Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Science ; 372(6542): 592-600, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33958470

ABSTRACT

The mammalian sex chromosome system (XX female/XY male) is ancient and highly conserved. The sex chromosome karyotype of the creeping vole (Microtus oregoni) represents a long-standing anomaly, with an X chromosome that is unpaired in females (X0) and exclusively maternally transmitted. We produced a highly contiguous male genome assembly, together with short-read genomes and transcriptomes for both sexes. We show that M. oregoni has lost an independently segregating Y chromosome and that the male-specific sex chromosome is a second X chromosome that is largely homologous to the maternally transmitted X. Both maternally inherited and male-specific sex chromosomes carry fragments of the ancestral Y chromosome. Consequences of this recently transformed sex chromosome system include Y-like degeneration and gene amplification on the male-specific X, expression of ancestral Y-linked genes in females, and X inactivation of the male-specific chromosome in male somatic cells. The genome of M. oregoni elucidates the processes that shape the gene content and dosage of mammalian sex chromosomes and exemplifies a rare case of plasticity in an ancient sex chromosome system.


Subject(s)
Abnormal Karyotype , Arvicolinae/genetics , Sex Determination Processes/genetics , X Chromosome/genetics , Animals , Base Sequence , Female , Gene Amplification , Genes, sry , Haplotypes , Male , Maternal Inheritance , X Chromosome Inactivation , Y Chromosome/genetics
2.
Am J Trop Med Hyg ; 102(5): 951-954, 2020 05.
Article in English | MEDLINE | ID: mdl-32124728

ABSTRACT

Four formulations of an investigational tetravalent dengue purified inactivated vaccine, administered as two doses one month (M) apart, were previously shown to be immunogenic and well-tolerated up to M13 of the phase I study NCT01702857. Here, we report results of the follow-up from M14 to year (Y) 3. One hundred healthy Puerto Rican adults, predominantly dengue virus (DENV)-primed, were randomized 1:1:1:1:1 to receive placebo or vaccine formulations: 1 µg/serotype/dose adjuvanted with aluminum, AS01E or AS03B, or aluminum-adjuvanted 4 µg/serotype/dose. No serious adverse events occurred. Two medically-attended potential immune-mediated disease cases, vaccination unrelated, were reported (groups 1 µg+Alum and 1 µg+AS03B). Of 14 instances of suspected dengue, none were laboratory confirmed. Geometric mean neutralizing antibody titers against DENV 1-4 waned from M14, but remained above pre-vaccination levels for DENV 1-3, with the highest values for group 1 µg+AS03B: 1220.1, 920.5, 819.4, and 940.5 (Y2), and 1329.3, 1169.2, 1219.8, and 718.9 (Y3). All formulations appeared to be safe and immunogenic during the 3-year follow-up.


Subject(s)
Dengue Vaccines/therapeutic use , Dengue Virus/immunology , Dengue/prevention & control , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Vaccines/administration & dosage , Dengue Vaccines/adverse effects , Dengue Vaccines/immunology , Female , Follow-Up Studies , Humans , Male , Puerto Rico
3.
PeerJ ; 7: e7783, 2019.
Article in English | MEDLINE | ID: mdl-31592350

ABSTRACT

Live trapping is a common tool used to assess demography of small mammals. However, live-trapping is often expensive and stressful to captured individuals. Thus, assessing the relative tradeoffs among study goals, project expenses, and animal well-being is necessary. Here, we evaluated how apparent bias and precision of estimates for apparent annual survival, abundance, capture probability, and recapture probability of Humboldt's flying squirrels (Glaucomys oregonensis) varied with the number of secondary trapping occasions. We used data from forested sites trapped on 12 consecutive occasions annually in the HJ Andrews Experimental Forest (9 sites, 6 years) and the Siuslaw National Forest (seven sites, three years) in Oregon. We used Huggins robust design models to estimate parameters of interest for the first 4, 8, and 12 trapping occasions. We also estimated the effect of attaching Tomahawk traps to tree boles on site- and year-specific flying squirrel capture frequencies. Our estimates with 12 occasions were similar to those from previous studies. Abundances and capture probabilities were variable among years on both sites; however, variation was much lower on the Siuslaw sites. Reducing the length of primary trapping occasions from 12 to 8 nights had very little impact on parameter estimates, but further reducing the length of primary trapping occasions to four nights caused substantial apparent bias in parameter estimates and decreased precision. We found that attaching Tomahawk traps to tree boles increased the site- and year-specific capture frequency of flying squirrels. Our results suggest that live-trapping studies targeting Humboldt's flying squirrels in the Pacific Northwest of the United States could reduce per-site costs and stress to captured individuals without biasing estimates by reducing the length of primary trapping occasions to 8 nights. We encourage similar analyses for other commonly-trapped species in these and other ecosystems.

4.
J Allergy Clin Immunol ; 141(1): 180-188.e3, 2018 01.
Article in English | MEDLINE | ID: mdl-28629749

ABSTRACT

BACKGROUND: Clonal mast cell disorders are known to occur in a subset of patients with systemic reactions to Hymenoptera stings. This observation has prompted the question of whether clonal mast cell disorders also occur in patients with idiopathic anaphylaxis (IA). OBJECTIVE: We sought to determine the prevalence of clonal mast cell disorders among patients with IA, criteria to identify those patients who require a bone marrow biopsy, and whether the pathogenesis of IA involves a hyperresponsive mast cell compartment. METHODS: We prospectively enrolled patients with IA (≥3 episodes/y) who then underwent a medical evaluation that included a serum tryptase determination, allele-specific quantitative PCR (ASqPCR) for the KIT D816V mutation, and a bone marrow examination. Mast cells were cultured from peripheral blood CD34+ cells and examined for releasability after FcεRI aggregation. RESULTS: Clonal mast cell disease was diagnosed in 14% of patients referred with IA. ASqPCR for the KIT D816V mutation was a useful adjunct in helping identify those with systemic mastocytosis but not monoclonal mast cell activation syndrome. A modified overall clonal prediction model was developed by using clinical findings, a serum tryptase determination, and ASqPCR. There was no evidence of a hyperresponsive mast cell phenotype in patients with IA. CONCLUSION: Patients with clonal mast cell disease can present as having IA. Distinct clinical and laboratory features can be used to select those patients more likely to have an underlying clonal mast cell disorder (monoclonal mast cell activation syndrome or systemic mastocytosis) and thus candidates for a bone marrow biopsy.


Subject(s)
Anaphylaxis/genetics , Anaphylaxis/immunology , Mast Cells/immunology , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/immunology , Mutation, Missense , Proto-Oncogene Proteins c-kit , Adolescent , Adult , Aged , Amino Acid Substitution , Anaphylaxis/pathology , Female , Humans , Male , Mast Cells/pathology , Mastocytosis, Systemic/pathology , Middle Aged , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology
5.
Nat Genet ; 48(12): 1564-1569, 2016 12.
Article in English | MEDLINE | ID: mdl-27749843

ABSTRACT

Elevated basal serum tryptase levels are present in 4-6% of the general population, but the cause and relevance of such increases are unknown. Previously, we described subjects with dominantly inherited elevated basal serum tryptase levels associated with multisystem complaints including cutaneous flushing and pruritus, dysautonomia, functional gastrointestinal symptoms, chronic pain, and connective tissue abnormalities, including joint hypermobility. Here we report the identification of germline duplications and triplications in the TPSAB1 gene encoding α-tryptase that segregate with inherited increases in basal serum tryptase levels in 35 families presenting with associated multisystem complaints. Individuals harboring alleles encoding three copies of α-tryptase had higher basal serum levels of tryptase and were more symptomatic than those with alleles encoding two copies, suggesting a gene-dose effect. Further, we found in two additional cohorts (172 individuals) that elevated basal serum tryptase levels were exclusively associated with duplication of α-tryptase-encoding sequence in TPSAB1, and affected individuals reported symptom complexes seen in our initial familial cohort. Thus, our findings link duplications in TPSAB1 with irritable bowel syndrome, cutaneous complaints, connective tissue abnormalities, and dysautonomia.


Subject(s)
Chronic Pain/genetics , Connective Tissue Diseases/genetics , DNA Copy Number Variations/genetics , Dysautonomia, Familial/genetics , Gastrointestinal Diseases/genetics , Pruritus/genetics , Skin Diseases/genetics , Tryptases/blood , Tryptases/genetics , Adolescent , Adult , Aged , Child , Chronic Pain/blood , Chronic Pain/enzymology , Connective Tissue Diseases/blood , Connective Tissue Diseases/enzymology , Dysautonomia, Familial/blood , Dysautonomia, Familial/enzymology , Female , Gastrointestinal Diseases/blood , Gastrointestinal Diseases/enzymology , Humans , Male , Middle Aged , Pruritus/blood , Pruritus/enzymology , Skin Diseases/blood , Skin Diseases/enzymology , Young Adult
6.
Stem Cell Res ; 15(1): 42-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26001169

ABSTRACT

Patients with systemic mastocytosis (SM) have a wide variety of problems, including skeletal abnormalities. The disease results from a mutation of the stem cell receptor (c-kit) in mast cells and we wondered if the function of bone marrow stromal cells (BMSCs; also known as MSCs or mesenchymal stem cells) might be affected by the invasion of bone marrow by mutant mast cells. As expected, BMSCs from SM patients do not have a mutation in c-kit, but they proliferate poorly. In addition, while osteogenic differentiation of the BMSCs seems to be deficient, their adipogenic potential appears to be increased. Since the hematopoietic supportive abilities of BMSCs are also important, we also studied the engraftment in NSG mice of human CD34(+) hematopoietic progenitors, after being co-cultured with BMSCs of healthy volunteers vs. BMSCs derived from patients with SM. BMSCs derived from the bone marrow of patients with SM could not support hematopoiesis to the extent that healthy BMSCs do. Finally, we performed an expression analysis and found significant differences between healthy and SM derived BMSCs in the expression of genes with a variety of functions, including the WNT signaling, ossification, and bone remodeling. We suggest that some of the symptoms associated with SM might be driven by epigenetic changes in BMSCs caused by dysfunctional mast cells in the bone marrow of the patients.


Subject(s)
Bone Marrow Cells/pathology , Mastocytosis, Systemic/pathology , Adipogenesis/genetics , Adult , Aged , Animals , Case-Control Studies , Cell Proliferation , Cell Shape , Colony-Forming Units Assay , Female , Gene Expression Profiling , Gene Regulatory Networks , Hematopoiesis/genetics , Humans , Male , Mice , Middle Aged , Mutation/genetics , Osteogenesis/genetics , Proto-Oncogene Proteins c-kit/genetics , Stromal Cells/pathology , Tissue Donors
7.
Environ Manage ; 55(4): 930-42, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25694035

ABSTRACT

We assessed the effects of the elimination of livestock in riparian systems at Hart Mountain National Antelope Refuge in southeastern Oregon, 23 years after the removal of cattle grazing, using 64 photos taken before grazing was removed compared with later retake photos. Two methods were used for this assessment: (1) a qualitative visual method comparing seven cover types and processes and (2) a new quantitative method of inserting digital line transects into photos. Results indicated that channel widths and eroding banks decreased in 64 and 73% of sites, respectively. We found a 90% decrease in the amount of bare soil (P < 0.001) and a 63% decrease in exposed channel (P < 0.001) as well as a significant increase in the cover of grasses/sedges/forbs (15% increase, P = 0.037), rushes (389% increase, P = 0.014), and willow (388% increase, P < 0.001). We also assessed the accuracy of the new method of inserting digital line transects into photo pairs. An overall accuracy of 91% (kappa 83%) suggests that digital line transects can be a useful tool for quantifying vegetation cover from photos. Our results indicate that the removal of cattle can result in dramatic changes in riparian vegetation, even in semi-arid landscapes and without replanting or other active restoration efforts.


Subject(s)
Animal Husbandry , Cattle , Conservation of Natural Resources , Ecosystem , Plants , Animals , Oregon , Qualitative Research , Soil
8.
J Allergy Clin Immunol ; 134(1): 178-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24582309

ABSTRACT

BACKGROUND: Mastocytosis associated with germline KIT activating mutations is exceedingly rare. We report the unique clinicopathologic features of a patient with systemic mastocytosis caused by a de novo germline KIT K509I mutation. OBJECTIVES: We sought to investigate the effect of the germline KIT K509I mutation on human mast cell development and function. METHODS: Primary human mast cells derived from CD34(+) peripheral blood progenitors were examined for growth, development, survival, and IgE-mediated activation. In addition, a mast cell transduction system that stably expressed the KIT K509I mutation was established. RESULTS: KIT K509I biopsied mast cells were round, CD25(-), and well differentiated. KIT K509I progenitors cultured in stem cell factor (SCF) demonstrated a 10-fold expansion compared with progenitors from healthy subjects and developed into mature hypergranular mast cells with enhanced antigen-mediated degranulation. KIT K509I progenitors cultured in the absence of SCF survived but lacked expansion and developed into hypogranular mast cells. A KIT K509I mast cell transduction system revealed SCF-independent survival to be reliant on the preferential splicing of KIT at the adjacent exonic junction. CONCLUSION: Germline KIT mutations associated with mastocytosis drive a well-differentiated mast cell phenotype distinct to that of somatic KIT D816V disease, the oncogenic potential of which might be influenced by SCF and selective KIT splicing.


Subject(s)
Germ-Line Mutation , Mast Cells/pathology , Mastocytosis, Systemic/genetics , Phenotype , Proto-Oncogene Proteins c-kit/genetics , Adult , Alternative Splicing , Cell Degranulation/drug effects , Cell Differentiation , Cell Proliferation/drug effects , Cell Survival , Cells, Cultured , Female , Gene Expression , Humans , Mast Cells/drug effects , Mast Cells/immunology , Mastocytosis, Systemic/immunology , Mastocytosis, Systemic/pathology , Stem Cell Factor/pharmacology , Transduction, Genetic
10.
Exp Hematol ; 41(10): 870-881.e2, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23743299

ABSTRACT

Stem cell factor-dependent KIT activation is an essential process for mast cell homeostasis. The two major splice variants of KIT differ by the presence or absence of four amino acids (GNNK) at the juxta-membrane region of the extracellular domain. We hypothesized that the expression pattern of these variants differs in systemic mastocytosis and that transcripts containing the KIT D816V mutation segregate preferentially to one GNNK variant. A quantitative real-time PCR assay to assess GNNK(-) and GNNK(+) transcripts from bone marrow mononuclear cells was developed. The GNNK(-)/GNNK(+) copy number ratio showed a trend toward a positive correlation with the percentage of neoplastic mast cell involvement, and KIT D816V containing transcripts displayed a significantly elevated GNNK(-)/GNNK(+) copy number ratio. Relative expression of only the GNNK(-) variant correlated with increasing percentage of neoplastic mast cell involvement. A mast cell transfection system revealed that the GNNK(-) isoform of wild type KIT was associated with increased granule formation, histamine content, and growth. When accompanying the KIT D816V mutation, the GNNK(-) isoform enhanced cytokine-free metabolism and moderately reduced sensitivity to the tyrosine kinase inhibitor, PKC412. These data suggest that neoplastic mast cells favor a GNNK(-) variant predominance, which in turn enhances the activating potential of the KIT D816V mutation and thus could influence therapeutic sensitivity in systemic mastocytosis.


Subject(s)
Gene Expression Regulation, Neoplastic , Mast Cells/metabolism , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/physiopathology , Mutation , Proto-Oncogene Proteins c-kit/genetics , Adult , Aged , Drug Resistance, Neoplasm/genetics , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Mast Cells/drug effects , Mast Cells/enzymology , Mastocytosis, Systemic/enzymology , Middle Aged , Protein Isoforms/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
12.
Blood ; 118(26): 6803-13, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22053105

ABSTRACT

Increased mast cell burden is observed in the inflamed tissues and affected organs and tissues of patients with mast cell proliferative disorders. However, normal mast cells participate in host defense, so approaches to preferentially target clonally expanding mast cells are needed. We found that mammalian target of rapamycin complex 1 (mTORC1) and 2 (mTORC2) are up-regulated in neoplastic and developing immature mast cells compared with their terminally differentiated counterparts. Elevated mTOR mRNA was also observed in bone marrow mononuclear cells of patients exhibiting mast-cell hyperplasia. Selective inhibition of mTORC1 and mTORC2 through genetic and pharmacologic manipulation revealed that, whereas mTORC1 may contribute to mast-cell survival, mTORC2 was only critical for homeostasis of neoplastic and dividing immature mast cells. The cytostatic effect of mTORC2 down-regulation in proliferating mast cells was determined to be via inhibition of cell-cycle progression. Because mTORC2 was observed to play little role in the homeostasis of differentiated, nonproliferating, mature mast cells, these data provide a rationale for adopting a targeted approaching selectively inhibiting mTORC2 to effectively reduce the proliferation of mast cells associated with inflammation and disorders of mast cell proliferation while leaving normal differentiated mast cells largely unaffected.


Subject(s)
Homeostasis , Mast Cells/metabolism , Mastocytosis, Systemic/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Profiling , Humans , Mast Cells/drug effects , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes , Naphthyridines/pharmacology , Proteins/antagonists & inhibitors , Proteins/genetics , RNA Interference , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
13.
Blood ; 118(20): 5466-75, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-21926349

ABSTRACT

Kit regulation of mast cell proliferation and differentiation has been intimately linked to the activation of phosphatidylinositol 3-OH kinase (PI3K). The activating D816V mutation of Kit, seen in the majority of mastocytosis patients, causes a robust activation of PI3K signals. However, whether increased PI3K signaling in mast cells is a key element for their in vivo hyperplasia remains unknown. Here we report that dysregulation of PI3K signaling in mice by deletion of the phosphatase and tensin homolog (Pten) gene (which regulates the levels of the PI3K product, phosphatidylinositol 3,4,5-trisphosphate) caused mast cell hyperplasia and increased numbers in various organs. Selective deletion of Pten in the mast cell compartment revealed that the hyperplasia was intrinsic to the mast cell. Enhanced STAT5 phosphorylation and increased expression of survival factors, such as Bcl-XL, were observed in PTEN-deficient mast cells, and these were further enhanced by stem cell factor stimulation. Mice carrying PTEN-deficient mast cells also showed increased hypersensitivity as well as increased vascular permeability. Thus, Pten deletion in the mast cell compartment results in a mast cell proliferative phenotype in mice, demonstrating that dysregulation of PI3K signals is vital to the observed mast cell hyperplasia.


Subject(s)
Capillary Permeability/immunology , Hypersensitivity/pathology , Mast Cells/pathology , Mast Cells/physiology , Mastocytosis/pathology , PTEN Phosphohydrolase/genetics , Adoptive Transfer , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Degranulation/immunology , Cell Division/immunology , Cell Survival/immunology , Cells, Cultured , Disease Models, Animal , Hypersensitivity/immunology , Hypersensitivity/physiopathology , Mastocytosis/immunology , Mastocytosis/physiopathology , Mice , Mice, Mutant Strains , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/immunology
14.
J Allergy Clin Immunol ; 128(5): 1086-92.e1-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21762978

ABSTRACT

BACKGROUND: IL-5 plays a central role in the development and maintenance of eosinophilia (EO) and eosinophil activation in a wide variety of eosinophilic disorders. Although IL-5, IL-3, and GM-CSF can modulate the expression of IL-5 receptor α (IL-5Rα) on eosinophils in vitro, little is known about soluble and surface IL-5Rα levels in vivo. OBJECTIVE: To assess soluble and surface IL-5Rα levels in patients with EO and/or mastocytosis. METHODS: Surface IL-5Rα expression was assessed by flow cytometry in blood and/or bone marrow from subjects with EO (n = 39) and systemic mastocytosis (n = 8) and from normal volunteers (n = 28). Soluble IL-5Rα (sIL-5Rα) level was measured in a cohort of 177 untreated subjects and correlated with EO, eosinophil activation, and serum tryptase and cytokine levels. RESULTS: IL-5Rα expression on eosinophils inversely correlated with EO (r = -0.48; P < .0001), whereas serum levels of sIL-5Rα increased with the eosinophil count (r = 0.56; P < .0001) and serum IL-5 (r = 0.40; P < .0001) and IL-13 (r = 0.29; P = .004) levels. Of interest, sIL-5Rα level was significantly elevated in patients with systemic mastocytosis without EO. Although sIL-5Rα levels correlated with serum tryptase levels in these patients, eosinophil activation, assessed by CD69 expression on eosinophils and serum eosinophil-derived neurotoxin levels, was increased compared with that in normal subjects. CONCLUSIONS: These data are consistent with an in vivo IL-5Rα regulatory pathway in human eosinophils similar to that described in vitro and involving a balance between soluble and surface receptor levels. This may have implications with respect to the use of novel therapeutic agents targeting IL-5 and its receptor in patients with EO and/or mastocytosis.


Subject(s)
Eosinophilia/metabolism , Interleukin-5 Receptor alpha Subunit/biosynthesis , Mastocytosis, Systemic/metabolism , Adult , Aged , Cell Separation , Cytokines/analysis , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Eosinophil-Derived Neurotoxin/analysis , Eosinophil-Derived Neurotoxin/biosynthesis , Eosinophil-Derived Neurotoxin/immunology , Eosinophilia/immunology , Eosinophils/immunology , Eosinophils/metabolism , Female , Flow Cytometry , Humans , Interleukin-5 Receptor alpha Subunit/immunology , Male , Mastocytosis, Systemic/immunology , Middle Aged , Tryptases/blood , Young Adult
15.
Haematologica ; 96(3): 459-63, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21134978

ABSTRACT

Cooperating genetic events are likely to contribute to the phenotypic diversity of KIT-D816V systemic mastocytosis. In this study, 44 patients with KIT-D816V systemic mastocytosis were evaluated for coexisting NRAS, KRAS, HRAS or MRAS mutations. Activating NRAS mutations were identified in 2 of 8 patients with advanced disease. NRAS mutations were not found in patients with indolent systemic mastocytosis. To better understand the clonal evolution of mastocytosis, we evaluated the cell compartments impacted by the NRAS and KIT mutations. Clonal mast cells harbored both mutations. KIT-D816V was not detected in bone marrow CD34(+) progenitors, whereas the NRAS mutation was present. These findings suggest that NRAS mutations may have the potential to precede KIT-D816V in clonal development. Unlike other mature lineages, mast cell survival is dependent on KIT and the presence of these two activating mutations may have a greater impact on the expansion of this cell compartment and in resultant disease severity.


Subject(s)
Mastocytosis, Systemic/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Aged , Antibodies/immunology , Antibodies/metabolism , Antigens, CD34/genetics , Antigens, CD34/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Clone Cells/metabolism , Clone Cells/pathology , Female , Flow Cytometry , Humans , Male , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mastocytosis, Systemic/metabolism , Mastocytosis, Systemic/pathology , Middle Aged , Mutation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-kit/metabolism
16.
J Immunol ; 182(9): 5770-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380825

ABSTRACT

Human chymase is a highly efficient angiotensin II-generating serine peptidase expressed by mast cells. When secreted from degranulating cells, it can interact with a variety of circulating antipeptidases, but is mostly captured by alpha(2)-macroglobulin, which sequesters peptidases in a cage-like structure that precludes interactions with large protein substrates and inhibitors, like serpins. The present work shows that alpha(2)-macroglobulin-bound chymase remains accessible to small substrates, including angiotensin I, with activity in serum that is stable with prolonged incubation. We used alpha(2)-macroglobulin capture to develop a sensitive, microtiter plate-based assay for serum chymase, assisted by a novel substrate synthesized based on results of combinatorial screening of peptide substrates. The substrate has low background hydrolysis in serum and is chymase-selective, with minimal cleavage by the chymotryptic peptidases cathepsin G and chymotrypsin. The assay detects activity in chymase-spiked serum with a threshold of approximately 1 pM (30 pg/ml), and reveals native chymase activity in serum of most subjects with systemic mastocytosis. alpha(2)-Macroglobulin-bound chymase generates angiotensin II in chymase-spiked serum, and it appears in native serum as chymostatin-inhibited activity, which can exceed activity of captopril-sensitive angiotensin-converting enzyme. These findings suggest that chymase bound to alpha(2)-macroglobulin is active, that the complex is an angiotensin-converting enzyme inhibitor-resistant reservoir of angiotensin II-generating activity, and that alpha(2)-macroglobulin capture may be exploited in assessing systemic release of secreted peptidases.


Subject(s)
Angiotensin II/biosynthesis , Chymases/blood , Mast Cells/enzymology , Serum/enzymology , alpha-Macroglobulins/metabolism , Adult , Child , Chymases/isolation & purification , Chymases/metabolism , Enzyme Activation , Enzyme Stability , Humans , Mast Cells/metabolism , Mastocytosis/blood , Mastocytosis/enzymology , Pilot Projects , Protein Binding , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Substrate Specificity , alpha-Macroglobulins/isolation & purification
17.
Haematologica ; 93(1): 49-56, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18166785

ABSTRACT

BACKGROUND: Translocations involving region 5q31-32 (PDGFRB) have been reported in a variety of myeloproliferative diseases and are often associated with significant peripheral eosinophilia. We report an unusual case of a patient presenting with peripheral basophilia and systemic mastocytosis in whom cytogenetic analysis revealed a t(4;5)(q21.1;q31.3). DESIGN AND METHODS: We used molecular analyses to determine the role of PDGFRB in this case. The patient was treated with imatinib. RESULTS: Fluorescence in situ hybridization (FISH) documented a breakpoint in PDGFRB. In agreement with this, the patient responded very well to imatinib with resolution of clinical symptoms, basophilia, and mast cell disease. Molecular analyses revealed that PDGFRB, encoding an imatinib-sensitive tyrosine kinase, was fused to PRKG2. The fusion gene incorporates the first two exons of PRKG2 fused to the truncated exon 12 of PDGFRB, resulting in the disruption of its juxtamembrane domain. Functional studies confirmed that the activity and transforming properties of PRKG2-PDGFRbeta were dependent on the disruption of the auto-inhibitory juxtamembrane domain. CONCLUSIONS: Our results identify a second case of the PRKG2-PDGFRB fusion and confirm the unusual PDGFRB breakpoint associated with this fusion. This work also illustrates the use of imatinib for the treatment of specific cases of systemic mastocytosis.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/genetics , Oncogene Proteins, Fusion/genetics , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/chemistry , Animals , Benzamides , Bone Marrow/pathology , Cytogenetics , Humans , Imatinib Mesylate , Immunophenotyping , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Male , Mastocytosis, Systemic/complications , Mice , Middle Aged
18.
Immunol Allergy Clin North Am ; 26(3): 549-73, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16931293

ABSTRACT

It is an exciting time in the treatment of systemic mastocytosis. Major advances in the past 2 decades have helped to define the molecular abnormalities associated with this disease and to delineate pathways involved in its pathogenesis. This has directly translated into the development of novel targeted therapies. These therapies hold great promise to patients and health care providers that a "cure" for systemic mastocytosis may someday be obtainable.


Subject(s)
Mastocytosis, Systemic/therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Counseling , Humans , Inflammation Mediators/immunology , Patient Education as Topic
SELECTION OF CITATIONS
SEARCH DETAIL