Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(4): 113989, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38536816

ABSTRACT

Attachment of circulating tumor cells to the endothelial cells (ECs) lining blood vessels is a critical step in cancer metastatic colonization, which leads to metastatic outgrowth. Breast and prostate cancers are common malignancies in women and men, respectively. Here, we observe that ß1-integrin is required for human prostate and breast cancer cell adhesion to ECs under shear-stress conditions in vitro and to lung blood vessel ECs in vivo. We identify IQGAP1 and neural Wiskott-Aldrich syndrome protein (NWASP) as regulators of ß1-integrin transcription and protein expression in prostate and breast cancer cells. IQGAP1 and NWASP depletion in cancer cells decreases adhesion to ECs in vitro and retention in the lung vasculature and metastatic lung nodule formation in vivo. Mechanistically, NWASP and IQGAP1 act downstream of Cdc42 to increase ß1-integrin expression both via extracellular signal-regulated kinase (ERK)/focal adhesion kinase signaling at the protein level and by myocardin-related transcription factor/serum response factor (SRF) transcriptionally. Our results identify IQGAP1 and NWASP as potential therapeutic targets to reduce early metastatic dissemination.


Subject(s)
Integrin beta1 , Neoplasm Metastasis , Serum Response Factor , ras GTPase-Activating Proteins , Humans , Integrin beta1/metabolism , Integrin beta1/genetics , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Cell Line, Tumor , Serum Response Factor/metabolism , Male , Female , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Animals , Trans-Activators/metabolism , Cell Adhesion , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Mice , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , cdc42 GTP-Binding Protein/metabolism
2.
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
3.
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
4.
Extracell Vesicles Circ Nucl Acids ; 4(3): 461-485, 2023 Sep.
Article in English | MEDLINE | ID: mdl-38707985

ABSTRACT

Over the past decade, extracellular vesicles and particles (EVPs) have emerged as critical mediators of intercellular communication, participating in numerous physiological and pathological processes. In the context of cancer, EVPs exert local effects, such as increased invasiveness, motility, and reprogramming of tumor stroma, as well as systemic effects, including pre-metastatic niche formation, determining organotropism, promoting metastasis and altering the homeostasis of various organs and systems, such as the liver, muscle, and circulatory system. This review provides an overview of the critical advances in EVP research during the past decade, highlighting the heterogeneity of EVPs, their roles in intercellular communication, cancer progression, and metastasis. Moreover, the clinical potential of systemic EVPs as useful cancer biomarkers and therapeutic agents is explored. Last but not least, the progress in EVP analysis technologies that have facilitated these discoveries is discussed, which may further propel EVP research in the future.

5.
EMBO J ; 41(18): e109288, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36052513

ABSTRACT

Intercellular cross talk between cancer cells and stromal and immune cells is essential for tumor progression and metastasis. Extracellular vesicles and particles (EVPs) are a heterogeneous class of secreted messengers that carry bioactive molecules and that have been shown to be crucial for this cell-cell communication. Here, we highlight the multifaceted roles of EVPs in cancer. Functionally, transfer of EVP cargo between cells influences tumor cell growth and invasion, alters immune cell composition and function, and contributes to stromal cell activation. These EVP-mediated changes impact local tumor progression, foster cultivation of pre-metastatic niches at distant organ-specific sites, and mediate systemic effects of cancer. Furthermore, we discuss how exploiting the highly selective enrichment of molecules within EVPs has profound implications for advancing diagnostic and prognostic biomarker development and for improving therapy delivery in cancer patients. Altogether, these investigations into the role of EVPs in cancer have led to discoveries that hold great promise for improving cancer patient care and outcome.


Subject(s)
Extracellular Vesicles , Neoplasms , Cell Communication , Humans , Immunotherapy , Neoplasms/pathology , Tumor Microenvironment
6.
Gut Microbes ; 14(1): 2105609, 2022.
Article in English | MEDLINE | ID: mdl-35915556

ABSTRACT

The gut microbiome is intricately coupled with immune regulation and metabolism, but its role in Coronavirus Disease 2019 (COVID-19) is not fully understood. Severe and fatal COVID-19 is characterized by poor anti-viral immunity and hypercoagulation, particularly in males. Here, we define multiple pathways by which the gut microbiome protects mammalian hosts from SARS-CoV-2 intranasal infection, both locally and systemically, via production of short-chain fatty acids (SCFAs). SCFAs reduced viral burdens in the airways and intestines by downregulating the SARS-CoV-2 entry receptor, angiotensin-converting enzyme 2 (ACE2), and enhancing adaptive immunity via GPR41 and 43 in male animals. We further identify a novel role for the gut microbiome in regulating systemic coagulation response by limiting megakaryocyte proliferation and platelet turnover via the Sh2b3-Mpl axis. Taken together, our findings have unraveled novel functions of SCFAs and fiber-fermenting gut bacteria to dampen viral entry and hypercoagulation and promote adaptive antiviral immunity.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Animals , Antiviral Agents/therapeutic use , Fatty Acids, Volatile , Male , Mammals/metabolism , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2
7.
Clin Exp Metastasis ; 39(2): 263-277, 2022 04.
Article in English | MEDLINE | ID: mdl-35072851

ABSTRACT

While immense strides have been made in understanding tumor biology and in developing effective treatments that have substantially improved the prognosis of cancer patients, metastasis remains the major cause of cancer-related death. Improvements in the detection and treatment of primary tumors are contributing to a growing, detailed understanding of the dynamics of metastatic progression. Yet challenges remain in detecting metastatic dissemination prior to the establishment of overt metastases and in predicting which patients are at the highest risk of developing metastatic disease. Further improvements in understanding the mechanisms governing metastasis have great potential to inform the adaptation of existing therapies and the development of novel approaches to more effectively control metastatic disease. This article presents a forward-looking perspective on the challenges that remain in the treatment of metastasis, and the exciting emerging approaches that promise to transform the treatment of metastasis in cancer patients.


Subject(s)
Neoplasms , Humans , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , Prognosis
8.
Mol Oncol ; 16(1): 130-147, 2022 01.
Article in English | MEDLINE | ID: mdl-34058066

ABSTRACT

Dormant, disseminated tumor cells (DTCs) are thought to be the source of breast cancer metastases several years or even decades after initial treatment. To date, a selective therapy that leads to their elimination has not been discovered. While dormant DTCs resist chemotherapy, evidence suggests that this resistance is driven not by their lack of proliferation, but by their engagement of the surrounding microenvironment, via integrin-ß1-mediated interactions. Because integrin-ß1-targeted agents have not been translated readily to the clinic, signaling nodes downstream of integrin-ß1 could serve as attractive therapeutic targets in order to sensitize dormant DTCs to therapy. By probing a number of kinases downstream of integrin-ß1, we determined that PI3K inhibition with either a tool compounds or a compound (PF-05212384; aka Gedatolisib) in clinical trials robustly sensitizes quiescent breast tumor cells seeded in organotypic bone marrow cultures to chemotherapy. These results motivated the preclinical study of whether Gedatolisib-with or without genotoxic therapy-would reduce DTC burden and prevent metastases. Despite promising results in organotypic culture, Gedatolisib failed to reduce DTC burden or delay, reduce or prevent metastasis in murine models of either triple-negative or estrogen receptor-positive breast cancer dissemination and metastasis. This result held true whether analyzing Gedatolisib on its own (vs. vehicle-treated animals) or in combination with dose-dense doxorubicin and cyclophosphamide (vs. animals treated only with dose-dense chemotherapies). These data suggest that PI3K is not the node downstream of integrin-ß1 that confers chemotherapeutic resistance to DTCs. More broadly, they cast doubt on the strategy to target PI3K in order to eliminate DTCs and prevent breast cancer metastasis.


Subject(s)
Breast Neoplasms , Phosphatidylinositol 3-Kinases , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Humans , Integrins , Mice , Morpholines , Phosphoinositide-3 Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases , Triazines , Tumor Microenvironment
9.
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
10.
iScience ; 23(12): 101844, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33376973

ABSTRACT

Liquid biopsies based on cell-free DNA (cfDNA) or exosomes provide a noninvasive approach to monitor human health and disease but have not been utilized for astronauts. Here, we profile cfDNA characteristics, including fragment size, cellular deconvolution, and nucleosome positioning, in an astronaut during a year-long mission on the International Space Station, compared to his identical twin on Earth and healthy donors. We observed a significant increase in the proportion of cell-free mitochondrial DNA (cf-mtDNA) inflight, and analysis of post-flight exosomes in plasma revealed a 30-fold increase in circulating exosomes and patient-specific protein cargo (including brain-derived peptides) after the year-long mission. This longitudinal analysis of astronaut cfDNA during spaceflight and the exosome profiles highlights their utility for astronaut health monitoring, as well as cf-mtDNA levels as a potential biomarker for physiological stress or immune system responses related to microgravity, radiation exposure, and the other unique environmental conditions of spaceflight.

11.
J Mammary Gland Biol Neoplasia ; 25(4): 305-317, 2020 12.
Article in English | MEDLINE | ID: mdl-33351162

ABSTRACT

Breast cancer (BC) is responsible for 15% of all the cancer deaths among women in the USA. The tumor microenvironment (TME) has the potential to act as a driver of breast cancer progression and metastasis. The TME is composed of stromal cells within an extracellular matrix and soluble cytokines, chemokines and extracellular vesicles and nanoparticles that actively influence cell behavior. Extracellular vesicles include exosomes, microvesicles and large oncosomes that orchestrate fundamental processes during tumor progression through direct interaction with target cells. Long before tumor cell spread to future metastatic sites, tumor-secreted exosomes enter the circulation and establish distant pre-metastatic niches, hospitable and permissive milieus for metastatic colonization. Emerging evidence suggests that breast cancer exosomes promote tumor progression and metastasis by inducing vascular leakiness, angiogenesis, invasion, immunomodulation and chemoresistance. Exosomes are found in almost all physiological fluids including plasma, urine, saliva, and breast milk, providing a valuable resource for the development of non-invasive cancer biomarkers. Here, we review work on the role of exosomes in breast cancer progression and metastasis, and describe the most recent advances in models of exosome secretion, isolation, characterization and functional analysis. We highlight the potential applications of plasma-derived exosomes as predictive biomarkers for breast cancer diagnosis, prognosis and therapy monitoring. We finally describe the therapeutic approaches of exosomes in breast cancer.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/diagnosis , Exosomes/pathology , Neovascularization, Pathologic/pathology , Animals , Breast/blood supply , Breast/cytology , Breast/pathology , Breast Neoplasms/blood , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm , Exosomes/metabolism , Exosomes/transplantation , Extracellular Matrix/pathology , Female , Humans , Liquid Biopsy/methods , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/diagnosis , Prognosis , Tumor Microenvironment , Xenograft Model Antitumor Assays
12.
Front Oncol ; 10: 1350, 2020.
Article in English | MEDLINE | ID: mdl-33042789

ABSTRACT

During the process of hematogenous metastasis, tumor cells interact with platelets and their precursors megakaryocytes, providing a selection driver for the metastatic phenotype. Cancer cells have evolved a plethora of mechanisms to engage platelet activation and aggregation. Platelet coating of tumor cells in the blood stream promotes the successful completion of multiple steps of the metastatic cascade. Along the same lines, clinical evidence suggests that anti-coagulant therapy might be associated with reduced risk of metastatic disease and better prognosis in cancer patients. Here, we review experimental and clinical literature concerning the contribution of platelets and megakaryocytes to cancer metastasis and provide insights into the clinical relevance of anti-coagulant therapy in cancer treatment.

13.
Cell ; 182(4): 1044-1061.e18, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32795414

ABSTRACT

There is an unmet clinical need for improved tissue and liquid biopsy tools for cancer detection. We investigated the proteomic profile of extracellular vesicles and particles (EVPs) in 426 human samples from tissue explants (TEs), plasma, and other bodily fluids. Among traditional exosome markers, CD9, HSPA8, ALIX, and HSP90AB1 represent pan-EVP markers, while ACTB, MSN, and RAP1B are novel pan-EVP markers. To confirm that EVPs are ideal diagnostic tools, we analyzed proteomes of TE- (n = 151) and plasma-derived (n = 120) EVPs. Comparison of TE EVPs identified proteins (e.g., VCAN, TNC, and THBS2) that distinguish tumors from normal tissues with 90% sensitivity/94% specificity. Machine-learning classification of plasma-derived EVP cargo, including immunoglobulins, revealed 95% sensitivity/90% specificity in detecting cancer. Finally, we defined a panel of tumor-type-specific EVP proteins in TEs and plasma, which can classify tumors of unknown primary origin. Thus, EVP proteins can serve as reliable biomarkers for cancer detection and determining cancer type.


Subject(s)
Biomarkers, Tumor/metabolism , Extracellular Vesicles/metabolism , Neoplasms/diagnosis , Animals , Biomarkers, Tumor/blood , Cell Line , HSC70 Heat-Shock Proteins/metabolism , Humans , Machine Learning , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Neoplasms/metabolism , Proteome/analysis , Proteome/metabolism , Proteomics/methods , Sensitivity and Specificity , Tetraspanin 29/metabolism , rap GTP-Binding Proteins/metabolism
14.
Cancers (Basel) ; 11(5)2019 May 06.
Article in English | MEDLINE | ID: mdl-31064120

ABSTRACT

Hepatic metastatic growth is dependent upon stromal factors including the matrisomal proteins that make up the extracellular matrix (ECM). Laminins are ECM glycoproteins with several functions relevant to tumour progression including angiogenesis. We investigated whether metastatic colon cancer cells produce the laminins required for vascular basement membrane assembly as a mechanism for the promotion of angiogenesis and liver metastasis growth. qPCR was performed using human-specific primers to laminin chains on RNA from orthotopic human colorectal liver metastases. Laminin α5 (LAMA5) expression was inhibited in colon cancer cells using shRNA. Notch pathway gene expression was determined in endothelia from hepatic metastases. Orthotopic hepatic metastases expressed human laminin chains α5, ß1 and γ1 (laminin 511), all of which are required for vascular basement membrane assembly. The expression of Laminin 511 was associated with reduced survival in several independent colorectal cancer cohorts and angiogenesis signatures or vessel density significantly correlated with LAMA5 expression. Colorectal cancer cells in culture made little LAMA5, but its levels were increased by culture in a medium conditioned by tumour-derived CD11b+ myeloid cells through TNFα/NFκB pathway signalling. Down-regulation of LAMA5 in cancer cells impaired liver metastatic growth and resulted in reduced intra-tumoural vessel branching and increased the expression of Notch pathway genes in metastasis-derived endothelia. This data demonstrates a mechanism whereby tumour inflammation induces LAMA5 expression in colorectal cancer cells. LAMA5 is required for the successful growth of hepatic metastases where it promotes branching angiogenesis and modulates Notch signalling.

15.
J Clin Invest ; 129(5): 1845-1862, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30907747

ABSTRACT

Because metastasis is associated with the majority of cancer-related deaths, its prevention is a clinical aspiration. Prostanoids are a large family of bioactive lipids derived from the activity of cyclooxygenase-1 (COX-1) and COX-2. Aspirin impairs the biosynthesis of all prostanoids through the irreversible inhibition of both COX isoforms. Long-term administration of aspirin leads to reduced distant metastases in murine models and clinical trials, but the COX isoform, downstream prostanoid, and cell compartment responsible for this effect are yet to be determined. Here, we have shown that aspirin dramatically reduced lung metastasis through inhibition of COX-1 while the cancer cells remained intravascular and that inhibition of platelet COX-1 alone was sufficient to impair metastasis. Thromboxane A2 (TXA2) was the prostanoid product of COX-1 responsible for this antimetastatic effect. Inhibition of the COX-1/TXA2 pathway in platelets decreased aggregation of platelets on tumor cells, endothelial activation, tumor cell adhesion to the endothelium, and recruitment of metastasis-promoting monocytes/macrophages, and diminished the formation of a premetastatic niche. Thus, platelet-derived TXA2 orchestrates the generation of a favorable intravascular metastatic niche that promotes tumor cell seeding and identifies COX-1/TXA2 signaling as a target for the prevention of metastasis.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Cyclooxygenase Inhibitors/pharmacology , Neoplasm Metastasis/drug therapy , Thromboxane A2/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Blood Platelets/metabolism , Cell Line, Tumor , Female , Humans , Lung Neoplasms , Macrophages/metabolism , Melanoma, Experimental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/metabolism , Neoplasm Transplantation , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Prostaglandins/metabolism , Protein Isoforms , Thrombosis
16.
Mol Cancer ; 12(1): 52, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23734815

ABSTRACT

BACKGROUND: Circulating microRNAs (miRNAs) have been found in many body fluids and represent reliable markers of several physio-pathological disorders, including cancer. In some cases, circulating miRNAs have been evaluated as markers of the efficacy of anticancer treatment but it is not yet clear if miRNAs are actively released by tumor cells or derive from dead tumor cells. RESULTS: We showed that a set of prostate cancer secretory miRNAs (PCS-miRNAs) were spontaneously released in the growth medium by DU-145 prostate cancer cells and that the release was greater after treatment with the cytotoxic drug fludarabine. We also found that the miRNAs were associated with exosomes, implying an active mechanism of miRNA release. It should be noted that in fludarabine treated cells the release of miR-485-3p, as well as its association with exosomes, was reduced suggesting that miR-485-3p was retained by surviving cells. Monitoring the intracellular level of miR-485-3p in these cells, we found that miR-485-3p was stably up regulated for several days after treatment. As a possible mechanism we suggest that fludarabine selected cells that harbor high levels of miR-485-3p, which in turn regulates the transcriptional repressor nuclear factor-Y triggering the transcription of topoisomerase IIα, multidrug resistance gene 1 and cyclin B2 pro-survival genes. CONCLUSIONS: Cytotoxic treatment of DU-145 cells enhanced the release of PCS-miRNAs with the exception of miR-485-3p which was retained by surviving cells. We speculate that the retention of miR-485-3p was a side effect of fludarabine treatment in that the high intracellular level of miR-485-3p plays a role in the sensitivity to fludarabine.


Subject(s)
Antineoplastic Agents/pharmacology , MicroRNAs/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Vidarabine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Humans , Male , Prostatic Neoplasms/metabolism , Vidarabine/pharmacology
17.
Br J Haematol ; 162(4): 433-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23691951

ABSTRACT

Inhibition of coagulation greatly limits cancer metastasis in many experimental models. Cancer cells trigger coagulation, through expression of tissue factor or P-selectin ligands that have correlated with worse prognosis in human clinical studies. Cancer cells also affect coagulation through expression of thrombin and release of microparticles that augment coagulation. In the cancer-bearing host, coagulation facilitates tumour progression through release of platelet granule contents, inhibition of Natural Killer cells and recruitment of macrophages. We are revisiting this literature in the light of recent studies in which treatment of clinical cohorts with anticoagulant drugs led to diminished metastasis.


Subject(s)
Anticoagulants/therapeutic use , Blood Coagulation , Blood Platelets/physiology , Neoplasm Metastasis/prevention & control , Neoplasms, Experimental/blood , Animals , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Blood Platelets/drug effects , Cysteine Endopeptidases/physiology , Cytoplasmic Granules/metabolism , Hirudins/pharmacology , Humans , Killer Cells, Natural/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Macrophages/immunology , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/physiology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neoplastic Cells, Circulating , Neuraminidase/pharmacology , Neuraminidase/therapeutic use , P-Selectin/physiology , Platelet Activation/physiology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Rats , Thrombin/metabolism , Thrombophilia/complications , Thrombophilia/drug therapy , Thromboplastin/physiology
18.
Oligonucleotides ; 21(1): 39-45, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21241187

ABSTRACT

Recently, it has been reported that, in several tumor cell lines, short double-stranded RNAs tailored for promoter regions of specific genes are able to activate their transcription. Such molecules (named RNA activators) act opposite to other double-stranded RNA molecules (named RNA inhibitors) in that the overexpression instead of underexpression of a given gene is triggered. In Dohh2 non-Hodgkin lymphoma cells, the transcriptional repressor BCL6, which negatively controls both p53 and p21, is overexpressed, so that the cells can escape the check point governed by p53 and proliferate. The aim of this work was to investigate whether the RNA activator p21 can represent a tool to circumvent the transcriptional control of BCL6 and induce the blockage of cell proliferation in Dohh2 non-Hodgkin lymphoma cells. For that, Dohh2 cells were transfected with either a control RNA activator (ds-NC) or an RNA activator specific for human p21 promoter (ds-p21). At various time points after transfection, the cells were collected and p21 was measured. Dohh2 cells transfected with ds-p21 showed a slight but significant overexpression of p21 at both mRNA and protein levels. Nonetheless, cell proliferation, cell cycle, and apoptosis were not significantly modified. In contrast, the exposure of Dohh2 cells transfected with ds-p21 to fludarabine potentiates the cytotoxicity of the drug, suggesting the RNA activator p21 complements the fludarabine-dependent cell death pathways.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins/metabolism , RNA, Double-Stranded , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , Drug Delivery Systems , Drug Synergism , Female , Gene Expression Regulation , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-6 , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Messenger/analysis , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/genetics , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Vidarabine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...