Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nature ; 618(7964): 374-382, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37225988

ABSTRACT

Cancer alters the function of multiple organs beyond those targeted by metastasis1,2. Here we show that inflammation, fatty liver and dysregulated metabolism are hallmarks of systemically affected livers in mouse models and in patients with extrahepatic metastasis. We identified tumour-derived extracellular vesicles and particles (EVPs) as crucial mediators of cancer-induced hepatic reprogramming, which could be reversed by reducing tumour EVP secretion via depletion of Rab27a. All EVP subpopulations, exosomes and principally exomeres, could dysregulate hepatic function. The fatty acid cargo of tumour EVPs-particularly palmitic acid-induced secretion of tumour necrosis factor (TNF) by Kupffer cells, generating a pro-inflammatory microenvironment, suppressing fatty acid metabolism and oxidative phosphorylation, and promoting fatty liver formation. Notably, Kupffer cell ablation or TNF blockade markedly decreased tumour-induced fatty liver generation. Tumour implantation or pre-treatment with tumour EVPs diminished cytochrome P450 gene expression and attenuated drug metabolism in a TNF-dependent manner. We also observed fatty liver and decreased cytochrome P450 expression at diagnosis in tumour-free livers of patients with pancreatic cancer who later developed extrahepatic metastasis, highlighting the clinical relevance of our findings. Notably, tumour EVP education enhanced side effects of chemotherapy, including bone marrow suppression and cardiotoxicity, suggesting that metabolic reprogramming of the liver by tumour-derived EVPs may limit chemotherapy tolerance in patients with cancer. Our results reveal how tumour-derived EVPs dysregulate hepatic function and their targetable potential, alongside TNF inhibition, for preventing fatty liver formation and enhancing the efficacy of chemotherapy.


Subject(s)
Extracellular Vesicles , Fatty Acids , Fatty Liver , Liver , Pancreatic Neoplasms , Animals , Mice , Cytochrome P-450 Enzyme System/genetics , Extracellular Vesicles/metabolism , Fatty Acids/metabolism , Fatty Liver/drug therapy , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Liver/metabolism , Liver/pathology , Liver/physiopathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Liver Neoplasms/secondary , Humans , Inflammation/metabolism , Palmitic Acid/metabolism , Kupffer Cells , Oxidative Phosphorylation , rab27 GTP-Binding Proteins/deficiency
2.
Nat Immunol ; 16(4): 397-405, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25729925

ABSTRACT

Signaling via the pre-T cell antigen receptor (pre-TCR) and the receptor Notch1 induces transient self-renewal (ß-selection) of TCRß(+) CD4(-)CD8(-) double-negative stage 3 (DN3) and DN4 progenitor cells that differentiate into CD4(+)CD8(+) double-positive (DP) thymocytes, which then rearrange the locus encoding the TCR α-chain (Tcra). Interleukin 7 (IL-7) promotes the survival of TCRß(-) DN thymocytes by inducing expression of the pro-survival molecule Bcl-2, but the functions of IL-7 during ß-selection have remained unclear. Here we found that IL-7 signaled TCRß(+) DN3 and DN4 thymocytes to upregulate genes encoding molecules involved in cell growth and repressed the gene encoding the transcriptional repressor Bcl-6. Accordingly, IL-7-deficient DN4 cells lacked trophic receptors and did not proliferate but rearranged Tcra prematurely and differentiated rapidly. Deletion of Bcl6 partially restored the self-renewal of DN4 cells in the absence of IL-7, but overexpression of BCL2 did not. Thus, IL-7 critically acts cooperatively with signaling via the pre-TCR and Notch1 to coordinate proliferation, differentiation and Tcra recombination during ß-selection.


Subject(s)
Interleukin-7/genetics , Receptor, Notch1/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Thymocytes/metabolism , Animals , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Cell Differentiation , Cell Proliferation , Cell Survival , Gene Expression Regulation , Interleukin-7/deficiency , Interleukin-7/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-bcl-6/deficiency , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Receptor, Notch1/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Recombination, Genetic , Signal Transduction , Thymocytes/cytology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
3.
STAR Protoc ; 5(1): 102754, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38096060

ABSTRACT

Characterization of isolated extracellular vesicles and particles (EVPs) is crucial for determining functions and biomarker potential. Here, we present a protocol to analyze size, number, morphology, and EVP protein cargo and to validate EVP proteins in both humans and mice. We describe steps for nanoparticle tracking analysis, transmission electron microscopy, single-EVP immunodetection, EVP proteomic mass spectrometry and bioinformatic analysis, and EVP protein validation by ExoELISA and western blot analysis. This allows for EVP cross-validation across different platforms. For complete details on the use and execution of this protocol, please refer to Hoshino et al.1.


Subject(s)
Extracellular Vesicles , Proteomics , Humans , Animals , Mice , Blotting, Western , Computational Biology , Mass Spectrometry
4.
Blood ; 117(4): 1184-95, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21097675

ABSTRACT

Lunatic Fringe (Lfng) enhances Notch1 activation by Delta-like 4 (DL4) to promote Notch1-dependent T-lineage commitment of thymus-seeding progenitors. Subsequently, Notch1 and T-cell receptor-ß (TCRß)-containing pre-TCR complexes signal CD4/CD8 double-negative 3 (DN3) committed T-cell progenitors to survive, proliferate, and differentiate into CD4/CD8 double-positive (DP) αß T-cell precursors. Few DP thymocytes develop without Notch1 or pre-TCR signals, whereas ectopic Notch1 activation causes T-cell leukemia. However, mechanisms of a Notch-pre-TCR collaboration during this "ß-selection" process are poorly understood. We genetically manipulated Lfng to attenuate or enhance Notch1 activation in DN3 thymocytes without inducing leukemogenesis. We show that Lfng temporally sustains DL-induced Notch1 signaling to prolong proliferative self-renewal of pre-DP thymocytes. Pre-TCR signaling greatly augmented Notch trophic functions to promote robust proliferation of pre-DP progenitors. In contrast, in the absence of DL/Notch signaling, pre-TCR-expressing progenitors rapidly atrophied and differentiated into DP thymocytes. Thus, Lfng prolongs Notch1 signaling to promote self-renewal more than differentiation during the early stages of ß-selection. Our data provide novel insights into the Notch-pre-TCR collaboration, and suggest that decreasing Lfng expression during the DN3-DP transition minimizes the potent leukemogenic potential of Notch1 signaling.


Subject(s)
Cell Proliferation , Glycosyltransferases/physiology , Intracellular Signaling Peptides and Proteins/physiology , Lymphoid Progenitor Cells/physiology , Membrane Proteins/physiology , Receptor, Notch1/physiology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/physiology , Adaptor Proteins, Signal Transducing , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Calcium-Binding Proteins , Cells, Cultured , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoid Progenitor Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/metabolism
5.
Front Oncol ; 13: 1163786, 2023.
Article in English | MEDLINE | ID: mdl-37350937

ABSTRACT

The discovery that primary tumors condition distant organ sites of future metastasis for seeding by disseminating tumor cells through a process described as the pre-metastatic niche (PMN) formation revolutionized our understanding of cancer progression and opened new avenues for therapeutic interventions. Given the inherent inefficiency of metastasis, PMN generation is crucial to ensure the survival of rare tumor cells in the otherwise hostile environments of metastatic organs. Early on, it was recognized that preparing the "soil" of the distal organ to support the outgrowth of metastatic cells is the initiating event in PMN development, achieved through the remodeling of the organ's extracellular matrix (ECM). Remote restructuring of ECM at future sites of metastasis under the influence of primary tumor-secreted factors is an iterative process orchestrated through the crosstalk between resident stromal cells, such as fibroblasts, epithelial and endothelial cells, and recruited innate immune cells. In this review, we will explore the ECM changes, cellular effectors, and the mechanisms of ECM remodeling throughout PMN progression, as well as its impact on shaping the PMN and ultimately promoting metastasis. Moreover, we highlight the clinical and translational implications of PMN ECM changes and opportunities for therapeutically targeting the ECM to hinder PMN formation.

6.
STAR Protoc ; 2(1): 100225, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33786456

ABSTRACT

We developed a modified protocol, based on differential ultracentrifugation (dUC), to isolate extracellular vesicles and particles (specifically exomeres) (EVPs) from various human and murine sources, including cell lines, surgically resected tumors and adjacent tissues, and bodily fluids, such as blood, lymphatic fluid, and bile. The diversity of these samples requires robust and highly reproducible protocols and refined isolation technology, such as asymmetric-flow field-flow fractionation (AF4). Our isolation protocol allows for preparation of EVPs for various downstream applications, including proteomic profiling. For complete details on the use and execution of this protocol, please refer to Hoshino et al. (2020).


Subject(s)
Body Fluids/chemistry , Centrifugation, Density Gradient , Extracellular Vesicles/chemistry , Fractionation, Field Flow , Proteomics , Animals , Cell Line , Humans , Mice
7.
Nat Cell Biol ; 21(11): 1403-1412, 2019 11.
Article in English | MEDLINE | ID: mdl-31685984

ABSTRACT

The development of effective therapies against brain metastasis is currently hindered by limitations in our understanding of the molecular mechanisms driving it. Here we define the contributions of tumour-secreted exosomes to brain metastatic colonization and demonstrate that pre-conditioning the brain microenvironment with exosomes from brain metastatic cells enhances cancer cell outgrowth. Proteomic analysis identified cell migration-inducing and hyaluronan-binding protein (CEMIP) as elevated in exosomes from brain metastatic but not lung or bone metastatic cells. CEMIP depletion in tumour cells impaired brain metastasis, disrupting invasion and tumour cell association with the brain vasculature, phenotypes rescued by pre-conditioning the brain microenvironment with CEMIP+ exosomes. Moreover, uptake of CEMIP+ exosomes by brain endothelial and microglial cells induced endothelial cell branching and inflammation in the perivascular niche by upregulating the pro-inflammatory cytokines encoded by Ptgs2, Tnf and Ccl/Cxcl, known to promote brain vascular remodelling and metastasis. CEMIP was elevated in tumour tissues and exosomes from patients with brain metastasis and predicted brain metastasis progression and patient survival. Collectively, our findings suggest that targeting exosomal CEMIP could constitute a future avenue for the prevention and treatment of brain metastasis.


Subject(s)
Brain Neoplasms/genetics , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Hyaluronoglucosaminidase/genetics , Neovascularization, Pathologic/genetics , Tumor Microenvironment/genetics , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CCL1/genetics , Chemokine CCL1/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Exosomes/pathology , Humans , Hyaluronoglucosaminidase/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/pathology , Signal Transduction , Survival Analysis , Tumor Burden , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
8.
Nat Rev Cancer ; 17(5): 302-317, 2017 05.
Article in English | MEDLINE | ID: mdl-28303905

ABSTRACT

It is well established that organs of future metastasis are not passive receivers of circulating tumour cells, but are instead selectively and actively modified by the primary tumour before metastatic spread has even occurred. Sowing the 'seeds' of metastasis requires the action of tumour-secreted factors and tumour-shed extracellular vesicles that enable the 'soil' at distant metastatic sites to encourage the outgrowth of incoming cancer cells. In this Review, we summarize the main processes and new mechanisms involved in the formation of the pre-metastatic niche.


Subject(s)
Neoplasm Metastasis/pathology , Neoplasms/pathology , Animals , Humans , Neoplasm Metastasis/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Tumor Microenvironment
9.
Sci Transl Med ; 6(236): 236ra62, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24828076

ABSTRACT

Intensified and central nervous system (CNS)-directed chemotherapy has improved outcomes for pediatric B cell acute lymphoblastic leukemia (B-ALL) but confers treatment-related morbidities. Moreover, many patients suffer relapses, underscoring the need to develop new molecular targeted B-ALL therapies. Using a mouse model, we show that leukemic B cells require pre-B cell receptor (pre-BCR)-independent spleen tyrosine kinase (SYK) signaling in vivo for survival and proliferation. In diagnostic samples from human pediatric and adult B-ALL patients, SYK and downstream targets were phosphorylated regardless of pre-BCR expression or genetic subtype. Two small-molecule SYK inhibitors, fostamatinib and BAY61-3606, attenuated the growth of 69 B-ALL samples in vitro, including high-risk (HR) subtypes. Orally administered fostamatinib reduced heavy disease burden after xenotransplantation of HR B-ALL samples into immunodeficient mice and decreased leukemia dissemination into spleen, liver, kidneys, and the CNS of recipient mice. Thus, SYK activation sustains the growth of multiple HR B-ALL subtypes, suggesting that SYK inhibitors may improve outcomes for HR and relapsed B-ALL.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cells, B-Lymphoid/cytology , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/metabolism , Spleen/enzymology , Administration, Oral , Adult , Aminopyridines , Animals , Cell Proliferation , Cell Survival , Child , Female , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Leukemia/drug therapy , Male , Mice , Mice, SCID , Morpholines , Mutation , Neoplasm Transplantation , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Oligonucleotide Array Sequence Analysis , Oxazines/chemistry , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Recurrence , Signal Transduction , Syk Kinase , Treatment Outcome
10.
Nat Cell Biol ; 15(7): 807-17, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23728425

ABSTRACT

In a significant fraction of breast cancer patients, distant metastases emerge after years or even decades of latency. How disseminated tumour cells (DTCs) are kept dormant, and what wakes them up, are fundamental problems in tumour biology. To address these questions, we used metastasis assays in mice and showed that dormant DTCs reside on microvasculature of lung, bone marrow and brain. We then engineered organotypic microvascular niches to determine whether endothelial cells directly influence breast cancer cell (BCC) growth. These models demonstrated that endothelial-derived thrombospondin-1 induces sustained BCC quiescence. This suppressive cue was lost in sprouting neovasculature; time-lapse analysis showed that sprouting vessels not only permit, but accelerate BCC outgrowth. We confirmed this surprising result in dormancy models and in zebrafish, and identified active TGF-ß1 and periostin as tumour-promoting factors derived from endothelial tip cells. Our work reveals that stable microvasculature constitutes a dormant niche, whereas sprouting neovasculature sparks micrometastatic outgrowth.


Subject(s)
Bone Marrow Neoplasms/secondary , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Endothelium, Vascular/pathology , Lung Neoplasms/secondary , Neoplasm, Residual/pathology , Neovascularization, Pathologic , Pericytes/pathology , Animals , Bone Marrow Neoplasms/blood supply , Bone Marrow Neoplasms/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/metabolism , Female , Fluorescent Antibody Technique , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Mice , Neoplasm, Residual/blood supply , Neoplasm, Residual/metabolism , Pericytes/metabolism , Stem Cell Niche/physiology , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , Zebrafish/growth & development , Zebrafish/metabolism
11.
Blood ; 109(5): 1887-96, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17077325

ABSTRACT

Mutations in ATM (ataxia-telangiectasia mutated) cause ataxia-telangiectasia (AT), a disease characterized by neurodegeneration, sterility, immunodeficiency, and T-cell leukemia. Defective ATM-mediated DNA damage responses underlie many aspects of the AT syndrome, but the basis for the immune deficiency has not been defined. ATM associates with DNA double-strand breaks (DSBs), and some evidence suggests that ATM may regulate V(D)J recombination. However, it remains unclear how ATM loss compromises lymphocyte development in vivo. Here, we show that T-cell receptor beta (TCRbeta)-dependent proliferation and production of TCRbeta(low) CD4+CD8+ (DP) thymocytes occurred normally in Atm-/- mice. In striking contrast, the postmitotic maturation of TCRbeta(low) DP precursors into TCRbeta(int) DP cells and TCRbeta(hi) mature thymocytes was profoundly impaired. Furthermore, Atm-/- thymocytes expressed abnormally low amounts of TCRalpha mRNA and protein. These defects were not attributable to the induction of a BCL-2-sensitive apoptotic pathway. Rather, they were associated with frequent biallelic loss of distal Va gene segments in DP thymocytes, revealing that ATM maintains Tcra locus integrity as it undergoes V(D)J recombination. Collectively, our data demonstrate that ATM loss increases the frequency of aberrant Tcra deletion events, which compromise DP thymocyte maturation and likely promote the generation of oncogenic TCR translocations.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , DNA-Binding Proteins/deficiency , Protein Serine-Threonine Kinases/deficiency , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/cytology , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Deletion , Mice , Mitosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Thymus Gland/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
12.
Immunol Rev ; 209: 142-58, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16448540

ABSTRACT

The immune system is capable of recognizing and eliminating an enormous array of pathogens due to the extremely diverse antigen receptor repertoire of T and B lymphocytes. However, the development of lymphocytes bearing receptors with unique specificities requires the generation of programmed double strand breaks (DSBs) coupled with bursts of proliferation, rendering lymphocytes susceptible to mutations contributing to oncogenic transformation. Consequently, mechanisms responsible for monitoring global genomic integrity must be activated during lymphocyte development to limit the oncogenic potential of antigen receptor locus recombination. Mutations in ATM (ataxia-telangiectasia mutated), a kinase that coordinates DSB monitoring and the response to DNA damage, result in impaired T-cell development and predispose to T-cell leukemia. Here, we review recent evidence providing insight into the mechanisms by which ATM promotes normal lymphocyte development and protects from neoplastic transformation.


Subject(s)
Cell Cycle Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , Lymphopoiesis/physiology , Protein Serine-Threonine Kinases/physiology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/immunology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Rearrangement, T-Lymphocyte , Genomic Instability , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Leukemia, T-Cell/enzymology , Lymphoma, T-Cell/enzymology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/pathology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , VDJ Recombinases
SELECTION OF CITATIONS
SEARCH DETAIL