Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Blood Adv ; 6(22): 5873-5883, 2022 11 22.
Article in English | MEDLINE | ID: mdl-35977111

ABSTRACT

Early alterations within the bone marrow microenvironment that contribute to the progression of multiple myeloma (MM) from its precursor stages could be the key to identifying novel therapeutic approaches. However, the intrinsic variability in cellular populations between patients and the differences in sample processing and analysis methods have made it difficult to identify consistent changes between data sets. Here, we used single-cell RNA sequencing of bone marrow cells from precursor stages, monoclonal gammopathy of unknown significance, smoldering MM, and newly diagnosed MM and analyzed our data in combination with a previously published data set that used a similar patient population and sample processing. Despite the vast interpatient heterogeneity, some alterations were consistently observed in both data sets. We identified changes in immune cell populations as the disease progressed, which were characterized by a substantial decrease in memory and naïve CD4 T cells, and an increase in CD8+ effector T cells and T-regulatory cells. These alterations were further accompanied by an enrichment of nonclonal memory B cells and an increase in CD14 and CD16 monocytes in MM compared with its precursor stages. These results provide crucial information on the immune changes associated with the progression to clinical MM and can help to develop immune-based strategies for patient stratification and early therapeutic intervention.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Paraproteinemias , Smoldering Multiple Myeloma , Humans , Multiple Myeloma/genetics , Bone Marrow , Tumor Microenvironment/genetics
2.
Nat Commun ; 13(1): 4517, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35922426

ABSTRACT

Deciphering Multiple Myeloma evolution in the whole bone marrow is key to inform curative strategies. Here, we perform spatial-longitudinal whole-exome sequencing, including 140 samples collected from 24 Multiple Myeloma patients during up to 14 years. Applying imaging-guided sampling we observe three evolutionary patterns, including relapse driven by a single-cell expansion, competing/co-existing sub-clones, and unique sub-clones at distinct locations. While we do not find the unique relapse sub-clone in the baseline focal lesion(s), we show a close phylogenetic relationship between baseline focal lesions and relapse disease, highlighting focal lesions as hotspots of tumor evolution. In patients with ≥3 focal lesions on positron-emission-tomography at diagnosis, relapse is driven by multiple distinct sub-clones, whereas in other patients, a single-cell expansion is typically seen (p < 0.01). Notably, we observe resistant sub-clones that can be hidden over years, suggesting that a prerequisite for curative therapies would be to overcome not only tumor heterogeneity but also dormancy.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Phylogeny , Positron-Emission Tomography , Exome Sequencing
3.
Nat Commun ; 12(1): 293, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436579

ABSTRACT

Smoldering myeloma (SMM) is associated with a high-risk of progression to myeloma (MM). We report the results of a study of 82 patients with both targeted sequencing that included a capture of the immunoglobulin and MYC regions. By comparing these results to newly diagnosed myeloma (MM) we show fewer NRAS and FAM46C mutations together with fewer adverse translocations, del(1p), del(14q), del(16q), and del(17p) in SMM consistent with their role as drivers of the transition to MM. KRAS mutations are associated with a shorter time to progression (HR 3.5 (1.5-8.1), p = 0.001). In an analysis of change in clonal structure over time we studied 53 samples from nine patients at multiple time points. Branching evolutionary patterns, novel mutations, biallelic hits in crucial tumour suppressor genes, and segmental copy number changes are key mechanisms underlying the transition to MM, which can precede progression and be used to guide early intervention strategies.


Subject(s)
Evolution, Molecular , Smoldering Multiple Myeloma/genetics , APOBEC Deaminases/genetics , Adult , Aged , Aged, 80 and over , Alleles , Biomarkers, Tumor/metabolism , Clone Cells , DNA Copy Number Variations/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genome, Human , Humans , Male , Middle Aged , Multivariate Analysis , Mutation/genetics , Mutation Rate , Progression-Free Survival , Proto-Oncogene Proteins p21(ras)/genetics , Smoldering Multiple Myeloma/diagnosis , Time Factors , Translocation, Genetic
4.
Eur J Haematol ; 106(2): 230-240, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33107092

ABSTRACT

Circulating cell-free DNA (cfDNA) has the potential to capture spatial genetic heterogeneity in myeloma (MM) patients. We assessed whether cfDNA levels vary according to risk status defined by the 70 gene expression profile (GEP70). cfDNA levels in 77 patients were significantly higher in the GEP70 high-risk (HR) group compared to the low-risk (LR) group and correlated weakly with clinical markers including lactate dehydrogenase, ß2 -microglobulin, and ISS. Patients with high cfDNA levels were associated with a worse PFS (hazard ratio 6.4; 95% CI of ratio 1.9-22) and OS (hazard ratio 4.4; 95% CI of ratio 1.2-15.7). Circulating tumor DNA (ctDNA) was elevated in the HR group and ctDNA correlated strongly with GEP70 risk score (Spearman r = .69, P = .0027). cfDNA concentrations were significantly elevated between days 3-5 after chemotherapy before falling back to baseline levels. ctDNA in two patients showed a similar spike in levels between days 3 and 5 after chemotherapy with a concomitant increase in allele fraction of KRAS mutations. We assessed cfDNA levels in 25 patients with smoldering myeloma with serial samples and showed increased allele fraction of mutated KRAS at progression in cfDNA. Our study shows that cfDNA is a dynamic tool to capture genetic events in myeloma.


Subject(s)
Biomarkers, Tumor , Circulating Tumor DNA , Multiple Myeloma/genetics , Alleles , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow , Computational Biology/methods , Disease Progression , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Liquid Biopsy , Multiple Myeloma/blood , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Prognosis , Smoldering Multiple Myeloma/blood , Smoldering Multiple Myeloma/diagnosis , Smoldering Multiple Myeloma/genetics , Smoldering Multiple Myeloma/therapy , Treatment Outcome , Tumor Burden , ras Proteins/genetics
5.
J Hematol Oncol ; 13(1): 108, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32762714

ABSTRACT

BACKGROUND: Multiple Myeloma (MM) is a hematological malignancy with genomic heterogeneity and poor survival outcome. Apart from the central role of genetic lesions, epigenetic anomalies have been identified as drivers in the development of the disease. METHODS: Alterations in the DNA methylome were mapped in 52 newly diagnosed MM (NDMM) patients of six molecular subgroups and matched with loci-specific chromatin marks to define their impact on gene expression. Differential DNA methylation analysis was performed using DMAP with a ≥10% increase (hypermethylation) or decrease (hypomethylation) in NDMM subgroups, compared to control samples, considered significant for all the subsequent analyses with p<0.05 after adjusting for a false discovery rate. RESULTS: We identified differentially methylated regions (DMRs) within the etiological cytogenetic subgroups of myeloma, compared to control plasma cells. Using gene expression data we identified genes that are dysregulated and correlate with DNA methylation levels, indicating a role for DNA methylation in their transcriptional control. We demonstrated that 70% of DMRs in the MM epigenome were hypomethylated and overlapped with repressive H3K27me3. In contrast, differentially expressed genes containing hypermethylated DMRs within the gene body or hypomethylated DMRs at the promoters overlapped with H3K4me1, H3K4me3, or H3K36me3 marks. Additionally, enrichment of BRD4 or MED1 at the H3K27ac enriched DMRs functioned as super-enhancers (SE), controlling the overexpression of genes or gene-cassettes. CONCLUSIONS: Therefore, this study presents the underlying epigenetic regulatory networks of gene expression dysregulation in NDMM patients and identifies potential targets for future therapies.


Subject(s)
Epigenesis, Genetic , Epigenome , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/genetics , Multiple Myeloma/genetics , Aneuploidy , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 14/ultrastructure , Chromosomes, Human, Pair 4/genetics , Chromosomes, Human, Pair 4/ultrastructure , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin D2/biosynthesis , Cyclin D2/genetics , DNA Methylation , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Profiling , Gene Ontology , Histone Code , Histones/metabolism , Humans , Multiple Myeloma/classification , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Plasma Cells/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/genetics , Translocation, Genetic
6.
Blood Cancer J ; 10(6): 70, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32555163

ABSTRACT

Primary plasma cell leukemia (pPCL) is a rare and aggressive form of multiple myeloma (MM) that is characterized by the presence of ≥20% circulating plasma cells. Overall survival remains poor despite advances of anti-MM therapy. The disease biology as well as molecular mechanisms that distinguish pPCL from non-pPCL MM remain poorly understood and, given the rarity of the disease, are challenging to study. In an attempt to identify key biological mechanisms that result in the aggressive pPCL phenotype, we performed whole-exome sequencing and gene expression analysis in 23 and 41 patients with newly diagnosed pPCL, respectively. The results reveal an enrichment of complex structural changes and high-risk mutational patterns in pPCL that explain, at least in part, the aggressive nature of the disease. In particular, pPCL patients with traditional low-risk features such as translocation t(11;14) or hyperdiploidy accumulated adverse risk genetic events that could account for the poor outcome in this group. Furthermore, gene expression profiling showed upregulation of adverse risk modifiers in pPCL compared to non-pPCL MM, while adhesion molecules and extracellular matrix proteins became increasingly downregulated. In conclusion, this is one of the largest studies to dissect pPCL on a genomic and molecular level.


Subject(s)
Leukemia, Plasma Cell/genetics , Mutation , Adult , Aged , Aged, 80 and over , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genomics , Humans , Leukemia, Plasma Cell/pathology , Male , Middle Aged , Mutation Rate , Transcriptome , Translocation, Genetic , Exome Sequencing
7.
Clin Cancer Res ; 26(10): 2422-2432, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31988198

ABSTRACT

PURPOSE: Copy-number changes and translocations have been studied extensively in many datasets with long-term follow-up. The impact of mutations remains debated given the short time to follow-up of most datasets. EXPERIMENTAL DESIGN: We performed targeted panel sequencing covering 125 myeloma-specific genes and the loci involved in translocations in 223 newly diagnosed myeloma samples recruited into one of the total therapy trials. RESULTS: As expected, the most commonly mutated genes were NRAS, KRAS, and BRAF, making up 44% of patients. Double-Hit and BRAF and DIS3 mutations had an impact on outcome alongside classical risk factors in the context of an intensive treatment approach. We were able to identify both V600E and non-V600E BRAF mutations, 58% of which were predicted to be hypoactive or kinase dead. Interestingly, 44% of the hypoactive/kinase dead BRAF-mutated patients showed co-occurring alterations in KRAS, NRAS, or activating BRAF mutations, suggesting that they play a role in the oncogenesis of multiple myeloma by facilitating MAPK activation and may lead to chemoresistance. CONCLUSIONS: Overall, these data highlight the importance of mutational screening to better understand newly diagnosed multiple myeloma and may lead to patient-specific mutation-driven treatment approaches.


Subject(s)
Biomarkers, Tumor/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Multiple Myeloma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA Mutational Analysis , Female , Follow-Up Studies , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Prognosis , Survival Rate
8.
Haematologica ; 105(4): 1055-1066, 2020 04.
Article in English | MEDLINE | ID: mdl-31221783

ABSTRACT

MYC is a widely acting transcription factor and its deregulation is a crucial event in many human cancers. MYC is important biologically and clinically in multiple myeloma, but the mechanisms underlying its dysregulation are poorly understood. We show that MYC rearrangements are present in 36.0% of newly diagnosed myeloma patients, as detected in the largest set of next generation sequencing data to date (n=1,267). Rearrangements were complex and associated with increased expression of MYC and PVT1, but not other genes at 8q24. The highest effect on gene expression was detected in cases where the MYC locus is juxtaposed next to super-enhancers associated with genes such as IGH, IGK, IGL, TXNDC5/BMP6, FAM46C and FOXO3 We identified three hotspots of recombination at 8q24, one of which is enriched for IGH-MYC translocations. Breakpoint analysis indicates primary myeloma rearrangements involving the IGH locus occur through non-homologous end joining, whereas secondary MYC rearrangements occur through microhomology-mediated end joining. This mechanism is different to lymphomas, where non-homologous end joining generates MYC rearrangements. Rearrangements resulted in overexpression of key genes and chromatin immunoprecipitation-sequencing identified that HK2, a member of the glucose metabolism pathway, is directly over-expressed through binding of MYC at its promoter.


Subject(s)
Genes, myc , Multiple Myeloma , RNA, Long Noncoding/genetics , Genes, Immunoglobulin Heavy Chain , Humans , In Situ Hybridization, Fluorescence , Multiple Myeloma/genetics , Protein Disulfide-Isomerases , Translocation, Genetic
9.
Blood ; 132(1): 59-66, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29784643

ABSTRACT

Spatial intratumor heterogeneity is frequently seen in multiple myeloma (MM) and poses a significant challenge for risk classifiers, which rely on tumor samples from the iliac crest. Because biopsy-based assessment of multiple skeletal sites is difficult, alternative strategies for risk stratification are required. Recently, the size of focal lesions (FLs) was shown to be a surrogate marker for spatial heterogeneity, suggesting that data from medical imaging could be used to improve risk stratification approaches. Here, we investigated the prognostic value of FL size in 404 transplant-eligible, newly diagnosed MM patients. Using diffusion-weighted magnetic resonance imaging with background suppression, we identified the presence of multiple large FLs as a strong prognostic factor. Patients with at least 3 large FLs with a product of the perpendicular diameters >5 cm2 were associated with poor progression-free survival (PFS) and overall survival (OS; median, 2.3 and 3.6 years, respectively). This pattern, seen in 13.8% of patients, was independent of the Revised International Staging System (RISS), gene expression profiling (GEP)-based risk score, gain(1q), or extramedullary disease (hazard ratio, 2.7 and 2.2 for PFS and OS in multivariate analysis, respectively). The number of FLs lost its negative impact on outcome after adjusting for FL size. In conclusion, the presence of at least 3 large FL is a feature of high risk, which can be used to refine the diagnosis of this type of disease behavior and as an entry criterion for risk-stratified trials.


Subject(s)
Diffusion Magnetic Resonance Imaging , Multiple Myeloma/diagnostic imaging , Multiple Myeloma/mortality , Adult , Disease-Free Survival , Female , Humans , Male , Middle Aged , Risk Assessment , Risk Factors , Survival Rate
10.
Leukemia ; 32(11): 2435-2444, 2018 11.
Article in English | MEDLINE | ID: mdl-29654269

ABSTRACT

Chromosomal rearrangements that result in oncogenic kinase activation are present in many solid and hematological malignancies, but none have been reported in multiple myeloma (MM). Here we analyzed 1421 samples from 958 myeloma patients using a targeted assay and detected fusion genes in 1.5% of patients. These fusion genes were in-frame and the majority of them contained kinase domains from either receptor tyrosine kinases (ALK, ROS1, NTRK3, and FGFR1) or cytoplasmic kinases (BRAF, MAP3K14, and MAPK14), which would result in the activation of MEK/ERK, NF-κB, or inflammatory signaling pathways. Fusion genes were present in smoldering MM, newly diagnosed MM, and relapse patient samples indicating they are not solely late events. Most fusion genes were subclonal in nature, but one EML4-ALK fusion was clonal indicating it is a driver of disease pathogenesis. Samples with fusions of receptor tyrosine kinases were not found in conjunction with clonal Ras/Raf mutations indicating a parallel mechanism of MEK/ERK pathway activation. Fusion genes involving MAP3K14 (NIK), which regulates the NF-κB pathway, were detected as were t(14;17) rearrangements involving NIK in 2% of MM samples. Activation of kinases in myeloma through rearrangements presents an opportunity to use treatments existing in other cancers.


Subject(s)
Gene Rearrangement/genetics , Multiple Myeloma/genetics , Phosphotransferases/genetics , Protein Domains/genetics , Amino Acid Sequence , Anaplastic Lymphoma Kinase/genetics , Humans , Mutation/genetics , NF-kappa B/genetics , Neoplasm Recurrence, Local/genetics , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics
11.
Leukemia ; 32(7): 1561-1566, 2018 07.
Article in English | MEDLINE | ID: mdl-29467487

ABSTRACT

PARP inhibitors can induce synthetic lethality in tumors characterized by homologous recombination deficiency (HRD), which can be detected by evaluating genome-wide loss of heterozygosity (LOH). Multiple myeloma (MM) is a genetically unstable tumor and we hypothesized that HRD-related LOH (HRD-LOH) could be detected in patient samples, supporting a potential role for PARP inhibition in MM. Using results from targeted next-generation sequencing studies (FoundationOne® Heme), we analyzed HRD-LOH in patients at all disease stages (MGUS (n = 7), smoldering MM (SMM, n = 30), newly diagnosed MM (NDMM, n = 71), treated MM (TRMM, n = 64), and relapsed MM (RLMM, n = 234)) using an algorithm to identify HRD-LOH segments. We demonstrated HRD-LOH in MM samples, increasing as disease progresses. The extent of genomic HRD-LOH correlated with high-risk disease markers. Outcome of RLMM patients, the biggest clinical group, was analyzed and patients with HRD-LOH above the third quartile (≥5% HRD-LOH) had significantly worse progression-free and overall survival than those with lower levels (p < 0.001). Mutations in key homologous recombination genes account for some, but not all, of the cases with an excess of HRD-LOH. These data support the further evaluation of PARP inhibitors in MM patients, particularly in the relapsed setting with a high unmet need for new treatments.


Subject(s)
Biomarkers, Tumor , DNA Repair , Homologous Recombination , Loss of Heterozygosity , Multiple Myeloma/genetics , Humans , Kaplan-Meier Estimate , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prognosis , Proportional Hazards Models
13.
Blood ; 128(13): 1735-44, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27516441

ABSTRACT

To elucidate the mechanisms underlying relapse from chemotherapy in multiple myeloma, we performed a longitudinal study of 33 patients entered into Total Therapy protocols investigating them using gene expression profiling, high-resolution copy number arrays, and whole-exome sequencing. The study illustrates the mechanistic importance of acquired mutations in known myeloma driver genes and the critical nature of biallelic inactivation events affecting tumor suppressor genes, especially TP53, the end result being resistance to apoptosis and increased proliferation rates, which drive relapse by Darwinian-type clonal evolution. The number of copy number aberration changes and biallelic inactivation of tumor suppressor genes was increased in GEP70 high risk, consistent with genomic instability being a key feature of high risk. In conclusion, the study highlights the impact of acquired genetic events, which enhance the evolutionary fitness level of myeloma-propagating cells to survive multiagent chemotherapy and to result in relapse.


Subject(s)
Clonal Evolution , Genes, Tumor Suppressor , Multiple Myeloma/genetics , Mutation , Adult , Aged , Cell Proliferation , DNA Copy Number Variations , Disease Progression , Female , Gene Expression Profiling , Genes, p53 , Genes, ras , Genomic Instability , Humans , Longitudinal Studies , Male , Middle Aged , Models, Genetic , Multiple Myeloma/pathology , Multiple Myeloma/therapy , Phosphatidylinositol 3-Kinases/genetics , Recurrence , Risk Factors , Stem Cell Transplantation , Transplantation, Autologous
14.
Stem Cells ; 31(8): 1683-95, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23681919

ABSTRACT

The KIT receptor tyrosine kinase has important roles in hematopoiesis. We have recently produced a mouse model for imatinib resistant gastrointestinal stromal tumor (GIST) carrying the Kit(V558Δ) and Kit(T669I) (human KIT(T670I) ) mutations found in imatinib-resistant GIST. The Kit(V558Δ;T669I/+) mice developed microcytic erythrocytosis with an increase in erythroid progenitor numbers, a phenotype previously seen only in mouse models of polycythemia vera with alterations in Epo or Jak2. Significantly, the increased hematocrit observed in Kit(V558Δ;T669I/+) mice normalized upon splenectomy. In accordance with increased erythroid progenitors, myeloerythroid progenitor numbers were also elevated in the Kit(V558Δ;T669I/+) mice. Hematopoietic stem cell (HSC) numbers in the bone marrow (BM) of Kit(V558Δ;T669I/+) mice were unchanged in comparison to wild-type mice. However, increased HSC numbers were observed in fetal livers and the spleen and peripheral blood of adult Kit(V558Δ;T669I/+) mice. Importantly, HSC from Kit(V558Δ;T669I/+) BM had a competitive advantage over wild-type HSC. In response to 5-fluorouracil treatment, elevated numbers of dividing Lin(-) Sca(+) cells were found in the Kit(V558Δ;T669I/+) BM compared to wild type. Our study demonstrates that signaling from the Kit(V558Δ;T669I/+) receptor has important consequences in hematopoiesis enhancing HSC self-renewal and resulting in increased erythropoiesis.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Cell Growth Processes/physiology , Disease Models, Animal , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Fluorouracil/pharmacology , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 109(34): E2276-83, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22652566

ABSTRACT

Most gastrointestinal stromal tumors (GISTs) harbor a gain-of-function mutation in the Kit receptor. GIST patients treated with the tyrosine kinase inhibitor imatinib frequently develop imatinib resistance as a result of second-site Kit mutations. To investigate the consequences of second-site Kit mutations on GIST development and imatinib sensitivity, we engineered a mouse model carrying in the endogenous Kit locus both the Kit(V558Δ) mutation found in a familial case of GIST and the Kit(T669I) (human KIT(T670I)) "gatekeeper" mutation found in imatinib-resistant GIST patients. Similar to Kit(V558/+) mice, Kit(V558;T669I/+) mice developed gastric and colonic interstitial cell of Cajal hyperplasia as well as cecal GIST. In contrast to the single-mutant Kit(V558/+) control mice, treatment of the Kit(V558;T669I/+) mice with either imatinib or dasatinib failed to inhibit oncogenic Kit signaling and GIST growth. However, this resistance could be overcome by treatment of Kit(V558;T669I/+) mice with sunitinib or sorafenib. Although tumor lesions were smaller in Kit(V558;T669I/+) mice than in single-mutant mice, both interstitial cell of Cajal hyperplasia and mast cell hyperplasia were exacerbated in Kit(V558;T669I/+) mice. Strikingly, the Kit(V558;T669I/+) mice developed a pronounced polycythemia vera-like erythrocytosis in conjunction with microcytosis. This mouse model should be useful for preclinical studies of drug candidates designed to overcome imatinib resistance in GIST and to investigate the consequences of oncogenic KIT signaling in hematopoietic as well as other cell lineages.


Subject(s)
Erythrocytes/cytology , Gastrointestinal Stromal Tumors/genetics , Mutation , Piperazines/pharmacology , Polycythemia/genetics , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Lineage , Dasatinib , Disease Models, Animal , Drug Resistance , Drug Resistance, Neoplasm/genetics , Exons , Gastrointestinal Stromal Tumors/drug therapy , Imatinib Mesylate , Mice , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Thiazoles/pharmacology
16.
Dev Biol ; 337(2): 199-210, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19874813

ABSTRACT

Juxtamembrane signaling via the membrane growth factor KitL is critical for Kit mediated functions. KitL has a conserved cytoplasmic domain and has been shown to possess a monomeric leucine-dependent basolateral targeting signal. To investigate the consequences in vivo of impaired basolateral KitL targeting in polarized epithelial cells, we have mutated this critical leucine to alanine using a knock-in strategy. KitL(L263A/L263A) mutant mice are pigmented normally and steady-state hematopoiesis is unaffected although peritoneal and skin mast cell numbers are significantly increased. KitL localization is affected in the Sertoli cells of the KitL(L263A/L263A) testis and testis size is reduced in these mice due to aberrant spermatogonial proliferation. Furthermore, the effect of the KitL L263A mutation on the testicular phenotype is dosage dependent. The tubules of hemizygous KitL(L263A/Sl) mice completely lack germ cells in contrast to the weaker testicular phenotype of KitL(L263A/L263A) mice. The onset of the testis phenotype coincides with the formation of tight junctions between Sertoli cells during postnatal development. Thus, the altered sorting of KitL is dispensable for hematopoietic and melanogenic lineages, yet is crucial in the testicular environment, where the basal membranes of adjacent polarized Sertoli cells form a niche for the proliferating spermatogonia.


Subject(s)
Cell Polarity , Hematopoiesis/physiology , Spermatogenesis/physiology , Stem Cell Factor/chemistry , Stem Cell Factor/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Count , Cell Proliferation , Exons/genetics , Gene Targeting , Lymphopoiesis , Male , Mast Cells/cytology , Mice , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary , Protein Transport , Sequence Deletion , Structure-Activity Relationship , Testis/metabolism , Testis/pathology
17.
Reprod Biomed Online ; 18(4): 555-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19400999

ABSTRACT

A role for oestrogen in regulating fluid reabsorption in the monkey epididymis was recently demonstrated. Here, these studies are extended to identify potential oestrogen-regulated proteins in the cauda region of monkey epididymis treated with vehicle and oestrogen receptor antagonist (ICI 182780). Two-dimensional electrophoretic analysis was used to identify the proteins. The results indicated down-regulation of WNT4 in the ICI-182780-treated monkey cauda. In addition, the Wnt4 mRNA concentration was also reduced in the caput regions of ICI-182780- treated rats and oestrogen receptor knockout mice. WNT4 is a key regulator of gonadal differentiation in humans and mice and plays a pivotal role in early mouse embryogenesis. The results of the present study establish the presence of WNT4 in the monkey epididymis and its regulation by oestrogen, and suggest a role for WNT4 in maintaining epididymal homeostasis.


Subject(s)
Epididymis/metabolism , Estrogens/metabolism , Macaca radiata/metabolism , Water-Electrolyte Balance/physiology , Wnt Proteins/metabolism , Animals , DNA Primers/genetics , Electrophoresis, Gel, Two-Dimensional , Estradiol/analogs & derivatives , Estradiol/pharmacology , Fulvestrant , Male , Mass Spectrometry , Mice , Mice, Knockout , Rats , Rats, Wistar , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Wnt4 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...