Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
iScience ; 26(6): 106943, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37332612

ABSTRACT

The bone marrow (BM) is a complex microenvironment, coordinating the production of billions of blood cells every day. Despite its essential role and its relevance to hematopoietic diseases, this environment remains poorly characterized. Here we present a high-resolution characterization of the niche in health and acute myeloid leukemia (AML) by establishing a single-cell gene expression database of 339,381 BM cells. We found significant changes in cell type proportions and gene expression in AML, indicating that the entire niche is disrupted. We then predicted interactions between hematopoietic stem and progenitor cells (HSPCs) and other BM cell types, revealing a remarkable expansion of predicted interactions in AML that promote HSPC-cell adhesion, immunosuppression, and cytokine signaling. In particular, predicted interactions involving transforming growth factor ß1 (TGFB1) become widespread, and we show that this can drive AML cell quiescence in vitro. Our results highlight potential mechanisms of enhanced AML-HSPC competitiveness and a skewed microenvironment, fostering AML growth.

3.
Nat Commun ; 13(1): 6320, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36329021

ABSTRACT

The plasma multimeric glycoprotein von Willebrand factor (VWF) plays a critical role in primary hemostasis by tethering platelets to exposed collagen at sites of vascular injury. Recent studies have identified additional biological roles for VWF, and in particular suggest that VWF may play an important role in regulating inflammatory responses. However, the molecular mechanisms through which VWF exerts its immuno-modulatory effects remain poorly understood. In this study, we report that VWF binding to macrophages triggers downstream MAP kinase signaling, NF-κB activation and production of pro-inflammatory cytokines and chemokines. In addition, VWF binding also drives macrophage M1 polarization and shifts macrophage metabolism towards glycolysis in a p38-dependent manner. Cumulatively, our findings define an important biological role for VWF in modulating macrophage function, and thereby establish a novel link between primary hemostasis and innate immunity.


Subject(s)
Hemostasis , von Willebrand Factor , von Willebrand Factor/metabolism , Hemostasis/physiology , Blood Platelets/metabolism , Immunity, Innate , Macrophages/metabolism
4.
J Thromb Haemost ; 20(10): 2350-2365, 2022 10.
Article in English | MEDLINE | ID: mdl-35722954

ABSTRACT

BACKGROUND: Breast cancer results in a three- to four-fold increased risk of venous thromboembolism (VTE), which is associated with reduced patient survival. Despite this, the mechanisms underpinning breast cancer-associated thrombosis remain poorly defined. Tumor cells can trigger endothelial cell (EC) activation resulting in increased von Willebrand factor (VWF) secretion. Importantly, elevated plasma VWF levels constitute an independent biomarker for VTE risk. Moreover, in a model of melanoma, treatment with low molecular weight heparin (LMWH) negatively regulated VWF secretion and attenuated tumor metastasis. OBJECTIVE: To investigate the role of VWF in breast cancer metastasis and examine the effect of LMWH in modulating EC activation and breast tumor transmigration. METHODS: von Willebrand factor levels were measured by ELISA. Primary ECs were used to assess tumor-induced activation, angiogenesis, tumor adhesion, and transendothelial migration. RESULTS AND CONCLUSION: Patients with metastatic breast cancer have markedly elevated plasma VWF:Ag levels that also correlate with poorer survival. MDA-MB-231 and MCF-7 breast cancer cells induce secretion of VWF, angiopoietin-2, and osteoprotegerin from ECs, which is further enhanced by the presence of platelets. Vascular endothelial growth factor-A (VEGF-A) plays an important role in modulating breast cancer-induced VWF release. Moreover, VEGF-A from breast tumor cells also contributes to a pro-angiogenic effect on ECs. VWF multimers secreted from ECs, in response to tumor-VEGF-A, mediate adhesion of breast tumor cells along the endothelium. LMWH inhibits VWF-breast tumor adhesion and transendothelial migration. Our findings highlight the significant crosstalk between tumor cells and the endothelium including increased VWF secretion which may contribute to tumor metastasis.


Subject(s)
Breast Neoplasms , Venous Thromboembolism , Angiopoietin-2/metabolism , Breast Neoplasms/metabolism , Endothelial Cells/metabolism , Female , Heparin, Low-Molecular-Weight/pharmacology , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Osteoprotegerin/metabolism , Transendothelial and Transepithelial Migration , Vascular Endothelial Growth Factor A/metabolism , Venous Thromboembolism/metabolism , von Willebrand Factor/metabolism
5.
Semin Thromb Hemost ; 47(2): 139-149, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33636745

ABSTRACT

The association between cancer and venous thromboembolism (VTE) has been established for more than 150 years. Nevertheless, cancer-associated thrombosis still remains a major clinical challenge and is associated with significant morbidity and mortality for patients with cancer. The clinical presentation of cancer-associated thrombosis can be distinct from that of a patient without an underlying malignancy. Moreover, specific cancer types, including pancreatic cancer and hematological malignancies, as well as advanced stage disease can confer a significant thrombotic risk. This risk is further augmented by specific anticancer treatment modalities. The pathophysiology of cancer-associated thrombosis is complex and multifactorial. However, understanding the biological mechanisms underpinning VTE risk may provide insight into novel targeted prophylaxis in cancer patients. Over the last decade, low-molecular-weight heparin has been the preferred anticoagulant agent for patients with cancer-associated thrombosis due to improved efficacy compared with vitamin K antagonists. However, the advent of direct oral anticoagulants (DOACs) has added to the repertoire of ammunition now at the disposal of clinicians to aid in the management of cancer-associated thrombosis. Several randomized controlled trials have now been published, demonstrating DOAC as a noninferior alternative for both the treatment and prevention of cancer-associated thrombosis. Notwithstanding this, limitations for their widespread use remain, with the potential for increased bleeding risk, drug interactions, and poor DOAC metabolism. This review discusses the evidence base for the incidence and risk factors associated with VTE in cancer, development, and refinement of risk prediction models and novel advances in the therapeutic management of cancer-associated thrombosis.


Subject(s)
Anticoagulants/therapeutic use , Neoplasms/complications , Thrombosis/drug therapy , Anticoagulants/pharmacology , Humans , Risk Factors , Thrombosis/etiology
6.
J Thromb Haemost ; 18(10): 2444-2456, 2020 10.
Article in English | MEDLINE | ID: mdl-32573945

ABSTRACT

Von Willebrand factor (VWF) is a multimeric procoagulant plasma glycoprotein that mediates platelet adhesion along the endothelium. In addition to its role maintaining normal hemostasis, more recently novel biological functions for VWF have been described, including inflammation, angiogenesis, and metastasis. Significantly increased plasma VWF levels have been reported across a variety of cancer patient cohorts. Given that VWF is established as a risk factor for venous thrombosis, this is of direct clinical importance. Moreover, elevated VWF has also been observed localized within the tumor microenvironment, correlating with advanced disease stage and poorer clinical outcome. Critically, evidence suggests that elevated VWF levels in cancer patients may not only contribute to cancer associated coagulopathies but may also mediate cancer progression and metastasis. Studies have shown that VWF can promote pro-inflammatory signaling, regulate angiogenesis and vascular permeability, which may facilitate tumor cell growth and extravasation across the vessel wall. Endothelial secreted VWF multimers contribute to the adhesion and transendothelial migration of tumor cells key for tumor dissemination. In support of this, VWF inhibition attenuated metastasis in vivo. Perhaps most intriguingly, specific tumor cells have been reported to acquire de novo VWF expression which increases tumor-platelet heteroaggregates and confers enhanced metastatic activity. Current knowledge on the roles of VWF in cancer and in particular its contribution to metastasis and cancer associated coagulopathies is summarized in this review.


Subject(s)
Neoplasms , von Willebrand Diseases , Blood Platelets , Humans , Platelet Adhesiveness , Tumor Microenvironment , von Willebrand Factor
8.
Sci Rep ; 8(1): 15752, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361682

ABSTRACT

Acute myeloid leukaemia (AML) is an aggressive cancer with 50-75% of patients relapsing even after successful chemotherapy. The role of the bone marrow microenvironment (BMM) in protecting AML cells from chemotherapeutics and causing consequent relapse is increasingly recognised. However the role that the anti-apoptotic Bcl-2 proteins play as effectors of BMM-mediated drug resistance are less understood. Here we show that bone marrow mesenchymal stromal cells (BMSC) provide resistance to AML cells against BH3-mimetics, cytarabine and daunorubicin, but this is not mediated by Bcl-2 and/or Bcl-XL as previously thought. Instead, BMSCs induced Mcl-1 expression over Bcl-2 and/or Bcl-XL in AML cells and inhibition of Mcl-1 with a small-molecule inhibitor, A1210477, or repressing its expression with the CDC7/CDK9 dual-inhibitor, PHA-767491 restored sensitivity to BH3-mimetics. Furthermore, combined inhibition of Bcl-2/Bcl-XL and Mcl-1 could revert BMSC-mediated resistance against cytarabine + daunorubicin. Importantly, the CD34+/CD38- leukemic stem cell-encompassing population was equally sensitive to the combination of PHA-767491 and ABT-737. These results indicate that Bcl-2/Bcl-XL and Mcl-1 act in a redundant fashion as effectors of BMM-mediated AML drug resistance and highlight the potential of Mcl-1-repression to revert BMM-mediated drug resistance in the leukemic stem cell population, thus, prevent disease relapse and ultimately improve patient survival.


Subject(s)
Bone Marrow/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Piperidones/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrroles/pharmacology , Antigens, CD/metabolism , Biphenyl Compounds/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Cytarabine/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Nitrophenols/pharmacology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Sulfonamides/pharmacology , Tumor Microenvironment/drug effects , bcl-X Protein/metabolism
9.
Drug Discov Today ; 21(9): 1464-1471, 2016 09.
Article in English | MEDLINE | ID: mdl-27130156

ABSTRACT

Acute myeloid leukaemia (AML) is a hierarchically structured malignancy in which aberrant leukemic stem cells drive the production of leukaemic blast cell clones. AML cells strictly depend on the bone marrow microenvironment (BMM) in which they reside. Classical AML cell cultures fail to mimic the BMM and, therefore, drug discovery studies are dominated by in vivo models. However, animal models are time consuming, labour intensive, provide limited mechanistic insight, and are unsuited for high-throughput studies, necessitating the development of novel AML models. The evolving ex vivo BMM-mimicking culture systems aim to fill this gap, with increasing success. Here, we discuss how AML-microenvironment co-culture models advance our understanding of this disease, and highlight their future potential for translational AML research.


Subject(s)
Drug Evaluation, Preclinical/methods , Leukemia, Myeloid, Acute/drug therapy , Animals , Bone Marrow , Coculture Techniques , Humans , Tissue Scaffolds
10.
Mol Biosyst ; 7(2): 403-10, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21042606

ABSTRACT

Aging is a multi-factorial and complex phenomenon. Saccharomyces cerevisiae is developed as a model of aging and has been widely studied in order to understand the mechanism of lifespan regulation. A large number of high-throughput studies were conducted to identify the genes which modulate lifespan. These studies provide the list of genes that regulates the lifespan in yeast; however the regulation of these aging associated genes had not been fully understood. In this study, we have shown that deletion of the genes which increase the replicative lifespan (RLS) of yeast show discrete expression patterns when compared with the genes that, on deletion, cause a decrease in lifespan. Expression of longlived (LL) genes decreases as the cell progresses from mid log to stationary phase, whereas expression of shortlived (SL) genes remains unchanged. This distinct expression of LL and SL gene-sets suggests their differential gene regulation. Further analysis of transcriptional regulation by transcription factors and epigenetic regulators (acetylation and methylation) suggests that this differential expression of the two gene-sets is due to their differential epigenetic regulations, rather than regulation by transcription factors. These results accentuate the importance of epigenetic modifications in aging. We deduce that future focused studies on epigenetic modification regulation will help lead to a better understanding of the aging process.


Subject(s)
Aging/genetics , Gene Expression Regulation, Fungal , Genes, Fungal , Saccharomyces cerevisiae/genetics , Epigenesis, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...