Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(11): 991, 2021 10 23.
Article in English | MEDLINE | ID: mdl-34689158

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive malignant disease that is responsible for approximately 15% of breast cancers. The standard of care relies on surgery and chemotherapy but the prognosis is poor and there is an urgent need for new therapeutic strategies. Recent in silico studies have revealed an inverse correlation between recurrence-free survival and the level of cyclin-dependent kinase 8 (CDK8) in breast cancer patients. CDK8 is known to have a role in natural killer (NK) cell cytotoxicity, but its function in TNBC progression and immune cell recognition or escape has not been investigated. We have used a murine model of orthotopic breast cancer to study the tumor-intrinsic role of CDK8 in TNBC. Knockdown of CDK8 in TNBC cells impairs tumor regrowth upon surgical removal and prevents metastasis. In the absence of CDK8, the epithelial-to-mesenchymal transition (EMT) is impaired and immune-mediated tumor-cell clearance is facilitated. CDK8 drives EMT in TNBC cells in a kinase-independent manner. In vivo experiments have confirmed that CDK8 is a crucial regulator of NK-cell-mediated immune evasion in TNBC. The studies also show that CDK8 is involved in regulating the checkpoint inhibitor programmed death-ligand 1 (PD-L1). The CDK8-PD-L1 axis is found in mouse and human TNBC cells, underlining the importance of CDK8-driven immune cell evasion in these highly aggressive breast cancer cells. Our data link CDK8 to PD-L1 expression and provide a rationale for investigating the possibility of CDK8-directed therapy for TNBC.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Killer Cells, Natural/metabolism , Triple Negative Breast Neoplasms/genetics , Animals , Humans , Mice , Triple Negative Breast Neoplasms/pathology
2.
Blood Adv ; 5(1): 39-53, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33570624

ABSTRACT

Studies of molecular mechanisms of hematopoiesis and leukemogenesis are hampered by the unavailability of progenitor cell lines that accurately mimic the situation in vivo. We now report a robust method to generate and maintain LSK (Lin-, Sca-1+, c-Kit+) cells, which closely resemble MPP1 cells. HPCLSKs reconstitute hematopoiesis in lethally irradiated recipient mice over >8 months. Upon transformation with different oncogenes including BCR/ABL, FLT3-ITD, or MLL-AF9, their leukemic counterparts maintain stem cell properties in vitro and recapitulate leukemia formation in vivo. The method to generate HPCLSKs can be applied to transgenic mice, and we illustrate it for CDK6-deficient animals. Upon BCR/ABLp210 transformation, HPCLSKsCdk6-/- induce disease with a significantly enhanced latency and reduced incidence, showing the importance of CDK6 in leukemia formation. Studies of the CDK6 transcriptome in murine HPCLSK and human BCR/ABL+ cells have verified that certain pathways depend on CDK6 and have uncovered a novel CDK6-dependent signature, suggesting a role for CDK6 in leukemic progenitor cell homing. Loss of CDK6 may thus lead to a defect in homing. The HPCLSK system represents a unique tool for combined in vitro and in vivo studies and enables the production of large quantities of genetically modifiable hematopoietic or leukemic stem/progenitor cells.


Subject(s)
Fusion Proteins, bcr-abl , Hematopoietic Stem Cells , Animals , Hematopoiesis , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Eur J Immunol ; 50(6): 880-890, 2020 06.
Article in English | MEDLINE | ID: mdl-32052406

ABSTRACT

NK cells are innate lymphocytes responsible for lysis of pathogen-infected and transformed cells. One of the major activating receptors required for target cell recognition is the NK group 2D (NKG2D) receptor. Numerous reports show the necessity of NKG2D for effective tumor immune surveillance. Further studies identified NKG2D as a key element allowing tumor immune escape. We here use a mouse model with restricted deletion of NKG2D in mature NKp46+ cells (NKG2DΔNK ). NKG2DΔNK NK cells develop normally, have an unaltered IFN-γ production but kill tumor cell lines expressing NKG2D ligands (NKG2DLs) less efficiently. However, upon long-term stimulation with IL-2, NKG2D-deficient NK cells show increased levels of the lytic molecule perforin. Thus, our findings demonstrate a dual function of NKG2D for NK cell cytotoxicity; while NKG2D is a crucial trigger for cytotoxicity of tumor cells expressing activating ligands it is also capable to limit perforin production in IL-2 activated NK cells.


Subject(s)
Interleukin-2/pharmacology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Pore Forming Cytotoxic Proteins/immunology , Animals , Cell Line, Tumor , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/pathology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , Pore Forming Cytotoxic Proteins/genetics
4.
Nat Commun ; 10(1): 4741, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31628323

ABSTRACT

Cyclin-dependent kinases (CDKs) are frequently deregulated in cancer and represent promising drug targets. We provide evidence that CDK8 has a key role in B-ALL. Loss of CDK8 in leukemia mouse models significantly enhances disease latency and prevents disease maintenance. Loss of CDK8 is associated with pronounced transcriptional changes, whereas inhibiting CDK8 kinase activity has minimal effects. Gene set enrichment analysis suggests that the mTOR signaling pathway is deregulated in CDK8-deficient cells and, accordingly, these cells are highly sensitive to mTOR inhibitors. Analysis of large cohorts of human ALL and AML patients reveals a significant correlation between the level of CDK8 and of mTOR pathway members. We have synthesized a small molecule YKL-06-101 that combines mTOR inhibition and degradation of CDK8, and induces cell death in human leukemic cells. We propose that simultaneous CDK8 degradation and mTOR inhibition might represent a potential therapeutic strategy for the treatment of ALL patients.


Subject(s)
Cyclin-Dependent Kinase 8/metabolism , Disease Models, Animal , Fusion Proteins, bcr-abl/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Humans , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
5.
Pharmaceuticals (Basel) ; 12(2)2019 Jun 19.
Article in English | MEDLINE | ID: mdl-31248103

ABSTRACT

Improvements in cancer therapy frequently stem from the development of new small-molecule inhibitors, paralleled by the identification of biomarkers that can predict the treatment response. Recent evidence supports the idea that cyclin-dependent kinase 8 (CDK8) may represent a potential drug target for breast and prostate cancer, although no CDK8 inhibitors have entered the clinics. As the available inhibitors have been recently reviewed, we focus on the biological functions of CDK8 and provide an overview of the complexity of CDK8-dependent signaling throughout evolution and CDK8-dependent effects that may open novel treatment avenues.

6.
Neoplasia ; 20(6): 632-642, 2018 06.
Article in English | MEDLINE | ID: mdl-29772458

ABSTRACT

Leukemic stem cells (LSCs) are an emerging target of curative anti-leukemia therapy. In acute lymphoblastic leukemia (ALL), LSCs frequently express CD34 and often lack CD38. However, little is known about markers and targets expressed in ALL LSCs. We have examined marker- and target expression profiles in CD34+/CD38- LSCs in patients with Ph+ ALL (n = 22) and Ph- ALL (n = 27) by multi-color flow cytometry and qPCR. ALL LSCs expressed CD19 (B4), CD44 (Pgp-1), CD123 (IL-3RA), and CD184 (CXCR4) in all patients tested. Moreover, in various subgroups of patients, LSCs also displayed CD20 (MS4A1) (10/41 = 24%), CD22 (12/20 = 60%), CD33 (Siglec-3) (20/48 = 42%), CD52 (CAMPATH-1) (17/40 = 43%), IL-1RAP (13/29 = 45%), and/or CD135 (FLT3) (4/20 = 20%). CD25 (IL-2RA) and CD26 (DPPIV) were expressed on LSCs in Ph+ ALL exhibiting BCR/ABL1p210, whereas in Ph+ ALL with BCR/ABL1p190, LSCs variably expressed CD25 but did not express CD26. In Ph- ALL, CD34+/CD38- LSCs expressed IL-1RAP in 6/18 patients (33%), but did not express CD25 or CD26. Normal stem cells stained negative for CD25, CD26 and IL-1RAP, and expressed only low amounts of CD52. In xenotransplantation experiments, CD34+/CD38- and CD34+/CD38+ cells engrafted NSG mice after 12-20 weeks, and targeting with antibodies against CD33 and CD52 resulted in reduced engraftment. Together, LSCs in Ph+ and Ph- ALL display unique marker- and target expression profiles. In Ph+ ALL with BCR/ABL1p210, the LSC-phenotype closely resembles the marker-profile of CD34+/CD38- LSCs in chronic myeloid leukemia, confirming the close biologic relationship of these neoplasms. Targeting of LSCs with specific antibodies or related immunotherapies may facilitate LSC eradication in ALL.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Stem Cells/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line , Female , Gene Expression Regulation, Leukemic/physiology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Inbred NOD
7.
Cancer Immunol Res ; 6(4): 458-466, 2018 04.
Article in English | MEDLINE | ID: mdl-29386186

ABSTRACT

Cyclin-dependent kinase 8 (CDK8) is a member of the transcription-regulating CDK family. CDK8 activates or represses transcription by associating with the mediator complex or by regulating transcription factors. Oncogenic activity of CDK8 has been demonstrated in several cancer types. Targeting CDK8 represents a potential therapeutic strategy. Because knockdown of CDK8 in a natural killer (NK) cell line enhances cytotoxicity and NK cells provide the first line of immune defense against transformed cells, we asked whether inhibiting CDK8 would improve NK-cell antitumor responses. In this study, we investigated the role of CDK8 in NK-cell function in vivo using mice with conditional ablation of CDK8 in NKp46+ cells (Cdk8fl/flNcr1Cre). Regardless of CDK8 expression, NK cells develop and mature normally in bone marrow and spleen. However, CDK8 deletion increased expression of the lytic molecule perforin, which correlated with enhanced NK-cell cytotoxicity in vitro This translates into improved NK cell-mediated tumor surveillance in vivo in three independent models: B16F10 melanoma, v-abl+ lymphoma, and a slowly developing oncogene-driven leukemia. Our results thereby define a suppressive effect of CDK8 on NK-cell activity. Therapies that target CDK8 in cancer patients may enhance NK-cell responses against tumor cells. Cancer Immunol Res; 6(4); 458-66. ©2018 AACR.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Gene Deletion , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Neoplasms/genetics , Neoplasms/immunology , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Immunity, Innate , Killer Cells, Natural/cytology , Melanoma, Experimental , Mice , Mice, Transgenic , Neoplasms/pathology
8.
Mol Neurobiol ; 55(4): 3490-3498, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28508149

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder. Accumulating evidences suggest that PD might have a strong neurodevelopmental component. Among the genetic cases, mutations in the leucine-rich repeat kinase 2 (LRRK2) are well known to be disease causing. Although the molecular mechanism of the pathogenic LRRK2 function is not fully clear, inhibition of microRNA (miRNA) activity has been suggested to be among the pathogenic LRRK2 targets. Here, we demonstrate that the miRNA activity inhibition function of pathogenic LRRK2 is directly antagonized by the neuronal cell fate determinant TRIM32. These findings suggest that TRIM32 might be a modifier for PD and could be a novel therapeutic target.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , MicroRNAs/metabolism , Mutation/genetics , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Argonaute Proteins/metabolism , Cell Differentiation , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , Neurons/metabolism , Neurons/pathology , Protein Binding , RNA-Induced Silencing Complex/metabolism
9.
Br J Haematol ; 179(2): 229-241, 2017 10.
Article in English | MEDLINE | ID: mdl-28707321

ABSTRACT

The p21-activated kinases (PAKs) are key nodes in oncogenic signalling pathways controlling growth, survival, and motility of cancer cells. Their activity is increased in many human cancers and is associated with poor prognosis. To date, PAK deregulation has mainly been studied in solid tumours, where PAK1 and PAK4 are the main isoforms deregulated. We show that PAK1 and PAK2 are the critical isoforms in a BCR/ABL1+ haematopoietic malignancy. In suspension, leukaemic cells deficient for PAK1 and PAK2 undergo apoptosis, while the loss of either protein is well tolerated. Transfer of medium conditioned by shPAK2- but not shPAK1-expressing leukaemic cells interferes with endothelial cell growth. We found that leukaemic cells produce exosomes containing PAK2. Transfer of isolated exosomes supports endothelial cell proliferation. In parallel, we found that leukaemic cells explicitly require PAK2 to grow towards an extracellular matrix. PAK2-deficient cells fail to form colonies in methylcellulose and to induce lymphomas in vivo. PAK2 might therefore be the critical isoform in leukaemic cells by controlling tumour growth in a dual manner: vascularization via exosome-mediated transfer to endothelial cells and remodelling of the extracellular matrix. This finding suggests that the PAK2 isoform represents a promising target for the treatment of haematological diseases.


Subject(s)
Cell Proliferation , Fusion Proteins, bcr-abl/metabolism , Hematologic Neoplasms/metabolism , Leukemia/metabolism , Lymphoma/metabolism , p21-Activated Kinases/metabolism , Animals , Cell Line, Tumor , Endothelial Cells/metabolism , Endothelial Cells/pathology , Exosomes/genetics , Exosomes/metabolism , Exosomes/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fusion Proteins, bcr-abl/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Leukemia/genetics , Leukemia/pathology , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred NOD , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , p21-Activated Kinases/genetics
10.
Stem Cells Dev ; 22(18): 2487-96, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23600457

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene cause familial as well as sporadic Parkinson's disease (PD) that is characterized by an age-dependent degeneration of dopaminergic neurons. LRRK2 is strongly expressed in neural stem cells (NSCs), but still the exact molecular function of LRRK2 in these cells remains unknown. By performing a systemic analysis of the gene expression profile of LRRK2-deficient NSCs, we found that the expression of several PD-associated genes, such as oxidation and reduction in mitochondria, are deregulated on LRRK2 absence. Our data, indeed, indicate that LRRK2 regulates the level of cellular oxidative stress and thereby influences the survival of NSCs. Furthermore, the lack of LRRK2 leads to an up-regulation of neuronal differentiation-inducing processes, including the Let-7a pathway. On the other hand, the constitutive mutant of LRRK2(R1441G), known to cause PD, leads to down-regulation of the same pathway. In agreement with the function of Let-7a during neuronal differentiation, LRRK2-deficient NSCs differentiate faster than wild-type cells, while LRRK2(R1441G)-expressing NSCs show impaired neuronal differentiation. These results might help better characterize the molecular mechanisms underlying the role of LRRK2 in NSCs and would further improve potential cell-replacement strategies as well as drug discovery approaches.


Subject(s)
Cell Differentiation/genetics , Neural Stem Cells/metabolism , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Base Sequence , Cell Survival/genetics , Cell- and Tissue-Based Therapy , Cells, Cultured , Down-Regulation , Gene Expression Profiling , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Neural Stem Cells/cytology , Oxidation-Reduction , Oxidative Stress/genetics , Parkinson Disease/metabolism , Parkinson Disease/therapy , Protein Serine-Threonine Kinases/deficiency , Sequence Analysis, DNA , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...