Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Cell Immunol ; 4(1): 19-28, 2022.
Article in English | MEDLINE | ID: mdl-36052149

ABSTRACT

Heterozygous mutations in ELANE, the gene for neutrophil elastase, cause cyclic and congenital neutropenia through the programed cell death of neutrophil progenitors in the bone marrow. Granulocyte colony-stimulating factor is an effective therapy for these diseases, but alternative therapies are needed, especially for patients who do not respond well or are at high risk of developing myeloid malignancies. We developed an HL60 cell model for ELANE neutropenia and previously demonstrated that transient and regulated expression of mutant ELANE causes cell death by accelerated apoptosis. Knocking down the mutant gene or exposure to a potent inhibitor of neutrophil elastase rescued neutrophil development. Because of the great diversity in causative ELANE mutations, we generated stable HL60 clones expressing mutant P139L, C151Y and G214R and compared the effects of elastase inhibitor exposure to an ELANE knock-out line on cell development and function. ATRA induced differentiation demonstrated comparably impaired myeloid cell development for all three lines with upregulated expression of GRP78/BIP, an abnormality corrected by exposure of these cells to the elastase inhibitor MK-0339. The inhibitor and KO of mutant ELANE led to formation of neutrophils with comparable chemotactic and bactericidal capacities. We concluded that both strategies have great potential for the treatment of cyclic and congenital neutropenia. However, an orally absorbed, cell permeable inhibitor of neutrophil elastase, if proven safe and effective in a clinical trial, might be the better alternative to G-CSF or gene editing to treat ELANE neutropenia.

2.
Blood ; 139(5): 779-791, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34115842

ABSTRACT

Severe congenital neutropenia is an inborn disorder of granulopoiesis. Approximately one third of cases do not have a known genetic cause. Exome sequencing of 104 persons with congenital neutropenia identified heterozygous missense variants of CLPB (caseinolytic peptidase B) in 5 severe congenital neutropenia cases, with 5 more cases identified through additional sequencing efforts or clinical sequencing. CLPB encodes an adenosine triphosphatase that is implicated in protein folding and mitochondrial function. Prior studies showed that biallelic mutations of CLPB are associated with a syndrome of 3-methylglutaconic aciduria, cataracts, neurologic disease, and variable neutropenia. However, 3-methylglutaconic aciduria was not observed and, other than neutropenia, these clinical features were uncommon in our series. Moreover, the CLPB variants are distinct, consisting of heterozygous variants that cluster near the adenosine triphosphate-binding pocket. Both genetic loss of CLPB and expression of CLPB variants result in impaired granulocytic differentiation of human hematopoietic progenitor cells and increased apoptosis. These CLPB variants associate with wild-type CLPB and inhibit its adenosine triphosphatase and disaggregase activity in a dominant-negative fashion. Finally, expression of CLPB variants is associated with impaired mitochondrial function but does not render cells more sensitive to endoplasmic reticulum stress. Together, these data show that heterozygous CLPB variants are a new and relatively common cause of congenital neutropenia and should be considered in the evaluation of patients with congenital neutropenia.


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , Endopeptidase Clp/genetics , Neutropenia/congenital , Cells, Cultured , Endopeptidase Clp/chemistry , Exome , Female , Genetic Variation , Heterozygote , Humans , Infant , Male , Models, Molecular , Mutation , Neutropenia/genetics
3.
Blood ; 136(26): 2994-3003, 2020 12 24.
Article in English | MEDLINE | ID: mdl-32870250

ABSTRACT

Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome is a rare primary immunodeficiency caused by gain-of-function mutations in the CXCR4 gene. We report the safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary efficacy of mavorixafor from a phase 2 open-label dose-escalation and extension study in 8 adult patients with genetically confirmed WHIM syndrome. Mavorixafor is an oral small molecule selective antagonist of the CXCR4 receptor that increases mobilization and trafficking of white blood cells from the bone marrow. Patients received escalating doses of mavorixafor, up to 400 mg once daily. Five patients continued on the extension study for up to 28.6 months. Mavorixafor was well tolerated with no treatment-related serious adverse events. At a median follow-up of 16.5 months, we observed dose-dependent increases in absolute neutrophil count (ANC) and absolute lymphocyte count (ALC). At doses ≥300 mg/d, ANC was maintained at >500 cells per microliter for a median of 12.6 hours, and ALC was maintained at >1000 cells per microliter for up to 16.9 hours. Continued follow-up on the extension study resulted in a yearly infection rate that decreased from 4.63 events (95% confidence interval, 3.3-6.3) in the 12 months prior to the trial to 2.27 events (95% confidence interval, 1.4-3.5) for patients on effective doses. We observed an average 75% reduction in the number of cutaneous warts. This study demonstrates that mavorixafor, 400 mg once daily, mobilizes neutrophil and lymphocytes in adult patients with WHIM syndrome and provides preliminary evidence of clinical benefit for patients on long-term therapy. The trial was registered at www.clinicaltrials.gov as #NCT03005327.


Subject(s)
Aminoquinolines/administration & dosage , Benzimidazoles/administration & dosage , Butylamines/administration & dosage , Primary Immunodeficiency Diseases/drug therapy , Receptors, CXCR4/antagonists & inhibitors , Warts/drug therapy , Administration, Oral , Adolescent , Adult , Aminoquinolines/adverse effects , Benzimidazoles/adverse effects , Butylamines/adverse effects , Female , Humans , Leukocyte Count , Male , Middle Aged , Neutrophils , Primary Immunodeficiency Diseases/blood , Primary Immunodeficiency Diseases/genetics , Prospective Studies , Receptors, CXCR4/genetics , Warts/blood , Warts/genetics
4.
Curr Opin Hematol ; 27(1): 11-17, 2020 01.
Article in English | MEDLINE | ID: mdl-31652152

ABSTRACT

PURPOSE OF REVIEW: WHIM syndrome (warts, hypogammaglobulinemia, immunodeficiency, myelokathexis, or WHIMs) is a very rare autosomal dominant immunodeficiency disorder attributable to mutations in CXCR4. We reviewed clinical manifestations in 24 patients in 9 families to expand understanding of this syndrome. RECENT FINDINGS: Warts, cellulitis and respiratory infections are common in patients with WHIMs. Less commonly these patients have congenital heart disease, human papilloma virus-associated malignancies (cervical and vulvular) and lymphomas. Hearing loss because of recurrent otitis media is another important complication. Treatment with granulocyte colony-stimulating factor is controversial; this review indicates that it is effective to prevent and treat infections based upon long-term observations of patients enrolled in the Severe Chronic Neutropenia International Registry. Understanding the natural history and diversity of this syndrome are important for ongoing clinical trials of novel agents to treat WHIMs. SUMMARY: WHIM syndrome has diverse manifestations; some features occur consistently in almost all patients, for example, neutropenia, lymphocytopenia and mild hypogammaglobulinemia. However, the clinical consequences are quite variable across patient cohorts and within families. Each complication is important as a cause for morbidity and a source for patient and family concerns.


Subject(s)
Agammaglobulinemia , Family , Mutation , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Registries , Warts , Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Agammaglobulinemia/pathology , Agammaglobulinemia/therapy , Female , Humans , Male , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/therapy , Risk Factors , Warts/diagnosis , Warts/genetics , Warts/pathology , Warts/therapy
5.
Curr Opin Hematol ; 26(1): 16-21, 2019 01.
Article in English | MEDLINE | ID: mdl-30451720

ABSTRACT

PURPOSE OF REVIEW: Glycogen storage disease Ib (GSD Ib) is characterized by hepatomegaly, hypoglycemia, neutropenia, enterocolitis and recurrent bacterial infections. It is attributable to mutations in G6PT1, the gene for the glucose-6-phosphate transporter responsible for transport of glucose into the endoplasmic reticulum. Neutropenia in GSD Ib is now frequently treated with granulocyte colony-stimulating factor (G-CSF). We formed a cooperative group to review outcomes of the long-term treatment of GSD Ib patients treated with G-CSF. RECENT FINDINGS: The study enrolled 103 patients (48 men and 55 women), including 47 currently adult patients. All of these patients were treated with G-CSF, starting at a median age of 3.8 years (range 0.04-33.9 years) with a median dose of 3.0 mcg/kg/day (range 0.01-93.1 mcg/kg/day) for a median of 10.3 years (range 0.01-29.3 years). Neutrophils increased in response to G-CSF in all patients (median values before G-CSF 0.2 × 10/l, on G-CSF 1.20 x 10/l). Treatment increased spleen size (before G-CSF, 47%, on treatment on G-CSF 76%), and splenomegaly was the dose-limiting adverse effect of treatment (pain and early satiety). Clinical observations and records attest to reduce frequency of infectious events and the severity of inflammatory bowel symptoms, but fever and recurrent infections remain a significant problem. In the cohort of patients followed carefully through the Severe Chronic Neutropenia International Registry, four patients have developed myelodysplasia or acute myeloid leukemia and we are aware of four other cases, (altogether seven on G-CSF, one never treated with G-CSF). Liver transplantation in five patients did not correct neutropenia. Four patients had hematopoietic stem cell transplantation; two adults and two children were transplanted; one adult and one child survived. SUMMARY: GSD Ib is a complex disorder of glucose metabolism causing severe chronic neutropenia. G-CSF is effective to raise blood neutrophil counts and reduce fevers and infections in most patients. In conjunction with other therapies (salicylates, mesalamine sulfasalazine and prednisone), G-CSF ameliorates inflammatory bowel symptoms, but doses must be limited because it increases spleen size associated with abdominal pain.


Subject(s)
Glycogen Storage Disease Type I/drug therapy , Glycogen Storage Disease Type I/mortality , Granulocyte Colony-Stimulating Factor/administration & dosage , Neutropenia/drug therapy , Neutropenia/mortality , Registries , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Glycogen Storage Disease Type I/blood , Glycogen Storage Disease Type I/pathology , Humans , Male , Middle Aged , Neutropenia/blood , Neutropenia/pathology , Neutrophils/metabolism , Neutrophils/pathology , Survival Rate
6.
J Leukoc Biol ; 102(4): 1143-1151, 2017 10.
Article in English | MEDLINE | ID: mdl-28754797

ABSTRACT

Mutations in ELANE, the gene for neutrophil elastase (NE), a protease expressed early in neutrophil development, are the most frequent cause of cyclic (CyN) and severe congenital neutropenia (SCN). We hypothesized that inhibitors of NE, acting either by directly inhibiting enzymatic activity or as chaperones for the mutant protein, might be effective as therapy for CyN and SCN. We investigated ß-lactam-based inhibitors of human NE (Merck Research Laboratories, Kenilworth, NJ, USA), focusing on 1 inhibitor called MK0339, a potent, orally absorbed agent that had been tested in clinical trials and shown to have a favorable safety profile. Because fresh, primary bone marrow cells are rarely available in sufficient quantities for research studies, we used 3 cellular models: patient-derived, induced pluripotent stem cells (iPSCs); HL60 cells transiently expressing mutant NE; and HL60 cells with regulated expression of the mutant enzyme. In all 3 models, the cells expressing the mutant enzyme had reduced survival as measured with annexin V and FACS. Coincubation with the inhibitors, particularly MK0339, promoted cell survival and increased formation of mature neutrophils. These studies suggest that cell-permeable inhibitors of neutrophil elastase show promise as novel therapies for ELANE-associated neutropenia.


Subject(s)
Enzyme Inhibitors/pharmacology , Leukocyte Elastase , Mutation , Neutropenia/congenital , Cell Survival , Congenital Bone Marrow Failure Syndromes , Female , HL-60 Cells , Humans , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Male , Neutropenia/drug therapy , Neutropenia/enzymology , Neutropenia/genetics
7.
Obstet Gynecol ; 125(1): 197-203, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25560125

ABSTRACT

OBJECTIVE: To report outcomes associated with the administration of granulocyte colony-stimulating factor (G-CSF) to women with chronic neutropenia during pregnancy. METHODS: We conducted an observational study of women of childbearing potential with congenital, cyclic, idiopathic, or autoimmune neutropenia enrolled in the Severe Chronic Neutropenia International Registry to determine outcomes of pregnancies, without and with chronic G-CSF therapy, 1999-2014. Treatment decisions were made by the patients' personal physicians. A research nurse conducted telephone interviews of all enrolled U.S. women of childbearing potential using a standard questionnaire. Comparisons used Fisher's exact test analysis and Student's t test. RESULTS: One hundred seven women reported 224 pregnancies, 124 without G-CSF therapy and 100 on chronic G-CSF therapy (median dose 1.0 micrograms/kg per day, range 0.02-8.6 micrograms/kg per day). There were no significant differences in adverse events between the groups considering all pregnancies or individual mothers, for example, spontaneous terminations (all pregnancies: no G-CSF in 27/124, G-CSF in 13/100; P=.11, Fisher's exact test), preterm labors (all pregnancies, no G-CSF in 9/124, G-CSF in 2/100, P=.12). A study with at least 300 per group would be needed to detect a difference in these events with 80% statistical power (α=0.05). Four newborns of mothers with idiopathic or autoimmune neutropenia not on G-CSF (4/101) had life-threatening infections, whereas there were no similar events (0/90) in the treated group, but this difference was also not statistically significant (P=.124). Adverse events in the neonates were similar for the two groups. CONCLUSION: This observational study showed no significant adverse effects of administration of G-CSF to women with severe chronic neutropenia during pregnancy. LEVEL OF EVIDENCE: III.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Neutropenia/drug therapy , Neutrophils , Pregnancy Complications, Hematologic/drug therapy , Abortion, Spontaneous/epidemiology , Adolescent , Adult , Female , Gestational Age , Humans , Infant, Newborn , Infections/epidemiology , Leukocyte Count , Live Birth , Neutropenia/blood , Obstetric Labor, Premature/epidemiology , Pregnancy , Pregnancy Complications, Hematologic/blood , Young Adult
8.
Curr Opin Hematol ; 22(1): 3-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25427142

ABSTRACT

PURPOSE OF REVIEW: Mutations in the gene for neutrophil elastase, ELANE, cause cyclic neutropenia (CyN) and severe congenital neutropenia (SCN). This study summarized data from the Severe Chronic Neutropenia International Registry (SCNIR) on genotype-phenotype relationships of ELANE mutations to important clinical outcomes. We also summarize findings for ELANE mutations not observed in SCNIR patients. RECENT FINDINGS: There were 307 SCNIR patients with 104 distinctive ELANE mutations who were followed longitudinally for up to 27 years. The ELANE mutations were diverse; there were 65 single amino acid substitutions; 61 of these mutations (94%) were 'probably' or 'possibly damaging' by PolyPhen-2 analysis, and one of the 'benign' mutations was associated with two cases of acute myeloid leukemia (AML). All frame-shift mutations (19/19) were associated with the SCN. The pattern of mutations in the SCN versus CyN was significantly different (P < 10), but some mutations were observed in both groups (overlapping mutations). The cumulative incidence of severe adverse events, that is, myelodysplasia, AML, stem cell transplantation, or deaths was significantly greater for patients with SCN versus those with CyN or overlapping mutations. Specific mutations (i.e. G214R or C151Y) had a high risk for evolution to AML. SUMMARY: Sequencing is useful for predicting outcomes of ELANE-associated neutropenia.


Subject(s)
Genetic Diseases, Inborn/genetics , Leukocyte Elastase/genetics , Mutation , Neutropenia/genetics , Animals , Genetic Diseases, Inborn/enzymology , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukocyte Elastase/metabolism , Neutropenia/enzymology
9.
Hum Mutat ; 35(7): 824-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24753205

ABSTRACT

Severe congenital neutropenia (SCN) is a rare hematopoietic disorder, with estimated incidence of 1 in 200,000 individuals of European descent, many cases of which are inherited in an autosomal dominant pattern. Despite the fact that several causal genes have been identified, the genetic basis for >30% of cases remains unknown. We report a five-generation family segregating a novel single nucleotide variant (SNV) in TCIRG1. There is perfect cosegregation of the SNV with congenital neutropenia in this family; all 11 affected, but none of the unaffected, individuals carry this novel SNV. Western blot analysis show reduced levels of TCIRG1 protein in affected individuals, compared to healthy controls. Two unrelated patients with SCN, identified by independent investigators, are heterozygous for different, rare, highly conserved, coding variants in TCIRG1.


Subject(s)
Neutropenia/congenital , Vacuolar Proton-Translocating ATPases/genetics , Congenital Bone Marrow Failure Syndromes , DNA Mutational Analysis , Heterozygote , Humans , Mutation , Neutropenia/diagnosis , Neutropenia/genetics , Neutropenia/metabolism , Pedigree , Vacuolar Proton-Translocating ATPases/metabolism
10.
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
11.
Mol Endocrinol ; 20(2): 414-25, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16210345

ABSTRACT

Corticotroph-derived glycoprotein hormone (CGH), also referred to as thyrostimulin, is a noncovalent heterodimer of glycoprotein hormone alpha 2 (GPHA2) and glycoprotein hormone beta 5 (GPHB5). Here, we demonstrate that both subunits of CGH are expressed in the corticotroph cells of the human anterior pituitary, as well as in skin, retina, and testis. CGH activates the TSH receptor (TSHR); (125)I-CGH binding to cells expressing TSHR is saturable, specific, and of high affinity. In competition studies, unlabeled CGH is a potent competitor for (125)I-TSH binding, whereas unlabeled TSH does not compete for (125)I-CGH binding. Binding and competition analyses are consistent with the presence of two binding sites on the TSHR transfected baby hamster kidney cells, one that can interact with either TSH or CGH, and another that binds CGH alone. Transgenic overexpression of GPHB5 in mice produces elevations in serum T(4) levels, reductions in body weight, and proptosis. However, neither transgenic overexpression of GPHA2 nor deletion of GPHB5 produces an overt phenotype in mice. In vivo administration of CGH to mice produces a dose-dependent hyperthyroid phenotype including elevation of T(4) and hypertrophy of cells within the inner adrenal cortex. However, the distinctive expression patterns and binding characteristics of CGH suggest that it has endogenous biological roles that are discrete from those of TSH.


Subject(s)
Glycoproteins/metabolism , Receptors, Thyrotropin/metabolism , Animals , Binding Sites , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Glycoproteins/analysis , Glycoproteins/genetics , Glycoproteins/pharmacology , Humans , Hypertrophy , Male , Mice , Mice, Transgenic , Peptide Hormones/analysis , Peptide Hormones/metabolism , Pituitary Gland, Anterior/chemistry , Pituitary Gland, Anterior/metabolism , Retina/chemistry , Retina/metabolism , Skin/chemistry , Skin/metabolism , Testis/chemistry , Testis/metabolism , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroxine/blood , Tissue Distribution
12.
Int Arch Allergy Immunol ; 99(2-4): 411-415, 1992.
Article in English | MEDLINE | ID: mdl-34167267

ABSTRACT

We have previously demonstrated that incubation of splenic lymphocytes from an unimmunized mouse with IL-2 IFN-α or γ resulted in the development of a population of nonspecific regulatory cells (Ts). These cells were shown to block the ability of lymphocytes to generate mixed-lymphocyte responses in vitro. In the studies reported here, we have investigated the cell populations involved in this phenomenon. The Ts cells develop over a period of 2 or more days in culture with IL-2. Antibody to the 55-kD chain of the IL-2 receptor blocks Ts generation while stimulating T-cell proliferation. Although NK activity develops in these cultures, the mechanism of suppression is not via a lytic mechanism. Generation of the Ts in culture in the presence of IL-2 requires adherent cells as well as CD8+ cells. In studies using Con-A as the stimulus, the generation of Ts clearly requires both CD4+ T cells as well as CD8+ T cells and adherent cells. The evidence suggests that the CD4+ T cells serve as a source of IL-2 that is necessary in the activation of the IL-2-responsive CD8+, nonspecific Ts.

SELECTION OF CITATIONS
SEARCH DETAIL
...