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











Publication year range
1.
Front Immunol ; 14: 1154528, 2023.
Article in English | MEDLINE | ID: mdl-37539058

ABSTRACT

The desmoplastic reaction observed in many cancers is a hallmark of disease progression and prognosis, particularly in breast and pancreatic cancer. Stromal-derived extracellular matrix (ECM) is significantly altered in desmoplasia, and as such plays a critical role in driving cancer progression. Using fibroblast-derived matrices (FDMs), we show that cancer cells have increased growth on cancer associated FDMs, when compared to FDMs derived from non-malignant tissue (normal) fibroblasts. We assess the changes in ECM characteristics from normal to cancer-associated stroma at the primary tumor site. Compositional, structural, and mechanical analyses reveal significant differences, with an increase in abundance of core ECM proteins, coupled with an increase in stiffness and density in cancer-associated FDMs. From compositional changes of FDM, we derived a 36-ECM protein signature, which we show matches in large part with the changes in pancreatic ductal adenocarcinoma (PDAC) tumor and metastases progression. Additionally, this signature also matches at the transcriptomic level in multiple cancer types in patients, prognostic of their survival. Together, our results show relevance of FDMs for cancer modelling and identification of desmoplastic ECM components for further mechanistic studies.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Prognosis , Pancreatic Neoplasms/pathology , Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/pathology , Extracellular Matrix Proteins , Pancreatic Neoplasms
2.
Adv Healthc Mater ; 11(1): e2100684, 2022 01.
Article in English | MEDLINE | ID: mdl-34734500

ABSTRACT

Metastatic cancer spread is responsible for most cancer-related deaths. To colonize a new organ, invading cells adapt to, and remodel, the local extracellular matrix (ECM), a network of proteins and proteoglycans underpinning all tissues, and a critical regulator of homeostasis and disease. However, there is a major lack in tools to study cancer cell behavior within native 3D ECM. Here, an in-house designed bioreactor, where mouse organ ECM scaffolds are perfused and populated with cells that are challenged to colonize it, is presented. Using a specialized bioreactor chamber, it is possible to monitor cell behavior microscopically (e.g., proliferation, migration) within the organ scaffold. Cancer cells in this system recapitulate cell signaling observed in vivo and remodel complex native ECM. Moreover, the bioreactors are compatible with co-culturing cell types of different genetic origin comprising the normal and tumor microenvironment. This degree of experimental flexibility in an organ-specific and 3D context, opens new possibilities to study cell-cell and cell-ECM interplay and to model diseases in a controllable organ-specific system ex vivo.


Subject(s)
Extracellular Matrix , Tissue Scaffolds , Animals , Bioreactors , Mice , Perfusion , Proteoglycans , Tissue Engineering
3.
J Invasive Cardiol ; 33(12): E998-E1003, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34817395

ABSTRACT

OBJECTIVE: We aimed to characterize outcomes associated with cangrelor administration used in an antiplatelet bridging strategy in real-world clinical scenarios within a large academic medical system. BACKGROUND: Cangrelor has been used for antiplatelet bridging in perioperative settings or for patients unable to take oral medications. Prior studies in these settings have reported bleeding rates from 0%-40%. METHODS: Patients were retrospectively identified via chart review and included if they were over 18 years old, had coronary or peripheral arterial stents, and had received at least 1 hour of cangrelor infusion during inpatient admission. The primary endpoint was Bleeding Academic Research Consortium (BARC) 3-5 bleeding during cangrelor infusion or within 48 hours of discontinuation; secondary endpoints were bleeding events defined by Thrombolysis in Myocardial Infarction (TIMI), Global Use of Strategies to Open Occluded Arteries (GUSTO), and International Society on Thrombosis and Hemostasis (ISTH) criteria, as well as BARC 2 bleeding. RESULTS: Thirty-one patients met the inclusion criteria. Cangrelor indications were bridging to procedure in 22 patients (71.0%) and inability to take oral P2Y12 inhibitors in 9 patients (29.0%). Twenty-three patients (74.2%) were men, 11 patients (35.5%) were in cardiogenic shock, and 4 patients (12.9%) were on extracorporeal membrane oxygenation (ECMO) at the time of administration. No patients received cangrelor for routine percutaneous coronary intervention. Of the 31 patients, 13 (41.9%) had BARC 3-5 bleeding and 7 (22.6%) expired during hospitalization. All 4 patients on ECMO suffered BARC 3-5 bleeding. CONCLUSIONS: We reviewed the use of cangrelor for antiplatelet bridging in real-world clinical scenarios and observed higher rates of clinically significant bleeding than seen in other similar studies. Our study suggests careful consideration when using cangrelor in a sick patient population.


Subject(s)
Adenosine Monophosphate , Blood Platelets/drug effects , Platelet Aggregation Inhibitors , Adenosine Monophosphate/administration & dosage , Adenosine Monophosphate/adverse effects , Adenosine Monophosphate/analogs & derivatives , Adolescent , Humans , Infusions, Intravenous , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/adverse effects , Retrospective Studies
4.
Cancers (Basel) ; 13(13)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34283050

ABSTRACT

During the metastatic process, breast cancer cells must come into contact with the extra-cellular matrix (ECM) at every step. The ECM provides both structural support and biochemical cues, and cell-ECM interactions can lead to changes in drug response. Here, we used fibroblast-derived ECM (FDM) to perform high throughput drug screening of 4T1 breast cancer cells on metastatic organ ECM (lung), and we see that drug response differs from treatment on plastic. The FDMs that we can produce from different organs are abundant in and contains a complex mixture of ECM proteins. We also show differences in ECM composition between the primary site and secondary organ sites. Furthermore, we show that global kinase signalling of 4T1 cells on the ECM is relatively unchanged between organs, while changes in signalling compared to plastic are significant. Our study highlights the importance of context when testing drug response in vitro, showing that consideration of the ECM is critically important.

5.
Nat Commun ; 12(1): 3414, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099731

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) patients have a 5-year survival rate of only 8% largely due to late diagnosis and insufficient therapeutic options. Neutrophils are among the most abundant immune cell type within the PDAC tumor microenvironment (TME), and are associated with a poor clinical prognosis. However, despite recent advances in understanding neutrophil biology in cancer, therapies targeting tumor-associated neutrophils are lacking. Here, we demonstrate, using pre-clinical mouse models of PDAC, that lorlatinib attenuates PDAC progression by suppressing neutrophil development and mobilization, and by modulating tumor-promoting neutrophil functions within the TME. When combined, lorlatinib also improves the response to anti-PD-1 blockade resulting in more activated CD8 + T cells in PDAC tumors. In summary, this study identifies an effect of lorlatinib in modulating tumor-associated neutrophils, and demonstrates the potential of lorlatinib to treat PDAC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Lactams, Macrocyclic/pharmacology , Neutrophils/drug effects , Pancreatic Neoplasms/drug therapy , Aminopyridines , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Synergism , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Lactams , Lactams, Macrocyclic/therapeutic use , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Transgenic , Neutrophils/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Pyrazoles , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
Adv Healthc Mater ; 9(7): e1901669, 2020 04.
Article in English | MEDLINE | ID: mdl-32129003

ABSTRACT

In native tissues, the interaction between cells and the surrounding extracellular matrix (ECM) is reciprocal, as cells not only receive signals from the ECM but also actively remodel it through secretion of cell-derived ECM. However, very little is known about the reciprocal interaction between cells and their secreted ECM within synthetic biomaterials that mimic the ECM for use in engineering of tissues for regenerative medicine or as tissue models. Here, poly(ethylene glycol) (PEG) hydrogels with fully defined biomaterial properties are used to investigate the emerging role of cell-derived ECM on culture outcomes. It is shown that human mesenchymal stromal cells (MSCs) secrete ECM proteins into the pericellular space early after encapsulation and that, even in the absence of material-presented cell adhesion motifs, cell-derived fibronectin enables cell spreading. Then, it is investigated how different culture conditions influence MSC ECM expression in hydrogels. Most strikingly, it is found by RNA sequencing that the fibroblast growth factor 2 (FGF-2) changes ECM gene expression and, in particular, decreases the expression of structural ECM components including fibrillar collagens. In summary, this work shows that cell-derived ECM is a guiding cue in 3D hydrogels and that FGF-2 is a potentially important ECM regulator within bioengineered cell and tissue systems.


Subject(s)
Hydrogels , Mesenchymal Stem Cells , Cell Adhesion , Extracellular Matrix , Fibroblast Growth Factor 2/pharmacology , Humans , Hydrogels/pharmacology
7.
Life Sci Alliance ; 2(3)2019 06.
Article in English | MEDLINE | ID: mdl-31160380

ABSTRACT

Mesenchymal stromal cells (MSCs) are key contributors of the tumour microenvironment and are known to promote cancer progression through reciprocal communication with cancer cells, but how they become activated is not fully understood. Here, we investigate how breast cancer cells from different stages of the metastatic cascade convert MSCs into tumour-associated MSCs (TA-MSCs) using unbiased, global approaches. Using mass spectrometry, we compared the secretomes of MCF-7 cells, invasive MDA-MB-231 cells, and sublines isolated from bone, lung, and brain metastases and identified ECM and exosome components associated with invasion and organ-specific metastasis. Next, we used synthetic hydrogels to investigate how these different secretomes activate MSCs in bioengineered 3D microenvironments. Using kinase activity profiling and RNA sequencing, we found that only MDA-MB-231 breast cancer secretomes convert MSCs into TA-MSCs, resulting in an immunomodulatory phenotype that was particularly prominent in response to bone-tropic cancer cells. We have investigated paracrine signalling from breast cancer cells to TA-MSCs in 3D, which may highlight new potential targets for anticancer therapy approaches aimed at targeting tumour stroma.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Mesenchymal Stem Cells/metabolism , Metabolome , Proteome , Tumor Microenvironment , Biomarkers , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Chromatography, Liquid , Female , Gene Expression Profiling , Humans , Mass Spectrometry , Metabolomics/methods , Proteomics/methods , Tumor Cells, Cultured , Tumor Microenvironment/genetics
8.
Oncotarget ; 9(53): 30173-30188, 2018 Jul 10.
Article in English | MEDLINE | ID: mdl-30046396

ABSTRACT

Every year more than 8 million people suffer from cancer-related deaths worldwide [1]. Metastasis, the spread of cancer to distant sites, accounts for 90% of these deaths. A promising target for blocking tumor progression, without causing severe side effects [2], is Tumor Endothelial Marker 8 (TEM8), an integrin-like cell surface protein expressed predominantly in the tumor endothelium and in cancer cells [3, 4]. Here, we have investigated the role of TEM8 in cancer progression, angiogenesis and metastasis in invasive breast cancer, and validated the main findings and important results in colorectal cancer. We show that the loss of TEM8 in cancer cells results in inhibition of cancer progression, reduction in tumor angiogenesis and reduced metastatic burden in breast cancer mouse models. Furthermore, we show that TEM8 regulates cancer progression by affecting the expression levels of cell cycle-related genes. Taken together, our findings may have broad clinical and therapeutic potential for breast and colorectal primary tumor and metastasis treatment by targeting TEM8.

9.
EMBO Rep ; 19(8)2018 08.
Article in English | MEDLINE | ID: mdl-29967223

ABSTRACT

The fate of mesenchymal stem cells (MSCs) in the perivascular niche, as well as factors controlling their fate, is poorly understood. Here, we study MSCs in the perivascular microenvironment of endothelial capillaries by modifying a synthetic 3D biomimetic poly(ethylene glycol) (PEG)-hydrogel system in vitro We show that MSCs together with endothelial cells form micro-capillary networks specifically in soft PEG hydrogels. Transcriptome analysis of human MSCs isolated from engineered capillaries shows a prominent switch in extracellular matrix (ECM) production. We demonstrate that the ECM phenotypic switch of MSCs can be recapitulated in the absence of endothelial cells by functionalizing PEG hydrogels with the Notch-activator Jagged1. Moreover, transient culture of MSCs in Notch-inducing microenvironments reveals the reversibility of this ECM switch. These findings provide insight into the perivascular commitment of MSCs by use of engineered niche-mimicking synthetic hydrogels.


Subject(s)
Cell Lineage , Hydrogels/pharmacology , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Receptors, Notch/metabolism , Bone Marrow Cells/cytology , Capillaries/drug effects , Capillaries/physiology , Capillaries/ultrastructure , Cell Lineage/drug effects , Cellular Microenvironment/drug effects , Coculture Techniques , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Polyethylene Glycols/pharmacology
10.
Proteomics ; 18(2)2018 01.
Article in English | MEDLINE | ID: mdl-29178193

ABSTRACT

The nematode Caenorhabditis elegans is widely used as a model organism to study cell and developmental biology. Quantitative proteomics of C. elegans is still in its infancy and, so far, most studies have been performed on adult worm samples. Here, we used quantitative mass spectrometry to characterize protein level changes across the four larval developmental stages (L1-L4) of C. elegans. In total, we identified 4130 proteins, and quantified 1541 proteins that were present across all four stages in three biological replicates from independent experiments. Using hierarchical clustering and functional ontological analyses, we identified 21 clusters containing proteins with similar protein profiles across the four stages, and highlighted the most overrepresented biological functions in each of these protein clusters. In addition, we used the dataset to identify putative larval stage-specific proteins in each individual developmental stage, as well as in the early and late developmental stages. In summary, this dataset provides system-wide analysis of protein level changes across the four C. elegans larval developmental stages, which serves as a useful resource for the C. elegans research community. MS data were deposited in ProteomeXchange (http://proteomecentral.proteomexchange.org) via the PRIDE partner repository with the primary accession identifier PXD006676.


Subject(s)
Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/growth & development , Animals , Larva/chemistry , Proteomics , Tandem Mass Spectrometry
11.
Nat Med ; 23(7): 890-898, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28604702

ABSTRACT

The extracellular matrix (ECM) is a master regulator of cellular phenotype and behavior. It has a crucial role in both normal tissue homeostasis and disease pathology. Here we present a fast and efficient approach to enhance the study of ECM composition and structure. Termed in situ decellularization of tissues (ISDoT), it allows whole organs to be decellularized, leaving native ECM architecture intact. These three-dimensional decellularized tissues can be studied using high-resolution fluorescence and second harmonic imaging, and can be used for quantitative proteomic interrogation of the ECM. Our method is superior to other methods tested in its ability to preserve the structural integrity of the ECM, facilitate high-resolution imaging and quantitatively detect ECM proteins. In particular, we performed high-resolution sub-micron imaging of matrix topography in normal tissue and over the course of primary tumor development and progression to metastasis in mice, providing the first detailed imaging of the metastatic niche. These data show that cancer-driven ECM remodeling is organ specific, and that it is accompanied by comprehensive changes in ECM composition and topological structure. We also describe differing patterns of basement-membrane organization surrounding different types of blood vessels in healthy and diseased tissues. The ISDoT procedure allows for the study of native ECM structure under normal and pathological conditions in unprecedented detail.


Subject(s)
Basement Membrane/ultrastructure , Breast Neoplasms/ultrastructure , Extracellular Matrix/ultrastructure , Mammary Neoplasms, Experimental/ultrastructure , Proteomics , Tumor Microenvironment , Animals , Basement Membrane/metabolism , Breast Neoplasms/metabolism , Extracellular Matrix/metabolism , Female , Humans , Imaging, Three-Dimensional , Lactation , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/ultrastructure , Lymph Nodes/metabolism , Lymph Nodes/ultrastructure , Lymphatic Metastasis , Mammary Glands, Human/metabolism , Mammary Glands, Human/ultrastructure , Mammary Neoplasms, Experimental/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/ultrastructure , Mice , Optical Imaging , Peripheral Nerves/metabolism , Peripheral Nerves/ultrastructure , Tongue Neoplasms/metabolism , Tongue Neoplasms/ultrastructure
12.
Dev Neurobiol ; 77(6): 726-737, 2017 06.
Article in English | MEDLINE | ID: mdl-27618403

ABSTRACT

Hypoxic-ischemic (HI) brain injury is a major cause of neurological abnormalities in the perinatal period. Inflammation contributes to the evolution of HI brain injury. Inter-alpha inhibitor proteins (IAIPs) are a family of proteins that are part of the innate immune system. We have reported that endogenous IAIPs exhibit developmental changes in ovine brain and that exogenous IAIP treatment reduces neuronal death in HI neonatal rats. However, the effects of HI on endogenous IAIPs in brain have not been previously examined. In this study, we examined the effects of ischemia-reperfusion on endogenous IAIPs levels in fetal sheep brain. Cerebral cortex, cerebellum, cervical spinal cord, choroid plexus, and CSF were snap frozen from sham control fetuses at 127 days gestation and after 30-min of carotid occlusion and 4-, 24-, and 48-h of reperfusion. IAIP levels were determined by Western immunoblot. IAIP expressions of the 250 kDa Inter-alpha inhibitor (IaI) and 125 kDa Pre-alpha inhibitor (PaI) in cerebral cortex and PaI in cerebellum were reduced (p < 0.05) 4-h after ischemia compared with controls and returned toward control levels 24- and 48-h after ischemia. CSF PaI and IaI were reduced 48 h after ischemia. We conclude that IAIPs in cerebral cortex and cerebellum are reduced by brain ischemia, and return toward control levels between 24 and 48 h after ischemia. However, changes in CSF IAIPs were delayed, exhibiting decreases 48 h after ischemia. We speculate that the decreases in endogenous IAIPs reflect increased utilization, potentially suggesting that they have endogenous neuroprotective properties. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 726-737, 2017.


Subject(s)
Alpha-Globulins/metabolism , Central Nervous System/metabolism , Gene Expression Regulation, Developmental/physiology , Hypoxia-Ischemia, Brain/pathology , Animals , Central Nervous System/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Fetus/anatomy & histology , Hypoxia-Ischemia, Brain/blood , Molecular Weight , Sheep , Spinal Cord/pathology , Time Factors
13.
J Cell Sci ; 129(22): 4159-4163, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799358

ABSTRACT

The adhesion nexus is the site at which integrin receptors bridge intracellular cytoskeletal and extracellular matrix networks. The connection between integrins and the cytoskeleton is mediated by a dynamic integrin adhesion complex (IAC), the components of which transduce chemical and mechanical signals to control a multitude of cellular functions. In this Cell Science at a Glance article and the accompanying poster, we integrate the consensus adhesome, a set of 60 proteins that have been most commonly identified in isolated IAC proteomes, with the literature-curated adhesome, a theoretical network that has been assembled through scholarly analysis of proteins that localise to IACs. The resulting IAC network, which comprises four broad signalling and actin-bridging axes, provides a platform for future studies of the regulation and function of the adhesion nexus in health and disease.


Subject(s)
Integrins/metabolism , Proteome/metabolism , Animals , Cell Adhesion , Disease , Humans
14.
J Cell Biol ; 212(3): 349-64, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26833789

ABSTRACT

Integrin adhesion complexes (IACs) form mechanochemical connections between the extracellular matrix and actin cytoskeleton and mediate phenotypic responses via posttranslational modifications. Here, we investigate the modularity and robustness of the IAC network to pharmacological perturbation of the key IAC signaling components focal adhesion kinase (FAK) and Src. FAK inhibition using AZ13256675 blocked FAK(Y397) phosphorylation but did not alter IAC composition, as reported by mass spectrometry. IAC composition was also insensitive to Src inhibition using AZD0530 alone or in combination with FAK inhibition. In contrast, kinase inhibition substantially reduced phosphorylation within IACs, cell migration and proliferation. Furthermore using fluorescence recovery after photobleaching, we found that FAK inhibition increased the exchange rate of a phosphotyrosine (pY) reporter (dSH2) at IACs. These data demonstrate that kinase-dependent signal propagation through IACs is independent of gross changes in IAC composition. Together, these findings demonstrate a general separation between the composition of IACs and their ability to relay pY-dependent signals.


Subject(s)
Cell Adhesion/drug effects , Fibroblasts/enzymology , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/enzymology , Signal Transduction/drug effects , src-Family Kinases/metabolism , Animals , Cell Movement , Cell Proliferation , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Focal Adhesions/drug effects , Focal Adhesions/genetics , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Time Factors , Transfection , src Homology Domains , src-Family Kinases/antagonists & inhibitors
15.
Exp Cell Res ; 343(1): 7-13, 2016 04 10.
Article in English | MEDLINE | ID: mdl-26515553

ABSTRACT

Cell and tissue stiffness have been known to contribute to both developmental and pathological signalling for some time, but the underlying mechanisms remain elusive. Integrins and their associated adhesion signalling complexes (IACs), which form a nexus between the cell cytoskeleton and the extracellular matrix, act as a key force sensing and transducing unit in cells. Accordingly, there has been much interest in obtaining a systems-level understanding of IAC composition. Proteomic approaches have revealed the complexity of IACs and identified a large number of components that are regulated by cytoskeletal force. Here we review the function of the consensus adhesome, an assembly of core IAC proteins that emerged from a meta-analysis of multiple proteomic datasets, in the context of mechanosensing. As IAC components have been linked to a variety of diseases involved with rigidity sensing, the field is now in a position to define the mechanosensing function of individual IAC proteins and elucidate their mechanisms of action.


Subject(s)
Focal Adhesions/metabolism , Integrins/metabolism , Models, Biological , Coordination Complexes , Humans , Proteomics , Stress, Mechanical
16.
Proteomics Clin Appl ; 10(1): 51-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26147903

ABSTRACT

PURPOSE: Multipotent mesenchymal stem cells (MSCs) have the capability to differentiate down adipocyte, osteocyte and chondrocyte lineages and as such offer a range of potential therapeutic applications. The composition and stiffness of the extracellular matrix (ECM) environment that surrounds cells dictates their transcriptional programme, thereby affecting stem cell lineage decision-making. Cells sense force via linkages between themselves and their microenvironment, and this is transmitted by integrin receptors and associated adhesion signalling complexes. To identify regulators of MSC force sensing, we sought to catalogue MSC integrin-associated adhesion complex composition. EXPERIMENTAL DESIGN: Adhesion complexes formed by MSCs plated on the ECM ligand fibronectin were isolated and characterised by MS. Identified proteins were interrogated by comparison to a literature-based reference set of cell adhesion-related components and using ontological and protein-protein interaction network analyses. RESULTS: Adhesion complex-specific proteins in MSCs were identified that comprised predominantly cell adhesion-related adaptors and actin cytoskeleton regulators. Furthermore, LIM domain-containing proteins in MSC adhesion complexes were highlighted, which may act as force-sensing components. CONCLUSION AND CLINICAL RELEVANCE: These data provide a valuable resource of information regarding the molecular connections that link integrins and adhesion signalling in MSCs, and as such may present novel opportunities for therapeutic intervention.


Subject(s)
Databases, Protein , Integrins/metabolism , Mesenchymal Stem Cells/metabolism , Humans , Integrins/genetics , Proteomics
17.
Nat Cell Biol ; 17(12): 1577-1587, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26479319

ABSTRACT

Integrin receptor activation initiates the formation of integrin adhesion complexes (IACs) at the cell membrane that transduce adhesion-dependent signals to control a multitude of cellular functions. Proteomic analyses of isolated IACs have revealed an unanticipated molecular complexity; however, a global view of the consensus composition and dynamics of IACs is lacking. Here, we have integrated several IAC proteomes and generated a 2,412-protein integrin adhesome. Analysis of this data set reveals the functional diversity of proteins in IACs and establishes a consensus adhesome of 60 proteins. The consensus adhesome is likely to represent a core cell adhesion machinery, centred around four axes comprising ILK-PINCH-kindlin, FAK-paxillin, talin-vinculin and α-actinin-zyxin-VASP, and includes underappreciated IAC components such as Rsu-1 and caldesmon. Proteomic quantification of IAC assembly and disassembly detailed the compositional dynamics of the core cell adhesion machinery. The definition of this consensus view of integrin adhesome components provides a resource for the research community.


Subject(s)
Focal Adhesions/metabolism , Integrins/metabolism , Proteome/metabolism , Proteomics/methods , Actinin/metabolism , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Focal Adhesions/drug effects , Humans , Immunoblotting , K562 Cells , Kinetics , Mass Spectrometry , Mice , Microscopy, Fluorescence , Nocodazole/pharmacology , Paxillin/metabolism , Protein Interaction Maps , Proteome/classification , Talin/metabolism , Tubulin Modulators/pharmacology , Vinculin/metabolism , Zyxin/metabolism
18.
Proc Natl Acad Sci U S A ; 111(50): 17953-8, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25472840

ABSTRACT

Inactivating mutations in chromodomain helicase DNA binding protein 7 (CHD7) cause CHARGE syndrome, a severe multiorgan system disorder of which Isolated gonadotropin-releasing hormone (GnRH) deficiency (IGD) is a minor feature. Recent reports have described predominantly missense CHD7 alleles in IGD patients, but it is unclear if these alleles are relevant to causality or overall genetic burden of Kallmann syndrome (KS) and normosmic form of IGD. To address this question, we sequenced CHD7 in 783 well-phenotyped IGD patients lacking full CHARGE features; we identified nonsynonymous rare sequence variants in 5.2% of the IGD cohort (73% missense and 27% splice variants). Functional analyses in zebrafish using a surrogate otolith assay of a representative set of these CHD7 alleles showed that rare sequence variants observed in controls showed no altered function. In contrast, 75% of the IGD-associated alleles were deleterious and resulted in both KS and normosmic IGD. In two families, pathogenic mutations in CHD7 coexisted with mutations in other known IGD genes. Taken together, our data suggest that rare deleterious CHD7 alleles contribute to the mutational burden of patients with both KS and normosmic forms of IGD in the absence of full CHARGE syndrome. These findings (i) implicate a unique role or preferential sensitivity for CHD7 in the ontogeny of GnRH neurons, (ii) reiterate the emerging genetic complexity of this family of IGD disorders, and (iii) demonstrate how the coordinated use of well-phenotyped cohorts, families, and functional studies can inform genetic architecture and provide insights into the developmental biology of cellular systems.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Deficiency Diseases/genetics , Gonadotropin-Releasing Hormone/deficiency , Kallmann Syndrome/genetics , Phenotype , Zebrafish/genetics , Animals , Base Sequence , CHARGE Syndrome/genetics , CHARGE Syndrome/pathology , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Gonadotropin-Releasing Hormone/genetics , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Otolithic Membrane/pathology , Protein Structure, Tertiary , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL