Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Blood ; 143(23): 2414-2424, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38457657

ABSTRACT

ABSTRACT: Hyperactivation of the NF-κB cascade propagates oncogenic signaling and proinflammation, which together augments disease burden in myeloproliferative neoplasms (MPNs). Here, we systematically ablate NF-κB signaling effectors to identify core dependencies using a series of primary samples and syngeneic and patient-derived xenograft (PDX) mouse models. Conditional knockout of Rela attenuated Jak2V617F- and MPLW515L-driven onset of polycythemia vera and myelofibrosis disease hallmarks, respectively. In PDXs, RELA knockout diminished leukemic engraftment and bone marrow fibrosis while extending survival. Knockout of upstream effector Myd88 also alleviated disease burden; conversely, perturbation of negative regulator miR-146a microRNA induced earlier lethality and exacerbated disease. Perturbation of NF-κB effectors further skewed the abundance and distribution of hematopoietic multipotent progenitors. Finally, pharmacological targeting of interleukin-1 receptor-associated kinase 4 (IRAK4) with inhibitor CA-4948 suppressed disease burden and inflammatory cytokines specifically in MPN without inducing toxicity in nondiseased models. These findings highlight vulnerabilities in MPN that are exploitable with emerging therapeutic approaches.


Subject(s)
Myeloproliferative Disorders , NF-kappa B , Signal Transduction , Animals , Mice , Humans , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/metabolism , NF-kappa B/metabolism , Mice, Knockout , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics
2.
Exp Hematol ; 132: 104178, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38340948

ABSTRACT

Myeloproliferative neoplasms (MPNs) are driven by hyperactivation of JAK-STAT signaling but can demonstrate skewed hematopoiesis upon acquisition of additional somatic mutations. Here, using primary MPN samples and engineered embryonic stem cells, we demonstrate that mutations in JAK2 induced a significant increase in erythroid colony formation, whereas mutations in additional sex combs-like 1 (ASXL1) led to an erythroid colony defect. RNA-sequencing revealed upregulation of protein arginine methyltransferase 6 (PRMT6) induced by mutant ASXL1. Furthermore, genetic perturbation of PRMT6 exacerbated the MPN disease burden, including leukemic engraftment and splenomegaly, in patient-derived xenograft models, highlighting a novel tumor-suppressive function of PRMT6. However, augmented erythroid potential and bone marrow human CD71+ cells following PRMT6 knockdown were reserved only for primary MPN samples harboring ASXL1 mutations. Last, treatment of CD34+ hematopoietic/stem progenitor cells with the PRMT6 inhibitor EPZ020411 induced expression of genes involved in heme metabolism, hemoglobin, and erythropoiesis. These findings highlight interactions between JAK2 and ASXL1 mutations and a unique erythroid regulatory network in the context of mutant ASXL1.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , Erythropoiesis/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Signal Transduction , Mutation , Repressor Proteins/genetics , Repressor Proteins/metabolism , Nuclear Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism
3.
Exp Hematol ; 128: 48-66, 2023 12.
Article in English | MEDLINE | ID: mdl-37611729

ABSTRACT

Hyperactivation of JAK2 kinase is a unifying feature of human Ph- myeloproliferative neoplasms (MPNs), most commonly due to the JAK2 V617F mutation. Mice harboring a homologous mutation in the Jak2 locus exhibit a phenotype resembling polycythemia vera. NFκB pathway hyperactivation is present in myeloid neoplasms, including MPNs, despite scarcity of mutations in NFκB pathway genes. To determine the impact of NFκB pathway hyperactivation in conjunction with Jak2 V617F, we utilized Ikk2 (Ikk2-CA) mice. Pan-hematopoietic Ikk2-CA alone produced depletion of hematopoietic stem cells and B cells. When combined with the Jak2 V617F mutation, Ikk2-CA rescued the polycythemia vera phenotype of Jak2 V617F. Likewise, Jak2 V617F ameliorated defects in hematopoiesis produced by Ikk2-CA. Single-cell RNA sequencing of hematopoietic stem and progenitor cells revealed multiple genes antagonistically regulated by Jak2 and Ikk2, including subsets whose expression was altered by Jak2 V617F and/or Ikk2-CA but partly or fully rectified in the double mutant. We hypothesize that Jak2 promotes hematopoietic stem cell population self-renewal, whereas Ikk2 promotes myeloid lineage differentiation, and biases cell fates at several branch points in hematopoiesis. Jak2 and Ikk2 both regulate multiple genes affecting myeloid maturation and cell death. Therefore, the presence of dual Jak2 and NFκB hyperactivation may present neomorphic therapeutic vulnerabilities in myeloid neoplasms.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Mice , Humans , Animals , Polycythemia Vera/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Mutation , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Protein Serine-Threonine Kinases/genetics
4.
Am J Hematol ; 98(7): 1029-1042, 2023 07.
Article in English | MEDLINE | ID: mdl-37203407

ABSTRACT

Small molecule inhibitors targeting JAK2 provide symptomatic benefits for myeloproliferative neoplasm (MPN) patients and are among first-line therapeutic agents. However, despite all having potent capacity to suppress JAK-STAT signaling, they demonstrate distinct clinical profiles suggesting contributory effects in targeting other ancillary pathways. Here, we performed comprehensive profiling on four JAK2 inhibitors either FDA-approved (ruxolitinib, fedratinib, and pacritinib) or undergoing phase 3 studies (momelotinib) to better outline mechanistic and therapeutic efficacy. Across JAK2-mutant in vitro models, all four inhibitors demonstrated similar anti-proliferative phenotypes, whereas pacritinib yielded greatest potency on suppressing colony formation in primary samples, while momelotinib exhibited unique erythroid colony formation sparing. All inhibitors reduced leukemic engraftment, disease burden, and extended survival across patient-derived xenograft (PDX) models, with strongest effects elicited by pacritinib. Through RNA-sequencing and gene set enrichment analyses, differential suppressive degrees of JAK-STAT and inflammatory response signatures were revealed, which we validated with signaling and cytokine suspension mass cytometry across primary samples. Lastly, we assessed the capacity of JAK2 inhibitors to modulate iron regulation, uncovering potent suppression of hepcidin and SMAD signaling by pacritinib. These comparative findings provide insight into the differential and beneficial effects of ancillary targeting beyond JAK2 and may help guide the use of specific inhibitors in personalized therapy.


Subject(s)
Bone Marrow Neoplasms , Janus Kinase Inhibitors , Myeloproliferative Disorders , Humans , Janus Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Myeloproliferative Disorders/genetics , Janus Kinase 2/genetics
5.
Nat Commun ; 14(1): 1601, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36959190

ABSTRACT

Imaging Mass Cytometry (IMC) is an emerging multiplexed imaging technology for analyzing complex microenvironments using more than 40 molecularly-specific channels. However, this modality has unique data processing requirements, particularly for patient tissue specimens where signal-to-noise ratios for markers can be low, despite optimization, and pixel intensity artifacts can deteriorate image quality and downstream analysis. Here we demonstrate an automated content-aware pipeline, IMC-Denoise, to restore IMC images deploying a differential intensity map-based restoration (DIMR) algorithm for removing hot pixels and a self-supervised deep learning algorithm for shot noise image filtering (DeepSNiF). IMC-Denoise outperforms existing methods for adaptive hot pixel and background noise removal, with significant image quality improvement in modeled data and datasets from multiple pathologies. This includes in technically challenging human bone marrow; we achieve noise level reduction of 87% for a 5.6-fold higher contrast-to-noise ratio, and more accurate background noise removal with approximately 2 × improved F1 score. Our approach enhances manual gating and automated phenotyping with cell-scale downstream analyses. Verified by manual annotations, spatial and density analysis for targeted cell groups reveal subtle but significant differences of cell populations in diseased bone marrow. We anticipate that IMC-Denoise will provide similar benefits across mass cytometric applications to more deeply characterize complex tissue microenvironments.


Subject(s)
Algorithms , Tomography, X-Ray Computed , Humans , Signal-To-Noise Ratio , Tomography, X-Ray Computed/methods , Artifacts , Image Cytometry , Image Processing, Computer-Assisted/methods
6.
Nat Cancer ; 4(1): 108-127, 2023 01.
Article in English | MEDLINE | ID: mdl-36581736

ABSTRACT

Myeloproliferative neoplasms (MPNs) exhibit a propensity for transformation to secondary acute myeloid leukemia (sAML), for which the underlying mechanisms remain poorly understood, resulting in limited treatment options and dismal clinical outcomes. Here, we performed single-cell RNA sequencing on serial MPN and sAML patient stem and progenitor cells, identifying aberrantly increased expression of DUSP6 underlying disease transformation. Pharmacologic dual-specificity phosphatase (DUSP)6 targeting led to inhibition of S6 and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling while also reducing inflammatory cytokine production. DUSP6 perturbation further inhibited ribosomal S6 kinase (RSK)1, which we identified as a second indispensable candidate associated with poor clinical outcome. Ectopic expression of DUSP6 mediated JAK2-inhibitor resistance and exacerbated disease severity in patient-derived xenograft (PDX) models. Contrastingly, DUSP6 inhibition potently suppressed disease development across Jak2V617F and MPLW515L MPN mouse models and sAML PDXs without inducing toxicity in healthy controls. These findings underscore DUSP6 in driving disease transformation and highlight the DUSP6-RSK1 axis as a vulnerable, druggable pathway in myeloid malignancies.


Subject(s)
Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Animals , Mice , Humans , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Signal Transduction/genetics , Janus Kinases/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Dual Specificity Phosphatase 6/metabolism
7.
Exp Hematol ; 110: 47-59, 2022 06.
Article in English | MEDLINE | ID: mdl-35367529

ABSTRACT

Myelodysplastic syndromes (MDS) are hematopoietic stem cell disorders, the pathogenesis of which involves enhanced immune signaling that promotes or selects for mutant hematopoietic stem and progenitor cells (HSPCs). In particular, toll-like receptor (TLR) expression and signaling are enhanced in MDS, and their inhibition is an attractive therapeutic strategy. Although prior studies have reported increased expression of TLR2 and its binding partners TLR1 and TLR6 in the CD34+ cells of patients with MDS (especially those with low-risk disease), TLR expression in other cell types throughout the bone marrow is largely unknown. To address this, we used mass cytometry to assess the expression of TLR1, TLR2, and TLR6 and cytokines in the bone marrow hematopoietic cells of six low/intermediate-risk and six high-risk unmatched MDS bone marrow samples, as well as healthy controls, both at baseline and in response to TLR agonists. We observed several consistent differences between the groups. Most notably, TLR expression was upregulated in multiple cell populations in the low/intermediate-risk, but not high-risk, patients. In addition, many cytokines, including interleukin-6, interleukin-8, tumor necrosis factor α, transforming growth factor ß, macrophage inflammatory protein 1ß, and granzyme B, were highly expressed from various cell types in low/intermediate-risk patients. However, these same cytokines, with the exception of transforming growth factor ß, were expressed at lower levels in high-risk MDS. Together, these findings highlight the differential role of inflammation, and specifically TLR expression, in low/intermediate- versus high-risk MDS, and suggest that elevated TLR expression and cytokine production in multiple cell types likely influences the pathogenesis of MDS in lower-risk patients.


Subject(s)
Cytokines , Myelodysplastic Syndromes , Bone Marrow/pathology , Humans , Myelodysplastic Syndromes/metabolism , Toll-Like Receptor 1 , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , Toll-Like Receptors/metabolism , Transforming Growth Factor beta
9.
Blood Adv ; 6(2): 611-623, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34644371

ABSTRACT

Targeted inhibitors of JAK2 (eg ruxolitinib) often provide symptomatic relief for myeloproliferative neoplasm (MPN) patients, but the malignant clone persists and remains susceptible to disease transformation. These observations suggest that targeting alternative dysregulated signaling pathways may provide therapeutic benefit. Previous studies identified NFκB pathway hyperactivation in myelofibrosis (MF) and secondary acute myeloid leukemia (sAML) that was insensitive to JAK2 inhibition. Here, we provide evidence that NFκB pathway inhibition via pevonedistat targets malignant cells in MPN patient samples as well as in MPN and patient-derived xenograft mouse models that are nonredundant with ruxolitinib. Colony forming assays revealed preferential inhibition of MF colony growth compared with normal colony formation. In mass cytometry studies, pevonedistat blunted canonical TNFα responses in MF and sAML patient CD34+ cells. Pevonedistat also inhibited hyperproduction of inflammatory cytokines more effectively than ruxolitinib. Upon pevonedistat treatment alone or in combination with ruxolitinib, MPN mouse models exhibited reduced disease burden and improved survival. These studies demonstrating efficacy of pevonedistat in MPN cells in vitro as well as in vivo provide a rationale for therapeutic inhibition of NFκB signaling for MF treatment. Based on these findings, a Phase 1 clinical trial combining pevonedistat with ruxolitinib has been initiated.


Subject(s)
Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Primary Myelofibrosis , Animals , Cyclopentanes/therapeutic use , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , Primary Myelofibrosis/pathology , Pyrimidines
10.
Cancer Discov ; 11(12): 3126-3141, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34193440

ABSTRACT

Myeloproliferative neoplasms (MPN) are chronic blood diseases with significant morbidity and mortality. Although sequencing studies have elucidated the genetic mutations that drive these diseases, MPNs remain largely incurable with a significant proportion of patients progressing to rapidly fatal secondary acute myeloid leukemia (sAML). Therapeutic discovery has been hampered by the inability of genetically engineered mouse models to generate key human pathologies such as bone marrow fibrosis. To circumvent these limitations, here we present a humanized animal model of myelofibrosis (MF) patient-derived xenografts (PDX). These PDXs robustly engrafted patient cells that recapitulated the patient's genetic hierarchy and pathologies such as reticulin fibrosis and propagation of MPN-initiating stem cells. The model can select for engraftment of rare leukemic subclones to identify patients with MF at risk for sAML transformation and can be used as a platform for genetic target validation and therapeutic discovery. We present a novel but generalizable model to study human MPN biology. SIGNIFICANCE: Although the genetic events driving MPNs are well defined, therapeutic discovery has been hampered by the inability of murine models to replicate key patient pathologies. Here, we present a PDX system to model human myelofibrosis that reproduces human pathologies and is amenable to genetic and pharmacologic manipulation. This article is highlighted in the In This Issue feature, p. 2945.


Subject(s)
Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Animals , Clonal Evolution/genetics , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mutation , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics
11.
Cancer Discov ; 10(3): 342-344, 2020 03.
Article in English | MEDLINE | ID: mdl-32127404

ABSTRACT

Aoki and colleagues have utilized single-cell RNA sequencing and imaging mass cytometry to describe the landscape of reactive, nonmalignant immune-cell populations present in classic Hodgkin lymphoma (cHL), and delineate their spatial proximity to malignant Hodgkin-Reed-Sternberg cells. From this study, they have identified a LAG3-expressing Tr1-type Treg cell population as prevalent mainly in MHC-II-negative cHL, implying a potential functional relationship underlying the differential responsiveness of MHC-II-negative versus MHC-II-positive cHLs to immunotherapy.See related article by Aoki et al., p. 406.


Subject(s)
Hodgkin Disease , Reed-Sternberg Cells , Gene Expression Profiling , Humans , Transcriptome , Tumor Microenvironment
12.
Leukemia ; 33(8): 1978-1995, 2019 08.
Article in English | MEDLINE | ID: mdl-30718771

ABSTRACT

The distinct clinical features of myelofibrosis (MF) have been attributed in part to dysregulated inflammatory cytokine production. Circulating cytokine levels are elevated in MF patients; a subset of which have been shown to be poor prognostic indicators. In this study, cytokine overproduction was examined in MF patient plasma and in MF blood cells ex vivo using mass cytometry. Plasma cytokines measured following treatment with ruxolitinib remained markedly abnormal, indicating that aberrant cytokine production persists despite therapeutic JAK2 inhibition. In MF patient samples, 14/15 cytokines measured by mass cytometry were found to be constitutively overproduced, with the principal cellular source for most cytokines being monocytes, implicating a non-cell-autonomous role for monocyte-derived cytokines impacting disease-propagating stem/progenitor cells in MF. The majority of cytokines elevated in MF exhibited ex vivo hypersensitivity to thrombopoietin (TPO), toll-like receptor (TLR) ligands, and/or tumor necrosis factor (TNF). A subset of this group (including TNF, IL-6, IL-8, IL-10) was minimally sensitive to ruxolitinib. All TPO/TLR/TNF-sensitive cytokines, however, were sensitive to pharmacologic inhibition of NFκB and/or MAP kinase signaling. These results indicate that NFκB and MAP kinase signaling maintain cytokine overproduction in MF, and that inhibition of these pathways may provide optimal control of inflammatory pathophysiology in MF.


Subject(s)
Cytokines/biosynthesis , Janus Kinases/physiology , MAP Kinase Signaling System/physiology , NF-kappa B/physiology , Primary Myelofibrosis/immunology , STAT Transcription Factors/physiology , Signal Transduction/physiology , Humans , MAP Kinase Signaling System/drug effects , Monocytes/immunology , NF-kappa B/antagonists & inhibitors , Nitriles , Primary Myelofibrosis/drug therapy , Pyrazoles/therapeutic use , Pyrimidines , Thrombopoietin/pharmacology , Toll-Like Receptors/physiology
13.
Cytometry B Clin Cytom ; 96(1): 46-56, 2019 01.
Article in English | MEDLINE | ID: mdl-30426661

ABSTRACT

BACKGROUND: Background: Mass cytometry (CyTOF) is a powerful tool for analyzing cellular networks at the single cell level. Due to the high-dimensional nature of this approach, analysis algorithms have been developed to visualize and interpret mass cytometry data. In this study, we applied these approaches to a cohort of patients with secondary acute myeloid leukemia (sAML). METHODS: We utilized mass cytometry to interrogate localization and intensity of thrombopoietin-mediated intracellular signaling in sAML. Extracellular and intracellular phenotypes were dissected using SPADE, viSNE, and PhenoGraph. RESULTS: Healthy controls exhibited highly localized signaling responses largely restricted to the hematopoietic stem/progenitor cell (HSPC) compartment. In contrast, sAML samples contained subpopulations outside the HSPC compartment exhibiting thrombopoietin (TPO) sensitivity comparable to or greater than immunophenotypically defined HSPCs. We employed unsupervised clustering by PhenoGraph to elucidate distinct subpopulations within these heterogeneous samples. One metacluster composed almost exclusively of Lin- CD61+ CD34- CD38- CD45low cells was identified. This subpopulation was not readily identified by established manual gating approaches, and generally exhibited greater STAT phosphorylation in response to TPO stimulation than did Lin- CD61- CD34+ CD38- cells. Lin- CD61+ CD34- CD38- CD45low cells were identified in three additional sAML patients analyzed independently using a manual gating approach based upon PhenoGraph results. Each patient exhibited a similar TPO hypersensitivity to the PhenoGraph metacluster. CONCLUSIONS: The identification of this cellular subpopulation highlights the limitations of manual gating in sAML. Our study demonstrates the potential for mass cytometry to elucidate rare subpopulations in highly heterogeneous tumors by utilizing unsupervised high dimensional analysis. © 2018 International Clinical Cytometry Society.


Subject(s)
Flow Cytometry/methods , Immunophenotyping , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/pathology , Antigens, CD/metabolism , Cytokines/metabolism , Hematopoietic Stem Cells/pathology , Humans , Thrombopoietin/metabolism
14.
Cancer Cell ; 34(5): 741-756.e8, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30423295

ABSTRACT

How specific genetic lesions contribute to transformation of non-malignant myeloproliferative neoplasms (MPNs) and myelodysplastic syndromes (MDSs) to secondary acute myeloid leukemia (sAML) are poorly understood. JARID2 is lost by chromosomal deletions in a proportion of MPN/MDS cases that progress to sAML. In this study, genetic mouse models and patient-derived xenografts demonstrated that JARID2 acts as a tumor suppressor in chronic myeloid disorders. Genetic deletion of Jarid2 either reduced overall survival of animals with MPNs or drove transformation to sAML, depending on the timing and context of co-operating mutations. Mechanistically, JARID2 recruits PRC2 to epigenetically repress self-renewal pathways in hematopoietic progenitor cells. These studies establish JARID2 as a bona fide hematopoietic tumor suppressor and highlight potential therapeutic targets.


Subject(s)
Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Polycomb Repressive Complex 2/genetics , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Self Renewal/physiology , Cell Transformation, Neoplastic/pathology , Female , Gene Deletion , Gene Knockdown Techniques , Genes, Tumor Suppressor , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelodysplastic Syndromes/pathology , Myeloproliferative Disorders/pathology , N-Myc Proto-Oncogene Protein/metabolism , Polycomb Repressive Complex 2/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Transplantation, Heterologous
15.
Methods Mol Biol ; 1636: 371-392, 2017.
Article in English | MEDLINE | ID: mdl-28730492

ABSTRACT

Mass cytometry is a powerful technology that enables the measurement of >40 parameters at the single-cell level. The inherent spectral limitations of fluorescent flow cytometry are circumvented by the use of antibodies conjugated to metal isotope reporters, which are measured quantitatively using a CyTOF mass cytometer. The high dimensionality of mass cytometry is particularly useful for the analysis of cell signaling networks in complex biological samples. We describe here methods for cell preparation, antibody staining, data acquisition, and analysis of multidimensional data from a mass cytometry experiment.


Subject(s)
Mass Spectrometry , Signal Transduction , Single-Cell Analysis , Biomarkers , Bone Marrow Cells/metabolism , Cisplatin/pharmacology , Computational Biology/methods , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Humans , Permeability/drug effects , Signal Transduction/drug effects , Statistics as Topic
16.
Nat Genet ; 41(9): 977-85, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19701191

ABSTRACT

Mice homozygous for mutations in Dact1 (also called Dapper or Frodo) phenocopy human malformations involving the spine, genitourinary system and distal digestive tract. We traced this phenotype to disrupted germ-layer morphogenesis at the primitive streak. Notably, heterozygous mutation of Vangl2, a transmembrane component of the planar cell polarity (PCP) pathway, rescued recessive Dact1 phenotypes, whereas loss of Dact1 reciprocally rescued semidominant Vangl2 phenotypes. We show that Dact1, an intracellular protein, forms a complex with Vangl2. In Dact1 mutants, Vangl2 was increased at the primitive streak, where cells ordinarily undergo an epithelial-mesenchymal transition. This is associated with abnormal E-cadherin distribution and changes in biochemical measures of the PCP pathway. We conclude that Dact1 contributes to morphogenesis at the primitive streak by regulating Vangl2 upstream of cell adhesion and the PCP pathway.


Subject(s)
Congenital Abnormalities/etiology , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Nerve Tissue Proteins/metabolism , Alleles , Amino Acid Sequence , Animals , Congenital Abnormalities/genetics , Conserved Sequence , Crosses, Genetic , Homozygote , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Primitive Streak , Protein Structure, Tertiary , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...