Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Leukemia ; 36(11): 2634-2646, 2022 11.
Article in English | MEDLINE | ID: mdl-36163264

ABSTRACT

Disease progression and relapse of chronic myeloid leukemia (CML) are caused by therapy resistant leukemia stem cells (LSCs), and cure relies on their eradication. The microenvironment in the bone marrow (BM) is known to contribute to LSC maintenance and resistance. Although leukemic infiltration of the spleen is a hallmark of CML, it is unknown whether spleen cells form a niche that maintains LSCs. Here, we demonstrate that LSCs preferentially accumulate in the spleen and contribute to disease progression. Spleen LSCs were located in the red pulp close to red pulp macrophages (RPM) in CML patients and in a murine CML model. Pharmacologic and genetic depletion of RPM reduced LSCs and decreased their cell cycling activity in the spleen. Gene expression analysis revealed enriched stemness and decreased myeloid lineage differentiation in spleen leukemic stem and progenitor cells (LSPCs). These results demonstrate that splenic RPM form a niche that maintains CML LSCs in a quiescent state, resulting in disease progression and resistance to therapy.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid , Humans , Mice , Animals , Spleen , Neoplastic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid/genetics , Macrophages/metabolism , Disease Progression , Tumor Microenvironment
2.
Nat Commun ; 11(1): 1632, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32242021

ABSTRACT

Co-stimulatory signals, cytokines and transcription factors regulate the balance between effector and memory cell differentiation during T cell activation. Here, we analyse the role of the TRAF2-/NCK-interacting kinase (TNIK), a signaling molecule downstream of the tumor necrosis factor superfamily receptors such as CD27, in the regulation of CD8+ T cell fate during acute infection with lymphocytic choriomeningitis virus. Priming of CD8+ T cells induces a TNIK-dependent nuclear translocation of ß-catenin with consecutive Wnt pathway activation. TNIK-deficiency during T cell activation results in enhanced differentiation towards effector cells, glycolysis and apoptosis. TNIK signaling enriches for memory precursors by favouring symmetric over asymmetric cell division. This enlarges the pool of memory CD8+ T cells and increases their capacity to expand after re-infection in serial re-transplantation experiments. These findings reveal that TNIK is an important regulator of effector and memory T cell differentiation and induces a population of stem cell-like memory T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Humans , Immunologic Memory , Lymphocyte Activation , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Wnt Signaling Pathway
3.
Oncogene ; 38(5): 622-636, 2019 01.
Article in English | MEDLINE | ID: mdl-30171261

ABSTRACT

Oncogenic KRAS mutations comprise the largest subset of lung cancer defined by genetic alterations, but in the clinic no targeted therapies are available that effectively control mutational KRAS activation. Consequently, patients with KRAS-driven tumors are routinely treated with cytotoxic chemotherapy, which is often transiently effective owing to development of drug resistance. In this study, we show that hyperactivated mammalian target of rapamycin (mTOR) pathway is a characteristic hallmark of KRAS-mutant lung adenocarcinoma after chemotherapy treatment, and that KRAS-mutant lung cancer cells rely on persistent mTOR signaling to resist chemotherapeutic drugs. Coherently, mTOR inhibition circumvents the refractory phenotype and restores sensitivity of resistant KRAS-mutant lung cancer cells to chemotherapy. Importantly, drug combinations of clinically approved mTOR inhibitors and chemotherapy drugs synergize in inhibiting cell proliferation of KRAS-mutant cancer cells in vitro and in vivo, and the efficacy of this combination treatment correlates with the magnitude of mTOR activity induced by chemotherapy alone. These results pinpoint mTOR as a mechanism of resistance to chemotherapy in KRAS-mutant lung cancer and validate a rational and readily translatable strategy that combines mTOR inhibitors with standard chemotherapy to treat KRAS-mutant adenocarcinoma, the most common and deadliest lung cancer subset.


Subject(s)
Adenocarcinoma of Lung/enzymology , Drug Resistance, Neoplasm , Lung Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins p21(ras)/genetics , TOR Serine-Threonine Kinases/genetics
4.
Cancer Res ; 79(2): 346-359, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30389698

ABSTRACT

Hematopoiesis in patients with cancer is characterized by reduced production of red blood cells and an increase in myelopoiesis, which contributes to the immunosuppressive environment in cancer. Some tumors produce growth factors that directly stimulate myelopoiesis such as G-CSF or GM-CSF. However, for a majority of tumors that do not directly secrete hematopoietic growth factors, the mechanisms involved in the activation of myelopoiesis are poorly characterized. In this study, we document in different murine tumor models activated hematopoiesis with increased proliferation of long-term and short-term hematopoietic stem cells and myeloid progenitor cells. As a consequence, the frequency of myeloid-derived suppressor cells and its ratio to CD8+ T cells increased in tumor-bearing mice. Activation of hematopoiesis and myeloid differentiation in tumor-bearing mice was induced by TNFα, which was mainly secreted by activated CD4+ T cells. Therefore, the activated adaptive immune system in cancer induces emergency myelopoiesis and immunosuppression. SIGNIFICANCE: These findings characterize a regulatory circuit linking activated T cells to suppression of tumor-specific immune responses, providing a conceptual advance in the understanding of emergency-hematopoiesis in cancer and opening new targets for therapeutic approaches. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/2/346/F1.large.jpg.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Myelopoiesis/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Tumor Necrosis Factor-alpha/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology
5.
J Exp Med ; 210(3): 605-21, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23401488

ABSTRACT

Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasia arising from the oncogenic break point cluster region/Abelson murine leukemia viral oncogene homolog 1 translocation in hematopoietic stem cells (HSCs), resulting in a leukemia stem cell (LSC). Curing CML depends on the eradication of LSCs. Unfortunately, LSCs are resistant to current treatment strategies. The host's immune system is thought to contribute to disease control, and several immunotherapy strategies are under investigation. However, the interaction of the immune system with LSCs is poorly defined. In the present study, we use a murine CML model to show that LSCs express major histocompatibility complex (MHC) and co-stimulatory molecules and are recognized and killed by leukemia-specific CD8(+) effector CTLs in vitro. In contrast, therapeutic infusions of effector CTLs into CML mice in vivo failed to eradicate LSCs but, paradoxically, increased LSC numbers. LSC proliferation and differentiation was induced by CTL-secreted IFN-γ. Effector CTLs were only able to eliminate LSCs in a situation with minimal leukemia load where CTL-secreted IFN-γ levels were low. In addition, IFN-γ increased proliferation and colony formation of CD34(+) stem/progenitor cells from CML patients in vitro. Our study reveals a novel mechanism by which the immune system contributes to leukemia progression and may be important to improve T cell-based immunotherapy against leukemia.


Subject(s)
Interferon-gamma/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Neoplastic Stem Cells/pathology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Amino Acid Sequence , Animals , Cell Proliferation , Cytotoxicity, Immunologic , Humans , Immunotherapy, Adoptive , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , Neoplastic Stem Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...