Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Proc Natl Acad Sci U S A ; 114(11): E2243-E2252, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28246330

ABSTRACT

Rapid and efficient protocols to generate oligodendrocytes (OL) from human induced pluripotent stem cells (iPSC) are currently lacking, but may be a key technology to understand the biology of myelin diseases and to develop treatments for such disorders. Here, we demonstrate that the induction of three transcription factors (SOX10, OLIG2, NKX6.2) in iPSC-derived neural progenitor cells is sufficient to rapidly generate O4+ OL with an efficiency of up to 70% in 28 d and a global gene-expression profile comparable to primary human OL. We further demonstrate that iPSC-derived OL disperse and myelinate the CNS of Mbpshi/shiRag-/- mice during development and after demyelination, are suitable for in vitro myelination assays, disease modeling, and screening of pharmacological compounds potentially promoting oligodendroglial differentiation. Thus, the strategy presented here to generate OL from iPSC may facilitate the studying of human myelin diseases and the development of high-throughput screening platforms for drug discovery.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Transcription Factors/genetics , Animals , Biomarkers , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cell Death/genetics , Cell Lineage/genetics , Cells, Cultured , Cluster Analysis , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Ectopic Gene Expression , Gene Expression Profiling , Humans , Mice , Mutation , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oxidative Stress , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Transcription Factors/metabolism , Transcriptome , tau Proteins/genetics , tau Proteins/metabolism
2.
Ann Neurol ; 81(6): 811-824, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28470695

ABSTRACT

OBJECTIVE: Degeneration of oligodendroglial distal processes has been identified as an early event in multiple sclerosis (MS) lesion development. Our objective was to further define the development of the "dying-back" oligodendrocyte lesion in situ and to model the development and potential reversibility of such responses using dissociated cultures of adult human brain-derived oligodendrocytes. METHODS: In situ analyses were performed on glutaraldehyde-fixed thin sections of clinically acute and pathologically active cases of MS. In vitro studies were conducted using adult human brain-derived oligodendrocytes challenged by metabolic stress conditions (low nutrient/glucose). RESULTS: In situ analyses indicated a spectrum of myelin changes in the presence of morphologically intact oligodendrocytes; these included degeneration of the inner cytoplasmic tongue with increasing sizes of intramyelinic bleb formation that could result in radial fractures of the myelin sheath. Macrophages with ingested myelin fragments were identified only once the fragmentation was established. In vitro studies indicated that oligodendrocyte process retraction, which was linked to reduced glycolytic respiratory activity, is reversible until a critical time point. Subsequent cell death was not linked to caspase-3-dependent programs. Gene expression studies conducted at the latest reversible time point revealed reduced expression of pathways associated with cell process outgrowth and myelination, as well as with metabolic activity. INTERPRETATION: Our findings reveal the potential to protect and possibly restore myelin elaborated by existent oligodendrocytes in early and evolving MS lesions, and suggest the necessity of ongoing studies of the mechanisms underlying subsequent adult human oligodendrocyte cell death. Ann Neurol 2017;81:811-824.


Subject(s)
Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Animals , Caspase 3/metabolism , Cell Death , Humans , Rats , Rats, Sprague-Dawley
3.
Nat Metab ; 4(6): 693-710, 2022 06.
Article in English | MEDLINE | ID: mdl-35760868

ABSTRACT

Elevated production of collagen-rich extracellular matrix is a hallmark of cancer-associated fibroblasts (CAFs) and a central driver of cancer aggressiveness. Here we find that proline, a highly abundant amino acid in collagen proteins, is newly synthesized from glutamine in CAFs to make tumour collagen in breast cancer xenografts. PYCR1 is a key enzyme for proline synthesis and highly expressed in the stroma of breast cancer patients and in CAFs. Reducing PYCR1 levels in CAFs is sufficient to reduce tumour collagen production, tumour growth and metastatic spread in vivo and cancer cell proliferation in vitro. Both collagen and glutamine-derived proline synthesis in CAFs are epigenetically upregulated by increased pyruvate dehydrogenase-derived acetyl-CoA levels. PYCR1 is a cancer cell vulnerability and potential target for therapy; therefore, our work provides evidence that targeting PYCR1 may have the additional benefit of halting the production of a pro-tumorigenic extracellular matrix. Our work unveils new roles for CAF metabolism to support pro-tumorigenic collagen production.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Pyrroline Carboxylate Reductases/metabolism , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Glutamine/metabolism , Humans , Proline , delta-1-Pyrroline-5-Carboxylate Reductase
4.
Clin Cancer Res ; 26(3): 657-668, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31611282

ABSTRACT

PURPOSE: Emerging data suggest immune checkpoint inhibitors have reduced efficacy in heavily pretreated triple-negative breast cancers (TNBC), but underlying mechanisms are poorly understood. To better understand the phenotypic evolution of TNBCs, we studied the genomic and transcriptomic profiles of paired tumors from patients with TNBC. EXPERIMENTAL DESIGN: We collected paired primary and metastatic TNBC specimens from 43 patients and performed targeted exome sequencing and whole-transcriptome sequencing. From these efforts, we ascertained somatic mutation profiles, tumor mutational burden (TMB), TNBC molecular subtypes, and immune-related gene expression patterns. Stromal tumor-infiltrating lymphocytes (stromal TIL), recurrence-free survival, and overall survival were also analyzed. RESULTS: We observed a typical TNBC mutational landscape with minimal shifts in copy number or TMB over time. However, there were notable TNBC molecular subtype shifts, including increases in the Lehmann/Pietenpol-defined basal-like 1 (BL1, 11.4%-22.6%) and mesenchymal (M, 11.4%-22.6%) phenotypes, and a decrease in the immunomodulatory phenotype (IM, 31.4%-3.2%). The Burstein-defined basal-like immune-activated phenotype was also decreased (BLIA, 42.2%-17.2%). Among downregulated genes from metastases, we saw enrichment of immune-related Kyoto Encyclopedia of Genes and Genomes pathways and gene ontology (GO) terms, and decreased expression of immunomodulatory gene signatures (P < 0.03) and percent stromal TILs (P = 0.03). There was no clear association between stromal TILs and survival. CONCLUSIONS: We observed few mutational shifts, but largely consistent transcriptomic shifts in longitudinally paired TNBCs. Transcriptomic and IHC analyses revealed significantly reduced immune-activating gene expression signatures and TILs in recurrent TNBCs. These data may explain the observed lack of efficacy of immunotherapeutic agents in heavily pretreated TNBCs. Further studies are ongoing to better understand these initial observations.See related commentary by Savas and Loi, p. 526.


Subject(s)
Triple Negative Breast Neoplasms , Biomarkers, Tumor , Humans , Lymphocytes, Tumor-Infiltrating , Phenotype , Transcriptome
5.
J Clin Invest ; 129(4): 1785-1800, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30753167

ABSTRACT

Understanding the tumor immune microenvironment (TIME) promises to be key for optimal cancer therapy, especially in triple-negative breast cancer (TNBC). Integrating spatial resolution of immune cells with laser capture microdissection gene expression profiles, we defined distinct TIME stratification in TNBC, with implications for current therapies including immune checkpoint blockade. TNBCs with an immunoreactive microenvironment exhibited tumoral infiltration of granzyme B+CD8+ T cells (GzmB+CD8+ T cells), a type 1 IFN signature, and elevated expression of multiple immune inhibitory molecules including indoleamine 2,3-dioxygenase (IDO) and programmed cell death ligand 1 (PD-L1), and resulted in good outcomes. An "immune-cold" microenvironment with an absence of tumoral CD8+ T cells was defined by elevated expression of the immunosuppressive marker B7-H4, signatures of fibrotic stroma, and poor outcomes. A distinct poor-outcome immunomodulatory microenvironment, hitherto poorly characterized, exhibited stromal restriction of CD8+ T cells, stromal expression of PD-L1, and enrichment for signatures of cholesterol biosynthesis. Metasignatures defining these TIME subtypes allowed us to stratify TNBCs, predict outcomes, and identify potential therapeutic targets for TNBC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/immunology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/pathology , Cholesterol/immunology , Female , Granzymes/immunology , Humans , Interferon Type I/immunology , Triple Negative Breast Neoplasms/pathology
6.
Sci Rep ; 8(1): 13437, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30194392

ABSTRACT

Multiple sclerosis (MS) is an autoimmune, neurodegenerative disease but the molecular mechanisms underlying neurodegenerative aspects of the disease are poorly understood. microRNAs (miRNAs) are powerful regulators of gene expression that regulate numerous mRNAs simultaneously and can thus regulate programs of gene expression. Here, we describe miRNA expression in neurons captured from mice subjected to experimental autoimmune encephalomyelitis (EAE), a model of central nervous system (CNS) inflammation. Lumbar motor neurons and retinal neurons were laser captured from EAE mice and miRNA expression was assessed by next-generation sequencing and validated by qPCR. We describe 14 miRNAs that are differentially regulated in both neuronal subtypes and determine putative mRNA targets though in silico analysis. Several upregulated neuronal miRNAs are predicted to target pathways that could mediate repair and regeneration during EAE. This work identifies miRNAs that are affected by inflammation and suggests novel candidates that may be targeted to improve neuroprotection in the context of pathological inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , MicroRNAs/biosynthesis , Retinal Neurons/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , MicroRNAs/genetics , Retinal Neurons/pathology
7.
Cell Rep ; 23(5): 1476-1490, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719259

ABSTRACT

AXL is activated by its ligand GAS6 and is expressed in triple-negative breast cancer cells. In the current study, we report AXL expression in HER2-positive (HER2+) breast cancers where it correlates with poor patient survival. Using murine models of HER2+ breast cancer, Axl, but not its ligand Gas6, was found to be essential for metastasis. We determined that AXL is required for intravasation, extravasation, and growth at the metastatic site. We found that AXL is expressed in HER2+ cancers displaying epithelial-to-mesenchymal transition (EMT) signatures where it contributes to sustain EMT. Interfering with AXL in a patient-derived xenograft (PDX) impaired transforming growth factor ß (TGF-ß)-induced cell invasion. Last, pharmacological inhibition of AXL specifically decreased the metastatic burden of mice developing HER2+ breast cancer. Our data identify AXL as a potential anti-metastatic co-therapeutic target for the treatment of HER2+ breast cancers.


Subject(s)
Breast Neoplasms/mortality , Epithelial-Mesenchymal Transition , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Heterografts , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor, ErbB-2/genetics , Axl Receptor Tyrosine Kinase
9.
Cancer Res ; 78(19): 5618-5630, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30093561

ABSTRACT

The role of the stromal compartment in tumor progression is best illustrated in breast cancer bone metastases, where the stromal compartment supports tumor growth, albeit through poorly defined mechanisms. p38MAPKα is frequently expressed in tumor cells and surrounding stromal cells, and its expression levels correlate with poor prognosis. This observation led us to investigate whether inhibition of p38MAPKα could reduce breast cancer metastases in a clinically relevant model. Orally administered, small-molecule inhibitors of p38MAPKα or its downstream kinase MK2 each limited outgrowth of metastatic breast cancer cells in the bone and visceral organs. This effect was primarily mediated by inhibition of the p38MAPKα pathway within the stromal compartment. Beyond effectively limiting metastatic tumor growth, these inhibitors reduced tumor-associated and chemotherapy-induced bone loss, which is a devastating comorbidity that drastically affects quality of life for patients with cancer. These data underscore the vital role played by stromal-derived factors in tumor progression and identify the p38MAPK-MK2 pathway as a promising therapeutic target for metastatic disease and prevention of tumor-induced bone loss.Significance: Pharmacologically targeting the stromal p38MAPK-MK2 pathway limits metastatic breast cancer growth, preserves bone quality, and extends survival. Cancer Res; 78(19); 5618-30. ©2018 AACR.


Subject(s)
Antineoplastic Agents/adverse effects , Bone and Bones/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Administration, Oral , Animals , Bone Neoplasms/secondary , Bone and Bones/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Drug Therapy , Female , HEK293 Cells , Humans , Induction Chemotherapy , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Neoplasm Metastasis , Osteoclasts/metabolism , Paclitaxel/pharmacology , Prognosis , Quality of Life , Stromal Cells/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL