Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 30(6): 832-850.e6, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267917

ABSTRACT

Remodeling of the tissue niche is often evident in diseases, yet, the stromal alterations and their contribution to pathogenesis are poorly characterized. Bone marrow fibrosis is a maladaptive feature of primary myelofibrosis (PMF). We performed lineage tracing and found that most collagen-expressing myofibroblasts were derived from leptin-receptor-positive (LepR+) mesenchymal cells, whereas a minority were from Gli1-lineage cells. Deletion of Gli1 did not impact PMF. Unbiased single-cell RNA sequencing (scRNA-seq) confirmed that virtually all myofibroblasts originated from LepR-lineage cells, with reduced expression of hematopoietic niche factors and increased expression of fibrogenic factors. Concurrently, endothelial cells upregulated arteriolar-signature genes. Pericytes and Sox10+ glial cells expanded drastically with heightened cell-cell signaling, suggesting important functional roles in PMF. Chemical or genetic ablation of bone marrow glial cells ameliorated fibrosis and improved other pathology in PMF. Thus, PMF involves complex remodeling of the bone marrow microenvironment, and glial cells represent a promising therapeutic target.


Subject(s)
Primary Myelofibrosis , Humans , Primary Myelofibrosis/drug therapy , Zinc Finger Protein GLI1/metabolism , Endothelial Cells/metabolism , Bone Marrow/metabolism , Neuroglia/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
2.
Expert Rev Anticancer Ther ; 21(6): 573-581, 2021 06.
Article in English | MEDLINE | ID: mdl-33593233

ABSTRACT

INTRODUCTION: Acute myeloid leukemia (AML) is an aggressive blood cancer that proves fatal for the majority of affected individuals. Older patients are particularly vulnerable due to more unfavorable disease biology and diminished ability to tolerate intensive induction chemotherapy (ICT). Safer, more efficacious therapies are desperately needed. AREAS COVERED: We briefly summarize the challenges facing AML treatment and introduce the rapidly expanding therapeutic landscape. Our focus is on the Hedgehog (Hh) pathway and how preclinical evidence has spurred the clinical development of selective inhibitors for oncology indications. Glasdegib is the first Hh pathway inhibitor approved for the treatment of a hematologic malignancy, and we review its pharmacology, safety, efficacy, and potential clinical impact in AML patients. EXPERT OPINION: Advances in the mechanistic understanding of AML have started to translate into improved therapeutic options for patients with contraindications to ICT. Glasdegib improved overall survival in this population when combined with low-dose cytarabine. While an encouraging development for these difficult to treat patients, alternative combination therapy approaches such as venetoclax plus azacitidine have gained greater clinical traction. Further investigation of glasdegib combination strategies and predictive biomarkers, particularly in regard to overcoming chemoresistance and preventing relapse, is needed to better define its clinical utility.


Subject(s)
Hedgehog Proteins , Leukemia, Myeloid, Acute , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzimidazoles , Cytarabine/adverse effects , Hedgehog Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Phenylurea Compounds/adverse effects
3.
Bioessays ; 40(3)2018 03.
Article in English | MEDLINE | ID: mdl-29384206

ABSTRACT

Various cell types cooperate to create a highly organized and dynamic micro-environmental niche in the bone marrow. Over the past several years, the field has increasingly recognized the critical roles of the interplay between bone marrow environment and hematopoietic cells in normal and deranged hematopoiesis. These advances rely on several new technologies that have allowed us to characterize the identity and roles of these niches in great detail. Here, we review the progress of the last several years, list some of the outstanding questions in the field and propose ways to target the diseased environment to better treat hematologic diseases. Understanding the extrinsic regulation by the niche will help boost hematopoiesis for regenerative medicine. Based on natural development of hematologic malignancies, we propose that combinatory targeting the niche and hematopoietic intrinsic mechanisms in early stages of hematopoietic malignancies may help eliminate minimal residual disease and have the highest efficacy.


Subject(s)
Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/drug therapy , Hematopoiesis/drug effects , Neoplastic Stem Cells/drug effects , Stem Cell Niche/drug effects , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Molecular Targeted Therapy/methods , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm, Residual , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Regenerative Medicine/methods , Stem Cell Niche/genetics , Tumor Microenvironment/genetics
4.
Leukemia ; 27(7): 1548-1557, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23455394

ABSTRACT

Acute promyelocytic leukemia (APL) is initiated by the PML-RARA (PR) fusion oncogene and has a characteristic expression profile that includes high levels of the Notch ligand Jagged-1 (JAG1). In this study, we used a series of bioinformatic, in vitro, and in vivo assays to assess the role of Notch signaling in human APL samples, and in a PML-RARA knock-in mouse model of APL (Ctsg-PML-RARA). We identified a Notch expression signature in both human primary APL cells and in Kit+Lin-Sca1+ cells from pre-leukemic Ctsg-PML-RARA mice. Both genetic and pharmacologic inhibition of Notch signaling abrogated the enhanced self-renewal seen in hematopoietic stem/progenitor cells from pre-leukemic Ctsg-PML-RARA mice, but had no influence on cells from age-matched wild-type mice. In addition, six of nine murine APL tumors tested displayed diminished growth in vitro when Notch signaling was inhibited pharmacologically. Finally, we found that genetic inhibition of Notch signaling with a dominant-negative Mastermind-like protein reduced APL growth in vivo in a subset of tumors. These findings expand the role of Notch signaling in hematopoietic diseases, and further define the mechanistic events important for PML-RARA-mediated leukemogenesis.


Subject(s)
Leukemia, Promyelocytic, Acute/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cathepsin G/genetics , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Oncogene Proteins, Fusion/genetics , Receptor, Notch1/genetics , Serrate-Jagged Proteins , Signal Transduction/genetics
5.
Blood ; 121(9): 1633-43, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23297133

ABSTRACT

Acute myeloid leukemia (AML) is characterized by dysregulated gene expression and abnormal patterns of DNA methylation; the relationship between these events is unclear. Many AML patients are now being treated with hypomethylating agents, such as decitabine (DAC), although the mechanisms by which it induces remissions remain unknown. The goal of this study was to use a novel stromal coculture assay that can expand primary AML cells to identify the immediate changes induced by DAC with a dose (100nM) that decreases total 5-methylcytosine content and reactivates imprinted genes (without causing myeloid differentiation, which would confound downstream genomic analyses). Using array-based technologies, we found that DAC treatment caused global hypomethylation in all samples (with a preference for regions with higher levels of baseline methylation), yet there was limited correlation between changes in methylation and gene expression. Moreover, the patterns of methylation and gene expression across the samples were primarily determined by the intrinsic properties of the primary cells, rather than DAC treatment. Although DAC induces hypomethylation, we could not identify canonical target genes that are altered by DAC in primary AML cells, suggesting that the mechanism of action of DAC is more complex than previously recognized.


Subject(s)
Azacitidine/analogs & derivatives , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/genetics , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/administration & dosage , Azacitidine/pharmacology , Cells, Cultured , CpG Islands/drug effects , CpG Islands/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Dose-Response Relationship, Drug , Gene Expression Profiling , Genome, Human/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Microarray Analysis , Primary Cell Culture , Time Factors
6.
Neurotherapeutics ; 6(3): 500-12, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19560740

ABSTRACT

Molecularly targeted therapies are transforming the care of patients with malignant gliomas, including glioblastoma, the most common malignant primary brain tumor of adults. With an arsenal of small molecule inhibitors and antibodies that target key components of the signal transduction machinery that are commonly activated in gliomas, neuro-oncologists and neurosurgeons are poised to transform the care of these patients. Nonetheless, successful application of targeted therapies remains a challenge. Strategies are lacking for directing kinase inhibitor or other pathway-specific therapies to individual patients most likely to benefit. In addition, response to targeted agents is determined not only by the presence of the key mutant kinases, but also by other critical changes in the molecular circuitry of cancer cells, such as loss of key tumor suppressor proteins, the selection for kinase-resistant mutants, and the deregulation of feedback loops. Understanding these signaling networks, and studying them in patients, will be critical for developing rational combination therapies to suppress resistance for malignant glioma patients. Here we review the current status of molecular targeted therapies for malignant gliomas. We focus initially on identifying some of the insights gained to date from targeting the EGFR/PI3K/Akt/mTOR signaling pathway in patients and on how this has led toward a reconceptualization of some of the challenges and directions for targeted treatment. We describe how advances from the world of genomics have the potential to transform our approaches toward targeted therapy, and describe how a deeper understanding of the complex nature of cancer, and its adeptness at rewiring molecular circuitry to evade targeted agents, has raised new challenges and identified new leads.


Subject(s)
Glioma/metabolism , Glioma/therapy , Signal Transduction/physiology , Animals , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Feedback, Physiological/physiology , Glioma/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...