Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Cell Sci ; 136(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37870164

ABSTRACT

Tumor initiation at either primary or metastatic sites is an inefficient process in which tumor cells must fulfill a series of conditions. One critical condition involves the ability of individual tumor-initiating cells to overcome 'isolation stress', enabling them to survive within harsh isolating microenvironments that can feature nutrient stress, hypoxia, oxidative stress and the absence of a proper extracellular matrix (ECM). In response to isolation stress, tumor cells can exploit various adaptive strategies to develop stress tolerance and gain stemness features. In this Opinion, we discuss how strategies such as the induction of certain cell surface receptors and deposition of ECM proteins enable tumor cells to endure isolation stress, thereby gaining tumor-initiating potential. As examples, we highlight recent findings from our group demonstrating how exposure of tumor cells to isolation stress upregulates the G-protein-coupled receptor lysophosphatidic acid receptor 4 (LPAR4), its downstream target fibronectin and two fibronectin-binding integrins, α5ß1 and αvß3. These responses create a fibronectin-rich niche for tumor cells, ultimately driving stress tolerance, cancer stemness and tumor initiation. We suggest that approaches to prevent cancer cells from adapting to stress by suppressing LPAR4 induction, blocking its downstream signaling or disrupting fibronectin-integrin interactions hold promise as potential strategies for cancer treatment.


Subject(s)
Fibronectins , Integrins , Fibronectins/metabolism , Cell Adhesion/physiology , Up-Regulation , Integrins/metabolism , Integrin alpha5beta1/metabolism , Extracellular Matrix/metabolism , Integrin alphaVbeta3/metabolism
3.
Proc Natl Acad Sci U S A ; 110(22): 9042-7, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671068

ABSTRACT

Lymph nodes are initial sites of tumor metastasis, yet whether the lymph node microenvironment actively promotes tumor metastasis remains unknown. We show here that VEGF-C/PI3Kα-driven remodeling of lymph nodes promotes tumor metastasis by activating integrin α4ß1 on lymph node lymphatic endothelium. Activated integrin α4ß1 promotes expansion of the lymphatic endothelium in lymph nodes and serves as an adhesive ligand that captures vascular cell adhesion molecule 1 (VCAM-1)(+) metastatic tumor cells, thereby promoting lymph node metastasis. Experimental induction of α4ß1 expression in lymph nodes is sufficient to promote tumor cell adhesion to lymphatic endothelium and lymph node metastasis in vivo, whereas genetic or pharmacological blockade of integrin α4ß1 or VCAM-1 inhibits it. As lymph node metastases accurately predict poor disease outcome, and integrin α4ß1 is a biomarker of lymphatic endothelium in tumor-draining lymph nodes from animals and patients, these results indicate that targeting integrin α4ß1 or VCAM to inhibit the interactions of tumor cells with the lymph node microenvironment may be an effective strategy to suppress tumor metastasis.


Subject(s)
Carcinoma, Ductal, Breast/pathology , Endothelium, Lymphatic/metabolism , Integrin alpha4beta1/metabolism , Lymph Nodes/metabolism , Neoplasm Metastasis/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Analysis of Variance , Animals , Cell Adhesion/physiology , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Lymphangiogenesis/physiology , Mice , Neoplasm Metastasis/prevention & control , Vascular Cell Adhesion Molecule-1/physiology
4.
Blood ; 122(25): 4140-9, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24108461

ABSTRACT

The Src family kinases (SFKs) c-Src and Yes mediate vascular leakage in response to various stimuli including lipopolysaccharide (LPS) and vascular endothelial growth factor (VEGF). Here, we define an opposing function of another SFK, Lyn, which in contrast to other SFKs, strengthens endothelial junctions and thereby restrains the increase in vascular permeability. Mice lacking Lyn displayed increased mortality in LPS-induced endotoxemia and increased vascular permeability in response to LPS or VEGF challenge compared with wild-type littermates. Lyn knockout mice repopulated with wild-type bone marrow-derived cells have higher vascular permeability than wild-type mice, suggesting a role of endothelial Lyn in the maintenance of the vascular barrier. Small interfering RNA-mediated down-regulation of Lyn disrupted endothelial barrier integrity, whereas expression of a constitutively active mutant of Lyn enhanced the barrier. However, down-regulation of Lyn did not affect LPS-induced endothelial permeability. We demonstrate that Lyn association with focal adhesion kinase (FAK) and phosphorylation of FAK at tyrosine residues 576/577 and 925 were required for Lyn-dependent stabilization of endothelial adherens junctions. Thus, in contrast to c-Src and Yes, which increase vascular permeability in response to stimuli, Lyn stabilizes endothelial junctions through phosphorylation of FAK. Therefore, therapeutics activating Lyn kinase may strengthen the endothelial barrier junction and hence have anti-inflammatory potential.


Subject(s)
Adherens Junctions/enzymology , Capillary Permeability/physiology , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , src-Family Kinases/metabolism , Adherens Junctions/genetics , Animals , Capillary Permeability/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/genetics , src-Family Kinases/genetics
5.
Blood ; 119(9): 2149-58, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22134168

ABSTRACT

Vascular development and angiogenesis initially depend on endothelial tip cell invasion, which is followed by a series of maturation steps, including lumen formation and recruitment of perivascular cells. Notch ligands expressed on the endothelium and their cognate receptors expressed on perivascular cells are involved in blood vessel maturation, though little is known regarding the Notch-dependent effectors that facilitate perivascular coverage of nascent vessels. Here, we report that vascular smooth muscle cell (VSMC) recognition of the Notch ligand Jagged1 on endothelial cells leads to expression of integrin αvß3 on VSMCs. Once expressed, integrin αvß3 facilitates VSMC adhesion to VWF in the endothelial basement membrane of developing retinal arteries, leading to vessel maturation. Genetic or pharmacologic disruption of Jagged1, Notch, αvß3, or VWF suppresses VSMC coverage of nascent vessels and arterial maturation during vascular development. Therefore, we define a Notch-mediated interaction between the developing endothelium and VSMCs leading to adhesion of VSMCs to the endothelial basement membrane and arterial maturation.


Subject(s)
Basement Membrane/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/metabolism , Integrins/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Notch/metabolism , Animals , Arteries/metabolism , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Integrins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/genetics , Protein Binding , RNA, Messenger/metabolism , Receptors, Notch/genetics , Serrate-Jagged Proteins , Signal Transduction/physiology , von Willebrand Factor/metabolism
6.
Nature ; 456(7223): 814-8, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-18997773

ABSTRACT

Angiogenesis and the development of a vascular network are required for tumour progression, and they involve the release of angiogenic factors, including vascular endothelial growth factor (VEGF-A), from both malignant and stromal cell types. Infiltration by cells of the myeloid lineage is a hallmark of many tumours, and in many cases the macrophages in these infiltrates express VEGF-A. Here we show that the deletion of inflammatory-cell-derived VEGF-A attenuates the formation of a typical high-density vessel network, thus blocking the angiogenic switch in solid tumours in mice. Vasculature in tumours lacking myeloid-cell-derived VEGF-A was less tortuous, with increased pericyte coverage and decreased vessel length, indicating vascular normalization. In addition, loss of myeloid-derived VEGF-A decreases the phosphorylation of VEGF receptor 2 (VEGFR2) in tumours, even though overall VEGF-A levels in the tumours are unaffected. However, deletion of myeloid-cell VEGF-A resulted in an accelerated tumour progression in multiple subcutaneous isograft models and an autochthonous transgenic model of mammary tumorigenesis, with less overall tumour cell death and decreased tumour hypoxia. Furthermore, loss of myeloid-cell VEGF-A increased the susceptibility of tumours to chemotherapeutic cytotoxicity. This shows that myeloid-derived VEGF-A is essential for the tumorigenic alteration of vasculature and signalling to VEGFR2, and that these changes act to retard, not promote, tumour progression.


Subject(s)
Carcinoma/metabolism , Gene Deletion , Mammary Neoplasms, Experimental/metabolism , Myeloid Cells/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Carcinoma/blood supply , Carcinoma/genetics , Cytotoxins/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia/genetics , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/pharmacology
7.
Nature ; 456(7223): 809-13, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-18997771

ABSTRACT

Angiogenesis does not only depend on endothelial cell invasion and proliferation: it also requires pericyte coverage of vascular sprouts for vessel stabilization. These processes are coordinated by vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) through their cognate receptors on endothelial cells and vascular smooth muscle cells (VSMCs), respectively. PDGF induces neovascularization by priming VSMCs/pericytes to release pro-angiogenic mediators. Although VEGF directly stimulates endothelial cell proliferation and migration, its role in pericyte biology is less clear. Here we define a role for VEGF as an inhibitor of neovascularization on the basis of its capacity to disrupt VSMC function. Specifically, under conditions of PDGF-mediated angiogenesis, VEGF ablates pericyte coverage of nascent vascular sprouts, leading to vessel destabilization. At the molecular level, VEGF-mediated activation of VEGF-R2 suppresses PDGF-Rbeta signalling in VSMCs through the assembly of a previously undescribed receptor complex consisting of PDGF-Rbeta and VEGF-R2. Inhibition of VEGF-R2 not only prevents assembly of this receptor complex but also restores angiogenesis in tissues exposed to both VEGF and PDGF. Finally, genetic deletion of tumour cell VEGF disrupts PDGF-Rbeta/VEGF-R2 complex formation and increases tumour vessel maturation. These findings underscore the importance of VSMCs/pericytes in neovascularization and reveal a dichotomous role for VEGF and VEGF-R2 signalling as both a promoter of endothelial cell function and a negative regulator of VSMCs and vessel maturation.


Subject(s)
Blood Vessels/metabolism , Neovascularization, Physiologic/physiology , Pericytes/metabolism , Platelet-Derived Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line , Cells, Cultured , Fibrosarcoma/blood supply , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Physiologic/drug effects , Pericytes/drug effects , Platelet-Derived Growth Factor/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction
8.
Cancer Res ; 84(10): 1630-1642, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38588407

ABSTRACT

Cancer stem/tumor-initiating cells display stress tolerance and metabolic flexibility to survive in a harsh environment with limited nutrient and oxygen availability. The molecular mechanisms underlying this phenomenon could provide targets to prevent metabolic adaptation and halt cancer progression. Here, we showed in cultured cells and live human surgical biopsies of non-small cell lung cancer that nutrient stress drives the expression of the epithelial cancer stem cell marker integrin αvß3 via upregulation of the ß3 subunit, resulting in a metabolic reprogramming cascade that allows tumor cells to thrive despite a nutrient-limiting environment. Although nutrient deprivation is known to promote acute, yet transient, activation of the stress sensor AMP-activated protein kinase (AMPK), stress-induced αvß3 expression via Src activation unexpectedly led to secondary and sustained AMPK activation. This resulted in the nuclear localization of peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC1α) and upregulation of glutamine metabolism, the tricarboxylic acid cycle, and oxidative phosphorylation. Pharmacological or genetic targeting of this axis prevented lung cancer cells from evading the effects of nutrient stress, thereby blocking tumor initiation in mice following orthotopic implantation of lung cancer cells. These findings reveal a molecular pathway driven by nutrient stress that results in cancer stem cell reprogramming to promote metabolic flexibility and tumor initiation. SIGNIFICANCE: Upregulation of integrin αvß3, a cancer stem cell marker, in response to nutrient stress activates sustained AMPK/PGC1α signaling that induces metabolic reprogramming in lung cancer cells to support their survival. See related commentary by Rainero, p. 1543.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Integrin alphaVbeta3 , Lung Neoplasms , Up-Regulation , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Animals , Integrin alphaVbeta3/metabolism , Mice , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , AMP-Activated Protein Kinases/metabolism , Stress, Physiological , Nutrients/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
9.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38959328

ABSTRACT

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Subject(s)
COVID-19 , Class Ib Phosphatidylinositol 3-Kinase , Inflammation , SARS-CoV-2 , Animals , Humans , Mice , Capillary Permeability/drug effects , Class Ib Phosphatidylinositol 3-Kinase/metabolism , COVID-19/pathology , COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Inflammation/pathology , Lung/pathology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice, Inbred C57BL , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , SARS-CoV-2/physiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/pathology
10.
Nature ; 446(7136): 690-4, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17377533

ABSTRACT

Inflammation enhances tumour promotion through NF-kappaB-dependent mechanisms. NF-kappaB was also proposed to promote metastatogenesis through epithelial-mesenchymal transition. Yet a mechanistic link between inflammation and metastasis is missing. We identified a role for IkappaB kinase alpha (IKKalpha), activated by receptor activator of NF-kappaB (RANK/TNFRSF11A), in mammary epithelial proliferation during pregnancy. Owing to similarities between mammary and prostate epithelia, we examined IKKalpha involvement in prostate cancer and its progression. Here we show that a mutation that prevents IKKalpha activation slows down CaP growth and inhibits metastatogenesis in TRAMP mice, which express SV40 T antigen in the prostate epithelium. Decreased metastasis correlated with elevated expression of the metastasis suppressor Maspin, the ablation of which restored metastatic activity. IKKalpha activation by RANK ligand (RANKL/TNFSF11) inhibits Maspin expression in prostate epithelial cells, whereas repression of Maspin transcription requires nuclear translocation of active IKKalpha. The amount of active nuclear IKKalpha in mouse and human prostate cancer correlates with metastatic progression, reduced Maspin expression and infiltration of prostate tumours with RANKL-expressing inflammatory cells. We propose that tumour-infiltrating RANKL-expressing cells lead to nuclear IKKalpha activation and inhibition of Maspin transcription, thereby promoting the metastatic phenotype.


Subject(s)
Cytokines/metabolism , I-kappa B Kinase/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serpins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Movement , Female , Gene Expression , Genes, Tumor Suppressor , Humans , Inflammation , Male , Mice , Neoplasm Metastasis , Prostatic Neoplasms/enzymology , Receptors, Tumor Necrosis Factor, Member 25/deficiency , Receptors, Tumor Necrosis Factor, Member 25/genetics , Serpins/genetics
11.
Nat Rev Cancer ; 2(2): 91-100, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12635172

ABSTRACT

As cancer cells undergo metastasis--invasion and migration of a new tissue--they penetrate and attach to the target tissue's basal matrix. This allows the cancer cell to pull itself forward into the tissue. The attachment is mediated by cell-surface receptors known as integrins, which bind to components of the extracellular matrix. Integrins are crucial for cell invasion and migration, not only for physically tethering cells to the matrix, but also for sending and receiving molecular signals that regulate these processes.


Subject(s)
Cell Movement , Integrins/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Apoptosis , Basement Membrane/pathology , Cell Survival , Clinical Trials as Topic , Extracellular Matrix/metabolism , Humans , Integrins/antagonists & inhibitors , Signal Transduction
12.
Proc Natl Acad Sci U S A ; 107(5): 2189-94, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080647

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies. To discover functionally relevant modulators of pancreatic neoplasia, we performed activity-based proteomic profiling on primary human ductal adenocarcinomas. Here, we identify retinoblastoma-binding protein 9 (RBBP9) as a tumor-associated serine hydrolase that displays elevated activity in pancreatic carcinomas. Whereas RBBP9 is expressed in normal and malignant tissues at similar levels, its elevated activity in tumor cells promotes anchorage-independent growth in vitro as well as pancreatic carcinogenesis in vivo. At the molecular level, RBBP9 activity overcomes TGF-beta-mediated antiproliferative signaling by reducing Smad2/3 phosphorylation, a previously unknown role for a serine hydrolase in cancer biology. Conversely, loss of endogenous RBBP9 or expression of mutationally inactive RBBP9 leads to elevated Smad2/3 phosphorylation, implicating this serine hydrolase as an essential suppressor of TGF-beta signaling. Finally, RBBP9-mediated suppression of TGF-beta signaling is required for E-cadherin expression as loss of the serine hydrolase activity leads to a reduction in E-cadherin levels and a concomitant decrease in the integrity of tumor cell-cell junctions. These data not only define a previously uncharacterized serine hydrolase activity associated with epithelial neoplasia, but also demonstrate the potential benefit of functional proteomics in the identification of new therapeutic targets.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/enzymology , Amino Acid Sequence , Animals , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/genetics , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Phosphorylation , Proteomics , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
13.
Proc Natl Acad Sci U S A ; 107(9): 4299-304, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20154271

ABSTRACT

Kinases are known to regulate fundamental processes in cancer including tumor proliferation, metastasis, neovascularization, and chemoresistance. Accordingly, kinase inhibitors have been a major focus of drug development, and several kinase inhibitors are now approved for various cancer indications. Typically, kinase inhibitors are selected via high-throughput screening using catalytic kinase domains at low ATP concentration, and this process often yields ATP mimetics that lack specificity and/or function poorly in cells where ATP levels are high. Molecules targeting the allosteric site in the inactive kinase conformation (type II inhibitors) provide an alternative for developing selective inhibitors that are physiologically active. By applying a rational design approach using a constrained amino-triazole scaffold predicted to stabilize kinases in the inactive state, we generated a series of selective type II inhibitors of PDGFRbeta and B-RAF, important targets for pericyte recruitment and endothelial cell survival, respectively. These molecules were designed in silico and screened for antivascular activity in both cell-based models and a Tg(fli1-EGFP) zebrafish embryogenesis model. Dual inhibition of PDGFRbeta and B-RAF cellular signaling demonstrated synergistic antiangiogenic activity in both zebrafish and murine models of angiogenesis, and a combination of previously characterized PDGFRbeta and RAF inhibitors validated the synergy. Our lead compound was selected as an orally active molecule with favorable pharmacokinetic properties which demonstrated target inhibition in vivo leading to suppression of murine orthotopic tumors in both the kidney and pancreas.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma, Renal Cell/pathology , Cell Division/drug effects , Kidney Neoplasms/pathology , Neovascularization, Pathologic , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Administration, Oral , Angiogenesis Inhibitors/therapeutic use , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Zebrafish
14.
Trends Cell Biol ; 2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37640611

ABSTRACT

While the tumor microenvironment is a critical contributor to cancer progression, early steps of tumor initiation and metastasis also rely on the ability of individual tumor cells to survive and thrive at locations where tumor stroma or immune infiltration has yet to be established. In this opinion article, we use the term 'isolation stress' to broadly describe the challenges that individual tumor cells must overcome during the initiation and expansion of the primary tumor beyond permissive boundaries and metastatic spread into distant sites, including a lack of cell-cell contact, adhesion to protumor extracellular matrix proteins, and access to nutrients, oxygen, and soluble factors that support growth. In particular, we highlight the ability of solitary tumor cells to autonomously generate a specialized fibronectin-enriched extracellular matrix to create their own pericellular niche that supports tumor initiation. Cancer cells that can creatively evade the effects of isolation stress not only become more broadly stress tolerant, they also tend to show enhanced stemness, drug resistance, tumor initiation, and metastasis.

15.
Nat Cell Biol ; 25(2): 309-322, 2023 02.
Article in English | MEDLINE | ID: mdl-36646789

ABSTRACT

Defining drivers of tumour initiation can provide opportunities to control cancer progression. Here we report that lysophosphatidic acid receptor 4 (LPAR4) becomes transiently upregulated on pancreatic cancer cells exposed to environmental stress or chemotherapy where it promotes stress tolerance, drug resistance, self-renewal and tumour initiation. Pancreatic cancer cells gain LPAR4 expression in response to stress by downregulating a tumour suppressor, miR-139-5p. Even in the absence of exogenous lysophosphatidic acid, LPAR4-expressing tumour cells display an enrichment of extracellular matrix genes that are established drivers of cancer stemness. Mechanistically, upregulation of fibronectin via an LPAR4/AKT/CREB axis is indispensable for LPAR4-induced tumour initiation and stress tolerance. Moreover, ligation of this fibronectin-containing matrix via integrins α5ß1 or αVß3 can transfer stress tolerance to LPAR4-negative cells. Therefore, stress- or drug-induced LPAR4 enhances cell-autonomous production of a fibronectin-rich extracellular matrix, allowing cells to survive 'isolation stress' and compensate for the absence of stromal-derived factors by creating their own tumour-initiating niche.


Subject(s)
MicroRNAs , Pancreatic Neoplasms , Receptors, Purinergic P2 , Humans , Fibronectins/genetics , Fibronectins/metabolism , Pancreatic Neoplasms/pathology , Extracellular Matrix/metabolism , Cell Transformation, Neoplastic/metabolism , Receptors, Purinergic P2/metabolism , MicroRNAs/genetics , Pancreatic Neoplasms
16.
Matrix Biol ; 124: 49-62, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37956856

ABSTRACT

Highly aggressive, metastatic, neuroendocrine prostate cancer, which typically develops from prostate cancer cells acquiring resistance to androgen deprivation therapy, is associated with limited treatment options and hence poor prognosis. We have previously demonstrated that the αVß3 integrin is over-expressed in neuroendocrine prostate cancer. We now show that LM609, a monoclonal antibody that specifically targets the human αVß3 integrin, hinders the growth of neuroendocrine prostate cancer patient-derived xenografts in vivo. Our group has recently identified a novel αVß3 integrin binding partner, NgR2, responsible for regulating the expression of neuroendocrine markers and for inducing neuroendocrine differentiation in prostate cancer cells. Through in vitro functional assays, we here demonstrate that NgR2 is crucial in promoting cell adhesion to αVß3 ligands. Moreover, we describe for the first time co-fractionation of αVß3 integrin and NgR2 in small extracellular vesicles derived from metastatic prostate cancer patients' plasma. These prostate cancer patient-derived small extracellular vesicles have a functional impact on human monocytes, increasing their adhesion to fibronectin. The monocytes incubated with small extracellular vesicles do not show an associated change in conventional polarization marker expression and appear to be in an early stage that may be defined as "adhesion competent". Overall, these findings allow us to better understand integrin-directed signaling and cell-cell communication during cancer progression. Furthermore, our results pave the way for new diagnostic and therapeutic perspectives for patients affected by neuroendocrine prostate cancer.


Subject(s)
Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Androgen Antagonists , Signal Transduction , Antibodies, Monoclonal , Integrins , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Cell Line, Tumor
17.
Int J Cancer ; 131(8): 1744-54, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22275185

ABSTRACT

The recepteur d'origine nantais (RON) receptor tyrosine kinase is overexpressed and stimulates invasive growth in pancreatic cancer cells, yet the mechanisms that underlie RON-mediated phenotypes remain poorly characterized. To better understand RON function in pancreatic cancer cells, we sought to identify novel RON interactants using multidimensional protein identification analysis. These studies revealed plectin, a large protein of the spectrin superfamily, to be a novel RON interactant. Plectin is a multifunctional protein that complexes with integrin-ß4 (ITGB4) to form hemidesmosomes, serves as a scaffolding platform crucial to the function of numerous protein signaling pathways and was recently described as an overexpressed protein in pancreatic cancer (Bausch D et al., Clin Cancer Res 2010; Kelly et al., PLoS Med 2008;5:e85). In this study, we demonstrate that on exposure to its ligand, macrophage-stimulating protein, RON binds to plectin and ITGB4, which results in disruption of the plectin-ITGB4 interaction and enhanced cell migration, a phenotype that can be recapitulated by small hairpin ribosomal nucleic acid (shRNA)-mediated suppression of plectin expression. We demonstrate that disruption of plectin-ITGB4 is dependent on RON and phosphoinositide-3 (PI3) kinase, but not mitogen-activated protein kinase (MEK), activity. Thus, in pancreatic cancer cells, plectin and ITGB4 form hemidesmosomes which serve to anchor cells to the extracellular matrix (ECM) and restrain migration. The current study defines a novel interaction between RON and plectin, provides new insight into RON-mediated migration and further supports efforts to target RON kinase activity in pancreatic cancer.


Subject(s)
Cell Movement , Hemidesmosomes/metabolism , Integrin beta4/metabolism , Pancreatic Neoplasms/pathology , Plectin/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Chromatography, Liquid , Fluorescent Antibody Technique , Humans , Kidney/cytology , Kidney/metabolism , Mitogen-Activated Protein Kinases , Pancreatic Neoplasms/metabolism , Phosphorylation , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Wound Healing
18.
Nature ; 439(7072): 95-9, 2006 Jan 05.
Article in English | MEDLINE | ID: mdl-16397500

ABSTRACT

Neuroblastoma, the most common paediatric solid tumour, arises from defective neural crest cells. Genetic alterations occur frequently in the most aggressive neuroblastomas. In particular, deletion or suppression of the proapoptotic enzyme caspase-8 is common in malignant, disseminated disease, although the effect of this loss on disease progression is unclear. Here we show that suppression of caspase-8 expression occurs during the establishment of neuroblastoma metastases in vivo, and that reconstitution of caspase-8 expression in deficient neuroblastoma cells suppressed their metastases. Caspase-8 status was not a predictor of primary tumour growth; rather, caspase-8 selectively potentiated apoptosis in neuroblastoma cells invading the collagenous stroma at the tumour margin. Apoptosis was initiated by unligated integrins by means of a process known as integrin-mediated death. Loss of caspase-8 or integrin rendered these cells refractory to integrin-mediated death, allowed cellular survival in the stromal microenvironment, and promoted metastases. These findings define caspase-8 as a metastasis suppressor gene that, together with integrins, regulates the survival and invasive capacity of neuroblastoma cells.


Subject(s)
Caspases/deficiency , Caspases/genetics , Neoplasm Metastasis/pathology , Neuroblastoma/enzymology , Neuroblastoma/pathology , Animals , Apoptosis , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , Cell Survival , Chick Embryo , Female , Gene Expression Regulation, Neoplastic , Humans , Integrins/metabolism , Kidney/pathology , Mice , Mice, Nude , Neoplasm Metastasis/genetics , Neoplasm Transplantation , Neuroblastoma/genetics , Ovary/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Curr Opin Hematol ; 18(3): 171-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21423013

ABSTRACT

PURPOSE OF REVIEW: It has been known for decades that in order to grow, tumors need to activate quiescent endothelial cells to form a functional vascular network, a process termed 'angiogenesis'. However, the molecular determinants that reverse this endothelial quiescence to facilitate pathological angiogenesis are not yet completely understood. This review examines a critical regulatory switch at the level of Ras that activates this angiogenic switch process and the role that microRNAs play in this process. RECENT FINDINGS: In the last few years, microRNAs, a new class of small RNA molecules, have emerged as key regulators of several cellular processes, including angiogenesis. MicroRNAs such as miR-126, miR-296, and miR-92a have been shown to play important roles in angiogenesis. We recently described how miR-132, an angiogenic growth factor inducible microRNA in the endothelium, facilitates pathological angiogenesis by downregulating p120RasGAP, a molecular brake for Ras. Importantly, targeting miR-132 with a complementary, synthetic antimicroRNA restored the brake and decreased angiogenesis and tumor burden in multiple tumor models. Taken together, emerging evidence suggests a central role for microRNAs downstream of multiple growth factors in regulating endothelial proliferation, migration, and vascular patterning. SUMMARY: Further research into miR-132-p120RasGAP biology and more broadly, microRNA regulation of Ras pathways in the endothelium will not only advance our understanding of angiogenesis but also provide opportunities for therapeutic intervention.


Subject(s)
MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , Animals , Endothelium, Vascular/metabolism , Humans , MicroRNAs/genetics
20.
Carcinogenesis ; 32(8): 1151-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21565828

ABSTRACT

The RON receptor tyrosine kinase (RTK) is overexpressed in the majority of pancreatic cancers, yet its role in pancreatic cancer cell biology remains to be clarified. Recent work in childhood sarcoma identified RON as a mediator of resistance to insulin-like growth factor receptor (IGF1-R)-directed therapy. To better understand RON function in pancreatic cancer cells, we sought to identify novel RON interactants. Using multidimensional protein identification analysis, IGF-1R was identified and confirmed to interact with RON in pancreatic cancer cell lines. IGF-1 induces rapid phosphorylation of RON, but RON signaling did not activate IGF-1R indicating unidirectional signaling between these RTKs. We next demonstrate that IGF-1 induces pancreatic cancer cell migration that is RON dependent, as inhibition of RON signaling by either shRNA-mediated RON knockdown or by a RON kinase inhibitor abrogated IGF-1 induced wound closure in a scratch assay. In pancreatic cancer cells, unlike childhood sarcoma, STAT-3, rather than RPS6, is activated in response to IGF-1, in a RON-dependent manner. The current study defines a novel interaction between RON and IGF-1R and taken together, these two studies demonstrate that RON is an important mediator of IGF1-R signaling and that this finding is consistent in both human epithelial and mesenchymal cancers. These findings demand additional investigation to determine if IGF-1R independent RON activation is associated with resistance to IGF-1R-directed therapies in vivo and to identify suitable biomarkers of activated RON signaling.


Subject(s)
Cell Movement/physiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Blotting, Western , Cell Adhesion , Humans , Immunoprecipitation , Pancreatic Neoplasms/genetics , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Tumor Cells, Cultured , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL