Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Exp Hematol ; 123: 28-33.e3, 2023 07.
Article in English | MEDLINE | ID: mdl-37209901

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive, noncurative, mature B-cell lymphoma, with a median overall survival of 6-7 years. This underlines a need for effective therapeutic strategies to treat MCL better. Epidermal growth factor-like 7 (EGFL7) is a protein secreted by endothelial cells shown to play a critical role in angiogenesis. Our laboratory has previously demonstrated that EGFL7 supports the growth of leukemic blasts in patients with acute myeloid leukemia (AML); however, its role in MCL has not been investigated yet. In this study, we report that EGFL7 messenger RNA (mRNA) is increased in the cells of patients with MCL compared with cells from healthy controls, and patients with high EGFL7 are associated with lower overall survival rates. Furthermore, EGFL7 is increased in the plasma of patients with MCL compared with the plasma from healthy controls. We further show that EGFL7 binds to epidermal growth factor receptor (EGFR) and activates AKT signaling pathway in MCL cells and that blocking EGFL7 in MCL in patient and cell lines decreases cell growth and increases apoptosis in vitro. Finally, anti-EGFL7 treatment inhibits tumor size and prolongs survival in a mouse model of MCL. In conclusion, our study reveals a role for EGFL7 in MCL cell proliferation and highlights EGFL7 inhibition as a promising new treatment for patients with MCL.


Subject(s)
Lymphoma, Mantle-Cell , Animals , Mice , Cell Line, Tumor , EGF Family of Proteins/metabolism , Endothelial Cells/metabolism , Lymphoma, Mantle-Cell/metabolism , Signal Transduction , Humans
2.
Front Immunol ; 13: 1033490, 2022.
Article in English | MEDLINE | ID: mdl-36505438

ABSTRACT

To date, the only curative treatment for high-risk or refractory hematologic malignancies non-responsive to standard chemotherapy is allogeneic hematopoietic transplantation (allo-HCT). Acute graft-versus-host disease (GVHD) is a donor T cell-mediated immunological disorder that is frequently fatal and the leading cause of non-relapse mortality (NRM) in patients post allo-HCT. The pathogenesis of acute GVHD involves recognition of minor and/or major HLA mismatched host antigens by donor T cells followed by expansion, migration and finally end-organ damage due to combination of inflammatory cytokine secretion and direct cytotoxic effects. The endothelium is a thin layer of endothelial cells (EC) that line the innermost portion of the blood vessels and a key regulator in vascular homeostasis and inflammatory responses. Endothelial cells are activated by a wide range of inflammatory mediators including bacterial products, contents released from dying/apoptotic cells and cytokines and respond by secreting cytokines/chemokines that facilitate the recruitment of innate and adaptive immune cells to the site of inflammation. Endothelial cells can also be damaged prior to transplant as well as by alloreactive donor T cells. Prolonged EC activation results in dysfunction that plays a role in multiple post-transplant complications including but not limited to veno-occlusive disease (VOD), transplant associated thrombotic microangiopathy (TA-TMA), and idiopathic pneumonia syndrome. In this mini review, we summarize the biology of endothelial cells, factors regulating EC activation and the role of ECs in inflammation and GVHD pathogenesis.


Subject(s)
Graft vs Host Disease , Hematologic Neoplasms , Humans , Endothelial Cells , Graft vs Host Disease/etiology , Transplantation, Homologous/adverse effects , Tissue Donors , Inflammation
3.
Blood Adv ; 6(7): 2403-2408, 2022 04 12.
Article in English | MEDLINE | ID: mdl-34654057

ABSTRACT

Acute graft-versus-host disease (aGVHD) is the second most common cause of death after allogeneic hematopoietic stem cell transplantation (allo-HSCT), underscoring the need for novel therapies. Based on previous work that endothelial cell dysfunction is present in aGVHD and that epidermal growth factor-like domain 7 (EGFL7) plays a significant role in decreasing inflammation by repressing endothelial cell activation and T-cell migration, we hypothesized that increasing EGFL7 levels after allo-HSCT will diminish the severity of aGVHD. Here, we show that treatment with recombinant EGFL7 (rEGFL7) in 2 different murine models of aGVHD decreases aGVHD severity and improves survival in recipient mice after allogeneic transplantation with respect to controls without affecting graft-versus-leukemia effect. Furthermore, we showed that rEGFL7 treatment results in higher thymocytes, T, B, and dendritic cell counts in recipient mice after allo-HSCT. This study constitutes a proof of concept of the ability of rEGFL7 therapy to reduce GHVD severity and mortality after allo-HSCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Transplantation/adverse effects , Endothelial Cells , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Mice , Transplantation, Homologous
4.
Front Immunol ; 13: 1125035, 2022.
Article in English | MEDLINE | ID: mdl-36685565

ABSTRACT

[This corrects the article DOI: 10.3389/fimmu.2022.1033490.].

5.
Front Oncol ; 11: 760789, 2021.
Article in English | MEDLINE | ID: mdl-34722316

ABSTRACT

Acute graft-versus-host disease (GVHD) is the leading cause of non-relapse mortality following allogeneic hematopoietic cell transplantation. The majority of patients non-responsive to front line treatment with steroids have an estimated overall 2-year survival rate of only 10%. Bromodomain and extra-terminal domain (BET) proteins influence inflammatory gene transcription, and therefore represent a potential target to mitigate inflammation central to acute GVHD pathogenesis. Using potent and selective BET inhibitors Plexxikon-51107 and -2853 (PLX51107 and PLX2853), we show that BET inhibition significantly improves survival and reduces disease progression in murine models of acute GVHD without sacrificing the beneficial graft-versus-leukemia response. BET inhibition reduces T cell alloreactive proliferation, decreases inflammatory cytokine production, and impairs dendritic cell maturation both in vitro and in vivo. RNA sequencing studies in human T cells revealed that BET inhibition impacts inflammatory IL-17 and IL-12 gene expression signatures, and Chromatin Immunoprecipitation (ChIP)-sequencing revealed that BRD4 binds directly to the IL-23R gene locus. BET inhibition results in decreased IL-23R expression and function as demonstrated by decreased phosphorylation of STAT3 in response to IL-23 stimulation in human T cells in vitro as well as in mouse donor T cells in vivo. Furthermore, PLX2853 significantly reduced IL-23R+ and pathogenic CD4+ IFNγ+ IL-17+ double positive T cell infiltration in gastrointestinal tissues in an acute GVHD murine model. Our findings identify a role for BET proteins in regulating the IL-23R/STAT3/IL-17 pathway. Based on our preclinical data presented here, PLX51107 will enter clinical trial for refractory acute GVHD in a Phase 1 safety, biological efficacy trial.

8.
Nat Commun ; 11(1): 3702, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32710081

ABSTRACT

Spinal cord injury (SCI) causes immune dysfunction, increasing the risk of infectious morbidity and mortality. Since bone marrow hematopoiesis is essential for proper immune function, we hypothesize that SCI disrupts bone marrow hematopoiesis. Indeed, SCI causes excessive proliferation of bone marrow hematopoietic stem and progenitor cells (HSPC), but these cells cannot leave the bone marrow, even after challenging the host with a potent inflammatory stimulus. Sequestration of HSPCs in bone marrow after SCI is linked to aberrant chemotactic signaling that can be reversed by post-injury injections of Plerixafor (AMD3100), a small molecule inhibitor of CXCR4. Even though Plerixafor liberates HSPCs and mature immune cells from bone marrow, competitive repopulation assays show that the intrinsic long-term functional capacity of HSPCs is still impaired in SCI mice. Together, our data suggest that SCI causes an acquired bone marrow failure syndrome that may contribute to chronic immune dysfunction.


Subject(s)
Bone Marrow Failure Disorders/etiology , Bone Marrow/metabolism , Spinal Cord Injuries/complications , Animals , Benzylamines , Bone Marrow/pathology , Bone Marrow Cells , Bone Marrow Failure Disorders/pathology , Cell Proliferation , Chemokine CXCL12 , Cyclams , Disease Models, Animal , Female , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction , Spinal Cord Injuries/immunology
10.
Haematologica ; 105(3): 721-729, 2020 03.
Article in English | MEDLINE | ID: mdl-31413100

ABSTRACT

Leukemia stem cells (LSC) are more resistant to standard chemotherapy and their persistence during remission can cause relapse, which is still one of the major clinical challenges in the treatment of acute myeloid leukemia (AML). A better understanding of the mutational patterns and the prognostic impact of molecular markers associated with stemness could lead to better clinical management and improve patients' outcomes. We applied a previously described 17-gene expression score comprising genes differently expressed between LSC and leukemic bulk blasts, for 934 adult patients with de novo AML, and studied associations of the 17-gene LSC score with clinical data and mutation status of 81 genes recurrently mutated in cancer and leukemia. We found that patients with a high 17-gene score were older and had more mutations. The 17-gene score was found to have a prognostic impact in both younger (aged <60 years) and older (aged ≥60 years) patients with AML. We also analyzed the 17-gene LSC score in the context of the 2017 European LeukemiaNet genetic-risk classification and found that for younger patients the score refined the classification, and identified patients currently classified in the European LeukemiaNet Favorable-risk category who had a worse outcome.


Subject(s)
Leukemia, Myeloid, Acute , Adult , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Middle Aged , Mutation , Prognosis , Stem Cells , Treatment Outcome
11.
Clin Cancer Res ; 26(3): 669-678, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31672772

ABSTRACT

PURPOSE: EGF-like domain 7 (EGFL7) is a secreted protein and recently has been shown to play an important role in acute myeloid leukemia (AML); however, the underlying mechanism by which EGFL7 promotes leukemogenesis is largely unknown. EXPERIMENTAL DESIGN: Using an antibody interaction array, we measured the ability of EGFL7 to bind directly approximately 400 proteins expressed by primary AML blasts. Primary patient samples were stimulated in vitro with recombinant EGFL7 (rEGFL7) or anti-EGFL7 blocking antibody to assess alterations in downstream signaling and the ability to effect blast differentiation and survival. We treated three independent AML models with anti-EGFL7 or IgG1 control to determine whether anti-EGFL7 could prolong survival in vivo. RESULTS: We found EGFL7 significantly binds several signaling proteins important for normal and malignant hematopoiesis including NOTCH. Stimulation of AML blasts with rEGFL7 reduced NOTCH intracellular domain and NOTCH target gene expression while treatment with an anti-EGFL7 blocking antibody resulted in reactivation of NOTCH signaling, increased differentiation, and apoptosis. Competitive ligand-binding assays showed rEGFL7 inhibits DELTA-like (DLL) 4-mediated NOTCH activation while anti-EGFL7 combined with DLL4 significantly increased NOTCH activation and induced apoptosis. Using three different AML mouse models, we demonstrated that in vivo treatment with anti-EGFL7 alone results in increased survival. CONCLUSIONS: Our data demonstrate that EGFL7 contributes to NOTCH silencing in AML by antagonizing canonical NOTCH ligand binding. Reactivation of NOTCH signaling in vivo using anti-EGFL7 results in prolonged survival of leukemic mice, supporting the use of EGFL7 as a novel therapeutic target in AML.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Calcium-Binding Proteins/metabolism , EGF Family of Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Notch/antagonists & inhibitors , Animals , Apoptosis , Calcium-Binding Proteins/genetics , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , EGF Family of Proteins/genetics , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Notch/metabolism , Signal Transduction
12.
Nat Commun ; 10(1): 5351, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31767858

ABSTRACT

Long non-coding RNAs (lncRNAs) are important regulatory molecules that are implicated in cellular physiology and pathology. In this work, we dissect the functional role of the HOXB-AS3 lncRNA in patients with NPM1-mutated (NPM1mut) acute myeloid leukemia (AML). We show that HOXB-AS3 regulates the proliferative capacity of NPM1mut AML blasts in vitro and in vivo. HOXB-AS3 is shown to interact with the ErbB3-binding protein 1 (EBP1) and guide EBP1 to the ribosomal DNA locus. Via this mechanism, HOXB-AS3 regulates ribosomal RNA transcription and de novo protein synthesis. We propose that in the context of NPM1 mutations, HOXB-AS3 overexpression acts as a compensatory mechanism, which allows adequate protein production in leukemic blasts.


Subject(s)
Leukemia, Myeloid/genetics , Mutation , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Ribosomal/genetics , Transcription, Genetic , Acute Disease , Animals , Cell Line, Tumor , Cell Proliferation , HEK293 Cells , Humans , K562 Cells , Leukemia, Myeloid/pathology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nucleophosmin , Protein Biosynthesis/genetics , THP-1 Cells , Transplantation, Heterologous
13.
Leukemia ; 33(9): 2169-2182, 2019 09.
Article in English | MEDLINE | ID: mdl-30858548

ABSTRACT

In acute myeloid leukemia (AML), novel therapies are needed to target not only the rapidly dividing AML blasts but also the distinct population of leukemia stem cells (LSCs), which have abnormal self-renewal capacity and increased chemotherapy resistance. Elucidation of the expression and function of deregulated genes in LSCs is critical to specifically target LSCs and may consequently lead to improving outcomes of AML patients. Here, we correlated long non-coding RNA (lncRNA) expression profiles obtained from two RNA-seq datasets of 375 younger (aged <60 years) 76 older (≥60 years) adults with cytogenetically normal AML with a 'core enriched' (CE) gene expression signature (GES) associated with LSCs. We identified a LSC-specific signature of 111 lncRNAs that correlated strongly with the CE-GES. Among the top upregulated LSC-associated lncRNAs, we identified the lncRNA DANCR. Further experiments confirmed that DANCR is upregulated in functionally validated LSC-enriched populations. DANCR knock-down in LSCs resulted in decreased stem-cell renewal and quiescence. Furthermore, we showed that targeting Dancr in vivo using a primary murine model of AML (expressing both Mll partial tandem duplication/Flt3 internal tandem duplication) prolonged the survival of mice after serial transplantation. Our data suggest that LSCs have a distinct lncRNA signature with functional relevance and therapeutic potential.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Acute Disease , Animals , Cell Self Renewal/genetics , Female , Humans , Male , Mice , Middle Aged
14.
Proc Natl Acad Sci U S A ; 115(51): 13069-13074, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30478046

ABSTRACT

MicroRNAs (miRNAs) have been extensively reported to be associated with hematological malignancies. The loss of miR-15a/16-1 at chromosome 13q14 is a hallmark of most of human chronic lymphocytic leukemia (CLL). Deletion of murine miR-15a/16-1 and miR-15b/16-2 has been demonstrated to promote B cell malignancies. Here, we evaluate the biological role of miR-15/16 clusters, crossbreeding miR-15a/16-1 and miR-15b/16-2 knockout mice. Unexpectedly, the complete deletion of both clusters promoted myeloproliferative disorders in the majority of the mice by the age of 5 months with a penetrance of 70%. These mice showed a significant enlargement of spleen and abnormal swelling of lymph nodes. Flow cytometry characterization demonstrated an expanded CD11b/Gr-1 double-positive myeloid population both in spleen and in bone marrow. The transplantation of splenocytes harvested from double-KO mice into wild-type recipient mice resulted in the development of myeloproliferative disorders, as observed in the donors. In vivo, miR-15/16 cluster deletion up-regulated the expression of Cyclin D1, Cyclin D2, and Bcl-2. Taken together, our findings identify a driver oncogenic role for miR-15/16 cluster deletion in different leukocytic cell lineages.


Subject(s)
Leukemia, Myeloid, Acute/etiology , MicroRNAs/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cyclins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/metabolism , Spleen/pathology
15.
Proc Natl Acad Sci U S A ; 114(23): E4641-E4647, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533390

ABSTRACT

Epithelial growth factor-like 7 (EGFL7) is a protein that is secreted by endothelial cells and plays an important role in angiogenesis. Although EGFL7 is aberrantly overexpressed in solid tumors, its role in leukemia has not been evaluated. Here, we report that levels of both EGFL7 mRNA and EGFL7 protein are increased in blasts of patients with acute myeloid leukemia (AML) compared with normal bone marrow cells. High EGFL7 mRNA expression associates with lower complete remission rates, and shorter event-free and overall survival in older (age ≥60 y) and younger (age <60 y) patients with cytogenetically normal AML. We further show that AML blasts secrete EGFL7 protein and that higher levels of EGFL7 protein are found in the sera from AML patients than in sera from healthy controls. Treatment of patient AML blasts with recombinant EGFL7 in vitro leads to increases in leukemic blast cell growth and levels of phosphorylated AKT. EGFL7 blockade with an anti-EGFL7 antibody reduced the growth potential and viability of AML cells. Our findings demonstrate that increased EGFL7 expression and secretion is an autocrine mechanism supporting growth of leukemic blasts in patients with AML.


Subject(s)
Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Angiogenic Proteins/antagonists & inhibitors , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Antibodies, Blocking/pharmacology , Calcium-Binding Proteins , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Disease-Free Survival , EGF Family of Proteins , Endothelial Growth Factors/antagonists & inhibitors , Female , Humans , Leukemia, Myeloid, Acute/therapy , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Prognosis , Proteins/metabolism , Proteins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Risk Factors , Up-Regulation , Young Adult
16.
Clin Cancer Res ; 22(24): 6142-6152, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27358488

ABSTRACT

PURPOSE: Selinexor, a selective inhibitor of XPO1, is currently being tested as single agent in clinical trials in acute myeloid leukemia (AML). However, considering the molecular complexity of AML, it is unlikely that AML can be cured with monotherapy. Therefore, we asked whether adding already established effective drugs such as topoisomerase (Topo) II inhibitors to selinexor will enhance its anti-leukemic effects in AML. EXPERIMENTAL DESIGN: The efficacy of combinatorial drug treatment using Topo II inhibitors (idarubicin, daunorubicin, mitoxantrone, etoposide) and selinexor was evaluated in established cellular and animal models of AML. RESULTS: Concomitant treatment with selinexor and Topo II inhibitors resulted in therapeutic synergy in AML cell lines and patient samples. Using a xenograft MV4-11 AML mouse model, we show that treatment with selinexor and idarubicin significantly prolongs survival of leukemic mice compared with each single therapy. CONCLUSIONS: Aberrant nuclear export and cytoplasmic localization of Topo IIα has been identified as one of the mechanisms leading to drug resistance in cancer. Here, we show that in a subset of patients with AML that express cytoplasmic Topo IIα, selinexor treatment results in nuclear retention of Topo IIα protein, resulting in increased sensitivity to idarubicin. Selinexor treatment of AML cells resulted in a c-MYC-dependent reduction of DNA damage repair genes (Rad51 and Chk1) mRNA and protein expression and subsequent inhibition of homologous recombination repair and increased sensitivity to Topo II inhibitors. The preclinical data reported here support further clinical studies using selinexor and Topo II inhibitors in combination to treat AML. Clin Cancer Res; 22(24); 6142-52. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Repair/drug effects , DNA Topoisomerases, Type II/metabolism , Hydrazines/pharmacology , Karyopherins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triazoles/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, SCID , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Topoisomerase II Inhibitors/pharmacology , Exportin 1 Protein
17.
J Clin Invest ; 124(4): 1512-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24590286

ABSTRACT

Aberrant expression of the secreted protein, acidic, cysteine-rich (osteonectin) (SPARC) gene, which encodes a matricellular protein that participates in normal tissue remodeling, is associated with a variety of diseases including cancer, but the contribution of SPARC to malignant growth remains controversial. We previously reported that SPARC was among the most upregulated genes in cytogenetically normal acute myeloid leukemia (CN-AML) patients with gene-expression profiles predictive of unfavorable outcome, such as mutations in isocitrate dehydrogenase 2 (IDH2-R172) and overexpression of the oncogenes brain and acute leukemia, cytoplasmic (BAALC) and v-ets erythroblastosis virus E26 oncogene homolog (ERG). In contrast, SPARC was downregulated in CN-AML patients harboring mutations in nucleophosmin (NPM1) that are associated with favorable prognosis. Based on these observations, we hypothesized that SPARC expression is clinically relevant in AML. Here, we found that SPARC overexpression is associated with adverse outcome in CN-AML patients and promotes aggressive leukemia growth in murine models of AML. In leukemia cells, SPARC expression was mediated by the SP1/NF-κB transactivation complex. Furthermore, secreted SPARC activated the integrin-linked kinase/AKT (ILK/AKT) pathway, likely via integrin interaction, and subsequent ß-catenin signaling, which is involved in leukemia cell self-renewal. Pharmacologic inhibition of the SP1/NF-κB complex resulted in SPARC downregulation and leukemia growth inhibition. Together, our data indicate that evaluation of SPARC expression has prognosticative value and SPARC is a potential therapeutic target for AML.


Subject(s)
Leukemia, Myeloid, Acute/etiology , Osteonectin/physiology , Adolescent , Adult , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Knockdown Techniques , Heterografts , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , NF-kappa B/metabolism , Nucleophosmin , Osteonectin/antagonists & inhibitors , Osteonectin/genetics , Prognosis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Young Adult , beta Catenin/metabolism
18.
Blood ; 122(23): 3778-83, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24085765

ABSTRACT

The coexpression of the MLL partial tandem duplication (PTD) and the FLT3 internal tandem duplication (ITD) mutations associate with a poor outcome in cytogenetically normal acute myeloid leukemia (AML). In mice, a double knock-in (dKI) of Mll(PTD/wt) and Flt3(ITD/wt) mutations induces spontaneous AML with an increase in DNA methyltransferases (Dnmt1, 3a, and 3b) and global DNA methylation index, thereby recapitulating its human AML counterpart. We determined that a regulator of Dnmts, miR-29b, is downregulated in bone marrow of dKI AML mice. Bortezomib exerted a dose-dependent increase in miR-29b expression in AML blasts ex vivo, followed by decreased Dnmts, reduced proliferation, and increased apoptosis. In vivo, bortezomib was not active against dKI AML, yet liposomal-encapsulated bortezomib, as a single agent, reversed downregulation of miR-29b in vivo and induced a long-term (90-day) disease-free remission in 80% of dKI AML mice that exhibited high leukemic burden at the start of therapy, yet showed no signs of relapse at autopsy. Taken together, these data support that liposomal bortezomib, as a single agent, eradicates Mll(PTD/wt):Flt3(ITD/wt) AML in mouse and may represent a powerful and potentially curative approach to high-risk human disease.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Myeloid-Lymphoid Leukemia Protein/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Agents/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , DNA Methylation , Drug Carriers , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Leukemia, Experimental/therapy , Leukemia, Myeloid, Acute/metabolism , Liposomes , Mice , Mice, Mutant Strains , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Proteasome Inhibitors/administration & dosage , Pyrazines/administration & dosage , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tandem Repeat Sequences
19.
J Clin Invest ; 123(10): 4144-57, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999433

ABSTRACT

The success of tyrosine kinase inhibitors (TKIs) in treating chronic myeloid leukemia (CML) depends on the requirement for BCR-ABL1 kinase activity in CML progenitors. However, CML quiescent HSCs are TKI resistant and represent a BCR-ABL1 kinase-independent disease reservoir. Here we have shown that persistence of leukemic HSCs in BM requires inhibition of the tumor suppressor protein phosphatase 2A (PP2A) and expression--but not activity--of the BCR-ABL1 oncogene. Examination of HSCs from CML patients and healthy individuals revealed that PP2A activity was suppressed in CML compared with normal HSCs. TKI-resistant CML quiescent HSCs showed increased levels of BCR-ABL1, but very low kinase activity. BCR-ABL1 expression, but not kinase function, was required for recruitment of JAK2, activation of a JAK2/ß-catenin survival/self-renewal pathway, and inhibition of PP2A. PP2A-activating drugs (PADs) markedly reduced survival and self-renewal of CML quiescent HSCs, but not normal quiescent HSCs, through BCR-ABL1 kinase-independent and PP2A-mediated inhibition of JAK2 and ß-catenin. This led to suppression of human leukemic, but not normal, HSC/progenitor survival in BM xenografts and interference with long-term maintenance of BCR-ABL1-positive HSCs in serial transplantation assays. Targeting the JAK2/PP2A/ß-catenin network in quiescent HSCs with PADs (e.g., FTY720) has the potential to treat TKI-refractory CML and relieve lifelong patient dependence on TKIs.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 2/metabolism , Animals , Apoptosis , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Enzyme Activators/pharmacology , Fingolimod Hydrochloride , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Humans , Janus Kinase 2/metabolism , K562 Cells , Mice , Mice, Transgenic , Neoplastic Stem Cells/enzymology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism
20.
Blood ; 121(13): 2474-82, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23335370

ABSTRACT

The p21-activated kinases (Paks) are serine/threonine kinases that are major effectors of the Rho guanosine 5'\x{2011}triphosphatase, Rac, and Cdc42. Rac and Cdc42 are known regulators of hematopoietic stem and progenitor cell (HSPC) function, however, a direct role for Paks in HSPCs has yet to be elucidated. Lin(-)Sca1(+)c-kit(+) (LSK) cells from wild-type mice were transduced with retrovirus expressing Pak inhibitory domain (PID), a well-characterized inhibitor of Pak activation. Defects in marrow homing and in vitro cell migration, assembly of the actin cytoskeleton, proliferation, and survival were associated with engraftment failure of PID-LSK. The PID-LSK demonstrated decreased phosphorylation of extracellular signal-regulated kinase (ERK), whereas constitutive activation of ERK in these cells led to rescue of hematopoietic progenitor cell proliferation in vitro and partial rescue of Pak-deficient HSPC homing and engraftment in vivo. Using conditional knock-out mice, we demonstrate that among group A Paks, Pak2(-/-) HSPC show reduced homing to the bone marrow and altered cell shape similar to PID-LSK cells in vitro and are completely defective in HSPC engraftment. These data demonstrate that Pak proteins are key components of multiple engraftment-associated HSPC functions and play a direct role in activation of ERK in HSPCs, and that Pak2 is specifically essential for HSPC engraftment.


Subject(s)
Cell Movement/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , p21-Activated Kinases/physiology , Animals , Cell Movement/physiology , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , p21-Activated Kinases/genetics , rac GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...