutations in TAZ. Flow cytometry analyses of cells with TAZ-specific, but not scrambled, shRNAs demonstrate nearly twofold increase in the proportion of annexin V-positive cells and significantly increased dissipation of mitochondrial membrane potential as determined by DIOC6 staining. Transfection of TAZ-specific shRNA had similar effects in U937 myeloid cells but not in lymphoid cell lines. Further studies in HL60 myeloid progenitor cells revealed aberrant release of cytochrome c from mitochondria and significantly elevated levels of activated caspase-3 in response to TAZ knockdown. Treatment with caspase-specific inhibitor zVAD-fmk resulted in substantially reduced apoptosis to near-normal levels. These data suggest that neutropenia in BTHS is attributable to increased dissipation of mitochondrial membrane potential, aberrant release of cytochrome c, activation of caspase-3, and accelerated apoptosis of myeloid progenitor cells, and that this defect can be partially restored in vitro by treatment with caspase-specific inhibitors.


Subject(s)
Barth Syndrome/complications , Neutropenia/etiology , Neutropenia/metabolism , Acyltransferases , Apoptosis/genetics , Cardiolipins/metabolism , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation , Gene Silencing , HL-60 Cells , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/genetics , Myeloid Progenitor Cells/metabolism , Precursor Cells, T-Lymphoid/metabolism , RNA, Small Interfering , Transcription Factors/genetics , U937 Cells
2.
Blood ; 118(18): 4963-6, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21835955

ABSTRACT

Mutations in CXCR4 cause severe leukopenia in myelokathexis or WHIM syndrome. Plerixafor inhibits binding of CXCR4 to its ligand CXCL12. We investigated the effects of plerixafor (0.04 to 0.24 mg/kg) administered at 2-4 day intervals in 6 patients. Outcome measures were the patients' complete blood cell counts, CD34(+) cell counts and lymphocyte subtypes compared with 5 normal subjects similarly treated with plerixafor. All patients showed prompt leukocytosis with maximum blood neutrophils and lymphocytes at 6-12 hours. Blood neutrophils peaked at 6-12 hours, increasing from a mean baseline of 0.4 ± 0.1 × 109/L, to mean peak of 4.5 ± 0.78 × 109/L. Lymphocytes also increased; the greatest increase was in B cells (CD19(+) cells), a > 40-fold increase over baseline at the 0.08 mg/kg dose. None of the patients experienced any significant adverse effects. Plerixafor is a promising therapy for this condition.


Subject(s)
Granulocyte Precursor Cells/drug effects , Heterocyclic Compounds/therapeutic use , Immunologic Deficiency Syndromes/drug therapy , Warts/drug therapy , Adult , Aged , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , Benzylamines , Bone Marrow Diseases/drug therapy , Bone Marrow Diseases/pathology , Cyclams , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Granulocyte Precursor Cells/pathology , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/adverse effects , Heterocyclic Compounds/pharmacokinetics , Humans , Immunologic Deficiency Syndromes/blood , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Leukopenia/complications , Leukopenia/drug therapy , Leukopenia/pathology , Male , Middle Aged , Primary Immunodeficiency Diseases , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Warts/blood , Warts/genetics , Warts/pathology
3.
Pediatr Blood Cancer ; 55(2): 314-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20582973

ABSTRACT

BACKGROUND: Cyclic neutropenia (CN) and severe congenital neutropenia (SCN) are disorders of neutrophil production that differ markedly in disease severity. Mutations of the ELANE gene (the symbol recently replacing ELA2) are considered largely responsible for most cases of CN and SCN, but specific mutations are typically associated with one or the other. PROCEDURE: We performed ELANE genotyping on all individuals and paternal sperm in an SCN kindred with eight SCN progeny of a sperm donor and six different mothers. RESULTS: One patient with CN had the same S97L ELANE mutation as seven patients with the SCN phenotype. The mutant allele was detected in the donor's spermatozoa, representing 18% of the ELANE gene pool, but not in DNA from his lymphocytes, neutrophils, or buccal mucosa, indicating gonadal mosaicism. CONCLUSIONS: The coexistence of CN and SCN phenotypes in this kindred with a shared paternal haplotype strongly suggests both a role for modifying genes in determination of congenital neutropenia disease phenotypes, and the classification of CN and SCN within a spectrum of phenotypes expressing varying degrees of the same disease process.


Subject(s)
Haplotypes , Inheritance Patterns/genetics , Leukocyte Elastase/genetics , Mutation , Neutropenia/genetics , Base Sequence , Child , Child, Preschool , Fathers , Genetic Predisposition to Disease , Humans , Infant , Male , Neutropenia/congenital , Pedigree , Phenotype , Spermatozoa/enzymology
4.
Br J Haematol ; 147(4): 535-42, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19775295

ABSTRACT

Severe congenital neutropenia (SCN) is a genetically heterogeneous syndrome associated with mutations of ELANE (ELA2), HAX1, GFI1, WAS, CSF3R or G6PC3. We investigated the prevalence of mutations of ELANE in a cohort of 162 SCN patients for whom blood or bone marrow samples were submitted to the North American Severe Chronic Neutropenia Tissue Repository. Mutations of ELANE were found in 90 of 162 patients (55.6%). Subsequently, we conducted an analysis of a subset of 73 of these cases utilising a high throughput sequencing approach to determine the prevalence of other mutations associated with SCN. Among the 73 patients, mutations of ELANE were detected in 28. In the remaining 45 patients with wild type ELANE alleles, five patients had mutations: GFI1 (1), SBDS (1), WAS (1) and G6PC3 (2); no mutations of HAX1 were detected. In approximately 40% of our cases, the genetic basis of SCN remains unknown. These data suggest that for genetic diagnosis of SCN, ELANE genotyping should first be performed. In patients without ELANE mutations, other known SCN-associated gene mutations will be found rarely and genotyping can be guided by the clinical features of each patient.


Subject(s)
Leukocyte Elastase/genetics , Mutation , Neutropenia/genetics , Adaptor Proteins, Signal Transducing , Chronic Disease , DNA Mutational Analysis/methods , DNA-Binding Proteins/genetics , Female , Genetic Predisposition to Disease , Genotype , Glucose-6-Phosphatase/genetics , Humans , Male , Neutropenia/congenital , Proteins/genetics , Transcription Factors/genetics , Wiskott-Aldrich Syndrome Protein/genetics
5.
Br J Haematol ; 142(4): 653-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18513286

ABSTRACT

Most severe congenital neutropenia (SCN) cases possess constitutive neutrophil elastase mutations; a smaller cohort has acquired mutations truncating the granulocyte colony-stimulating factor receptor (G-CSF-R). We have described a case with constitutive extracellular G-CSF-R mutation hyporesponsive to ligand. Here we report two independent acquired G-CSF-R truncation mutations and a novel constitutive neutrophil elastase mutation in this patient. Co-expression of a truncated receptor chain restored STAT5 signalling responses of the extracellular G-CSF-R mutant, while constitutively-active STAT5 enhanced its proliferative capacity. These data add to our knowledge of SCN and further highlight the importance of STAT5 in mediating proliferative responses to G-CSF.


Subject(s)
Leukocyte Elastase/genetics , Mutation/genetics , Neutropenia/congenital , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Child , DNA Mutational Analysis , Humans , Neutropenia/enzymology , Neutropenia/genetics , Polymerase Chain Reaction , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , STAT5 Transcription Factor/genetics , Serine Endopeptidases/genetics
6.
Br J Haematol ; 140(2): 210-3, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18028488

ABSTRACT

Severe congenital neutropenia (SCN) is a heterogeneous bone marrow failure syndrome predisposing to myelodysplastic syndrome and acute myeloid leukaemia (MDS/AML). We studied 82 North American and Australian SCN patients enrolled in the Severe Chronic Neutropenia International Registry who were on long-term treatment with granulocyte colony-stimulating factor and for whom the neutrophil elastase (ELA2) gene was sequenced. There was no significant difference in the risk of MDS/AML in patients with mutant versus wild-type ELA2: the respective cumulative incidences at 15 years were 36% and 25% (P = 0.96). Patients with either mutant or wild-type ELA2 should be followed closely for leukaemic transformation.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukocyte Elastase/genetics , Mutation , Neutropenia/genetics , Precancerous Conditions/genetics , Australia/epidemiology , Chronic Disease , Disease Progression , Dose-Response Relationship, Drug , Epidemiologic Methods , Genetic Predisposition to Disease , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Leukemia, Myeloid, Acute/epidemiology , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Neutropenia/congenital , Neutropenia/drug therapy , Neutropenia/epidemiology , Precancerous Conditions/congenital , Precancerous Conditions/drug therapy , Precancerous Conditions/epidemiology , United States/epidemiology
7.
Pediatr Blood Cancer ; 50(3): 630-2, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17096407

ABSTRACT

This report presents the case of a 15-year-old male with severe chronic neutropenia, leukopenia, and persistent tetraploid mosaicism in the bone marrow and peripheral blood. His father had mild neutropenia and bone marrow tetraploidy. Flow cytometric analysis of DNA content peripheral blood showed tetraploidy in 20% of granulocytes and 15% of monocytes. Sequence analysis of the ELA2 gene was normal, but the GFI1 gene exhibited transient appearance of single base changes the coding region and promoter. We speculate that an underlying genetic defect, inherited in an autosomal dominant pattern, leads to both disordered mitosis and neutropenia in this kindred.


Subject(s)
DNA-Binding Proteins/genetics , Mosaicism , Neutropenia/congenital , Polyploidy , Transcription Factors/genetics , Adolescent , Adult , Chronic Disease , Genes, Dominant , Granulocytes/ultrastructure , Humans , Leukemia/genetics , Leukopenia/congenital , Leukopenia/genetics , Lymphoma, Follicular/genetics , Male , Mitosis/genetics , Monocytes/ultrastructure , Mutagenesis , Neutropenia/genetics , Paraproteinemias/genetics , Pedigree
8.
Haematologica ; 91(5): 589-95, 2006 May.
Article in English | MEDLINE | ID: mdl-16670064

ABSTRACT

BACKGROUND AND OBJECTIVES: Severe congenital neutropenia (SCN) or Kostmann syndrome was originally reported to be an autosomal recessive disease of neutrophil production causing recurrent, life-threatening infections. Mutations in the neutrophil elastase gene (ELA-2) have previously been identified in patients with sporadic or autosomal dominant SCN. DESIGN AND METHODS: We studied 14 individuals (four patients with SCN and ten close relatives) belonging to the original Kostmann family in northern Sweden for mutations in the ELA-2 and the granulocyte colony-stimulating factor (G-CSF) receptor genes. RESULTS: One patient belonging to the original Kostmann family harbored a novel heterozygous ELA-2 mutation (g.2310T-->A;Leu92His) that was not inherited from her parents. The mutation was identified in DNA isolated from both whole blood and skin fibroblasts, suggesting a sporadic de novo mutation. As a young adult this patient sequentially acquired two mutations in the gene for the G-CSF receptor (G-CSFR) and therefore recently received a hematopoietic stem cell transplant, due to the risk of evolution to leukemia. Moreover, another patient developed acute leukemia and was treated with transplantation. No pathogenic ELA-2 or G-CSFR gene mutations were found in this patient or the other two patients, nor in any healthy relative. INTERPRETATION AND CONCLUSIONS: Our data are the first to document leukemia evolution and G-CSFR gene mutations in the original Kostmann kindred. In addition, our findings indicate that ELA-2 mutations are not the primary cause of SCN in the Swedish Kostmann family.


Subject(s)
Leukocyte Elastase/genetics , Neoplasm Proteins/genetics , Neutropenia/congenital , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Preleukemia/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Adult , Amino Acid Substitution , Cell Differentiation/genetics , Child , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Disease Progression , Female , Genes, Recessive , Hematopoietic Stem Cell Transplantation , Humans , Karyotyping , Male , Mutation, Missense , Neutropenia/epidemiology , Neutropenia/genetics , Neutropenia/surgery , Pedigree , Point Mutation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Preleukemia/epidemiology , Protein Structure, Tertiary , Sequence Analysis, DNA , Sweden/epidemiology , Syndrome , Transcription Factors/genetics , Wiskott-Aldrich Syndrome Protein/genetics
9.
J Periodontol ; 76(5): 837-44, 2005 May.
Article in English | MEDLINE | ID: mdl-15898946

ABSTRACT

BACKGROUND: Severe congenital neutropenia (SCN), also known as Kostmann syndrome, was originally reported as an autosomal recessive disease of neutrophil production. The disease is characterized by a maturation arrest of neutrophil precursors at the promyelocytic stage of differentiation and by extremely low levels of mature neutrophils in peripheral blood. METHODS: A 6-year-old male presented with a complaint of gingival swelling and bleeding, and swelling at the left side of his face. Upon clinical examination, severe inflammation of all gingival tissues was apparent, and a periapical abscess with mobility was noted on the left mandibular second molar. Medical and dental histories revealed numerous recurrent bacterial infections associated with oral and non-oral tissues. His medical history with recurrent infections led us to evaluate his 3-year-old sister to determine the status of her oral health. Inflammation of her oral tissues and recurrent bacterial infections were apparent. Their consanguineous parents were in good health. To assist in identifying possible systemic diseases underlying the inflammatory situation in the siblings, consultations were requested from the Pediatric Hematology Department at Selcuk University and Pediatric Oncology Department at Gulhane Military Medical Academy. RESULTS: Based on absolute neutrophil count (< or =200/mm(3)) and bone marrow aspiration findings consistent with early maturation arrest in myelopoiesis, the cases were diagnosed as SCN. No chromosomal abnormality was detected upon cytogenetic examination. Sequencing analysis also revealed no mutation in the neutrophil elastase or growth factor independent-1 (GFI-1) genes in these patients. Severe periodontal disease, attachment loss, and mobility for over 50% of the deciduous teeth were noted. Within 6 months, the male sibling lost all of his deciduous teeth due to periapical and periodontal infections. His sister presented with tooth mobility for all mandibular incisors. Monthly visits, including scaling, polishing, and 0.2% chlorhexidine digluconate irrigation were performed to support their oral hygiene and to avoid recurrent oral infections. We have been able to stabilize these patients' periodontal conditions during a 2-year follow-up period. CONCLUSION: This case report emphasizes the role of periodontists and pediatric dentists in the diagnosis of diseases linked with neutrophil and other systemic disorders and highlights the need to optimize the health of oral tissues with regular appointments.


Subject(s)
Neutropenia/congenital , Periodontal Diseases/etiology , Bacterial Infections/blood , Cell Differentiation , Child , Child, Preschool , Consanguinity , Dental Caries/therapy , Female , Humans , Leukocyte Count , Male , Neutropenia/complications , Neutropenia/drug therapy , Neutrophils/cytology , Pedigree , Radiography , Recurrence , Siblings , Tooth Loss/diagnostic imaging , Tooth Loss/etiology , Tooth Mobility/etiology , Tooth, Deciduous
10.
Blood ; 103(9): 3355-61, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14764541

ABSTRACT

Kostmann syndrome, or severe congenital neutropenia (SCN), is an autosomal recessive disorder of neutrophil production. To investigate the potential role of apoptosis in SCN, bone marrow aspirates and biopsies were obtained from 4 patients belonging to the kindred originally described by Kostmann and 1 patient with SCN of unknown inheritance. An elevated degree of apoptosis was observed in the bone marrow of these patients, and a selective decrease in B-cell lymphoma-2 (Bcl-2) expression was seen in myeloid progenitor cells. Furthermore, in vitro apoptosis of bone marrow-derived Kostmann progenitor cells was increased, and mitochondrial release of cytochrome c was detected in CD34(+) and CD33(+) progenitors from patients, but not in controls. Administration of granulocyte colony-stimulating factor (G-CSF) restored Bcl-2 expression and improved survival of myeloid progenitor cells. In addition, cytochrome c release was partially reversed upon incubation of progenitor cells with G-CSF. In sum, these studies establish a role for mitochondria-dependent apoptosis in the pathogenesis of Kostmann syndrome and yield a tentative explanation for the beneficial effect of growth factor administration in these patients.


Subject(s)
Apoptosis , Myeloid Progenitor Cells/pathology , Neutropenia/congenital , Neutropenia/etiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Apoptosis/drug effects , Bone Marrow , Case-Control Studies , Child, Preschool , Cytochromes c/metabolism , Family Health , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Infant , Infant, Newborn , Male , Mitochondria/metabolism , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Neutropenia/drug therapy , Neutropenia/pathology , Proto-Oncogene Proteins c-bcl-2/drug effects , Syndrome
12.
Exp Hematol ; 31(5): 372-81, 2003 May.
Article in English | MEDLINE | ID: mdl-12763135

ABSTRACT

Severe congenital neutropenia (SCN) is a rare hematological disease characterized by a selective decrease in the level of circulating neutrophils in peripheral blood, maturation arrest at the promyelocyte stage of differentiation in the bone marrow, recurrent severe infections, and evolution to acute myelogenous leukemia (AML). Cellular and molecular studies of 12 SCN patients, including 5 patients that evolved to develop AML, revealed impaired proliferative characteristics and accelerated apoptosis of bone marrow progenitor cells in SCN compared with 11 healthy controls as demonstrated by flow cytometry analysis. Sequencing analysis revealed heterozygous deletion or substitution mutations in the neutrophil elastase (NE) gene in 9 of 12 patients but not in healthy controls. Expression of various NE mutants, but not normal NE, resulted in accelerated apoptosis of human promyelocytic HL-60 progenitor cells, similar to impaired survival observed in patients' cells. Bone marrow-derived primitive CD34(+) and CD33(+)/CD34(-) progenitor cells from SCN patients evolving to AML, all with mutations in the granulocyte colony-stimulating factor receptor (G-CSFR) gene, demonstrated normal cell survival, whereas more differentiated CD15(+)/CD33(-)/CD34(-) cells negative for mutant G-CSFR gene, continue to exhibit accelerated apoptosis. These data demonstrate that impaired survival of bone marrow myeloid progenitor cells, probably driven by expression of mutant NE, is the cellular mechanism responsible for neutropenia in SCN. Furthermore, our results suggest that acquired G-CSFR mutations may initiate signaling events that override the pro-apoptotic effect of mutant NE in primitive progenitor cells, resulting in an expansion of the abnormal AML clone.


Subject(s)
Leukemia, Myeloid, Acute/etiology , Neutropenia/congenital , Antigens, CD34/analysis , Bone Marrow Cells/physiology , Cell Division , Cell Survival , Humans , Leukocyte Elastase/genetics , Mutation , Neutropenia/complications , Neutropenia/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics
13.
J Pediatr Hematol Oncol ; 24(9): 784-6, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12468929

ABSTRACT

Severe neutropenia is characterized by maturation arrest of myeloid cells at the promyelocyte stage of hematopoiesis. We reported that accelerated apoptosis of bone marrow myeloid progenitor cells was observed in both cyclic (CN) and severe congenital neutropenia (SCN). Short and long-term cultures of bone marrow CD34+ cells revealed reduced production of multipotent progenitors in SCN. In contrast, production of these cells was slightly elevated in CN compared with CD34+ cells from healthy volunteers. Production of myeloid-committed progenitor cells was significantly reduced in both CN and SCN. FACS analysis of CD34+ cells revealed G /G cell cycle arrest in SCN but not in CN.(0) (1) All CN patients and more than 90% of SCN patients have mutation in the neutrophil elastase (NE) gene. Molecular modeling of NE tertiary structure indicates that mutations observed in SCN are primarily located around the glycosylation sites, whereas CN mutations affect predominantly the active site. Transient expression of CN- or SCN-specific mutant NE cDNA results in impaired survival of human myeloid progenitor cells compared with control cells transfected with intact NE cDNA. We hypothesize that abnormal processing and subcellular localization of mutant NE might predetermine the etiology of cyclic or severe congenital neutropenia.


Subject(s)
Mutation , Neutropenia/congenital , Neutropenia/genetics , Pancreatic Elastase/genetics , Hematopoiesis , Hematopoietic Stem Cells/enzymology , Humans
14.
DNA Seq ; 13(4): 221-3, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12487025

ABSTRACT

Human cyclic neutropenia, also referred to as cyclic hematopoiesis, is a rare disease characterized by periodic fluctuations in blood cell production by the bone marrow and a corresponding recurrent severe neutropenia every 19-21 days. This results in bacterial infections and shortened life expectancy. Platelets, monocytes, lymphocytes, and reticulocytes cycle with the same periodicity. It has been determined that the neutrophil elastase (NE) gene is mutated in all cases of human cyclic hematopoiesis. Currently, the only animal model for this disease is the grey collie dog, in which there is a strikingly similar periodic neutropenia every 12-14 days. Towards the validation of this animal model, we have cloned and sequenced the canine NE cDNA from a normal dog.


Subject(s)
Leukocyte Elastase/genetics , Neutropenia/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/analysis , Disease Models, Animal , Dogs , Molecular Sequence Data , Mutation , Neutropenia/blood , Neutropenia/enzymology , Neutrophils/metabolism
15.
Semin Hematol ; 39(2): 89-94, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11957190

ABSTRACT

Cyclic neutropenia is a rare hematologic disorder, characterized by repetitive episodes of fever, mouth ulcers, and infections attributable to recurrent severe neutropenia. Fluctuations in blood cells are due to oscillatory production of cells by the bone marrow. Recent genetic, molecular, and cellular studies have shown that autosomal-dominant cyclic neutropenia and sporadic cases of this disease are due to a mutation in the gene for neutrophil elastase (ELA2), located at 19p13.3. This enzyme is synthesized in neutrophil precursors early in the process of primary granule formation. It is currently presumed that the mutant neutrophil elastase functions aberrantly within the cells to accelerate apoptosis of the precursors, resulting in effective and oscillatory production. Cyclic neutropenia is effectively treated with granulocyte colony-stimulating factor (G-CSF), usually at doses of 1 to 5 microg/kg/d (median dose, 2.5 microg/kg/d). Long-term, daily, or alternate-day administration reduces fever, mouth ulcers, and other inflammatory events associated with this disorder. Leukemic transformation is not a recognized risk for cyclic neutropenia, with or without treatment with G-CSF.


Subject(s)
Neutropenia/enzymology , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Leukocyte Elastase/genetics , Mutation , Neutropenia/drug therapy , Neutropenia/etiology , Periodicity , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...