Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Arch Pathol Lab Med ; 144(6): 706-724, 2020 06.
Article in English | MEDLINE | ID: mdl-31714809

ABSTRACT

CONTEXT.­: Cancer immunotherapy provides unprecedented rates of durable clinical benefit to late-stage cancer patients across many tumor types, but there remains a critical need for biomarkers to accurately predict clinical response. Although some cancer immunotherapy tests are associated with approved therapies and considered validated, other biomarkers are still emerging and at various states of clinical and translational exploration. OBJECTIVE.­: To provide pathologists with a current and practical update on the evolving field of cancer immunotherapy testing. The scientific background, clinical data, and testing methodology for the following cancer immunotherapy biomarkers are reviewed: programmed death ligand-1 (PD-L1), mismatch repair, microsatellite instability, tumor mutational burden, polymerase δ and ε mutations, cancer neoantigens, tumor-infiltrating lymphocytes, transcriptional signatures of immune responsiveness, cancer immunotherapy resistance biomarkers, and the microbiome. DATA SOURCES.­: Selected scientific publications and clinical trial data representing the current field of cancer immunotherapy. CONCLUSIONS.­: The cancer immunotherapy field, including the use of biomarker testing to predict patient response, is still in evolution. PD-L1, mismatch repair, and microsatellite instability testing are helping to guide the use of US Food and Drug Administration-approved therapies, but there remains a need for better predictors of response and resistance. Several categories of tumor and patient characteristics underlying immune responsiveness are emerging and may represent the next generation of cancer immunotherapy predictive biomarkers. Pathologists have important roles and responsibilities as the field of cancer immunotherapy continues to develop, including leadership of translational studies, exploration of novel biomarkers, and the accurate and timely implementation of newly approved and validated companion diagnostics.


Subject(s)
Biomarkers, Tumor/analysis , Immunotherapy/methods , Neoplasms/therapy , Humans
2.
Ann Allergy Asthma Immunol ; 123(6): 550-557, 2019 12.
Article in English | MEDLINE | ID: mdl-31494234

ABSTRACT

OBJECTIVE: To review the state of omics science specific to asthma and allergic diseases and discuss the current and potential applicability of omics in clinical disease prediction, treatment, and management. DATA SOURCES: Studies and reviews focused on the use of omics technologies in asthma and allergic disease research and clinical management were identified using PubMed. STUDY SELECTIONS: Publications were included based on relevance, with emphasis placed on the most recent findings. RESULTS: Omics-based research is increasingly being used to differentiate asthma and allergic disease subtypes, identify biomarkers and pathological mediators, predict patient responsiveness to specific therapies, and monitor disease control. Although most studies have focused on genomics and transcriptomics approaches, increasing attention is being placed on omics technologies that assess the effect of environmental exposures on disease initiation and progression. Studies using omics data to identify biological targets and pathways involved in asthma and allergic disease pathogenesis have primarily focused on a specific omics subtype, providing only a partial view of the disease process. CONCLUSION: Although omics technologies have advanced our understanding of the molecular mechanisms underlying asthma and allergic disease pathology, omics testing for these diseases are not standard of care at this point. Several important factors need to be addressed before these technologies can be used effectively in clinical practice. Use of clinical decision support systems and integration of these systems within electronic medical records will become increasingly important as omics technologies become more widely used in the clinical setting.


Subject(s)
Computational Biology , Hypersensitivity , Environmental Exposure , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Hypersensitivity/microbiology , Hypersensitivity/therapy
3.
Oncologist ; 24(10): 1287-1290, 2019 10.
Article in English | MEDLINE | ID: mdl-31366725

ABSTRACT

Drawing on discussions at a workshop hosted by the National Cancer Policy Forum, current challenges in pathology are reviewed and practical steps to facilitate high­quality cancer diagnosis and care through improved patient access to expertise in oncologic pathology are highlighted.


Subject(s)
Medical Oncology/methods , Neoplasms/diagnosis , Neoplasms/therapy , Quality of Health Care/standards , Humans
4.
Am J Clin Pathol ; 146(5): 585-593, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27769956

ABSTRACT

OBJECTIVES: To determine the effect of iterative refinement of standard ordering protocols on test utilization and results for bone marrow biopsy specimens. METHODS: Eighteen months of test utilization and result data were used to revise the protocols that determine cytogenetic and molecular test selection on bone marrow specimens and then compared with data obtained following protocol revision. RESULTS: Revision of protocols resulted in reduction in total tests and associated charges, due to a decrease in tests both concordant and discordant with the protocols. These reductions only occurred in diseases for which revisions were made and were limited to cases in which reflex testing was performed. There was an increase in the fraction of positive tests, which was also limited to reflex testing. CONCLUSIONS: Data-driven iterative revision of protocols further improves test utilization and performance, while reducing cost. Analysis of testing data can be used to continuously improve test ordering decisions.


Subject(s)
Bone Marrow Neoplasms/diagnosis , Bone Marrow/pathology , Evidence-Based Medicine , Practice Guidelines as Topic , Biopsy , Bone Marrow Cells/pathology , Bone Marrow Neoplasms/genetics , Bone Marrow Neoplasms/pathology , Costs and Cost Analysis , Cytogenetics/economics , Cytogenetics/statistics & numerical data , Hematology , Humans , Pathology , Pathology, Molecular/economics , Pathology, Molecular/statistics & numerical data
5.
Arch Pathol Lab Med ; 139(8): 989-1008, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25436904

ABSTRACT

CONTEXT: Genomic sequencing for cancer is offered by commercial for-profit laboratories, independent laboratory networks, and laboratories in academic medical centers and integrated health networks. The variability among the tests has created a complex, confusing environment. OBJECTIVE: To address the complexity, the Personalized Health Care (PHC) Committee of the College of American Pathologists proposed the development of a cancer genomics resource list (CGRL). The goal of this resource was to assist the laboratory pathology and clinical oncology communities. DESIGN: The PHC Committee established a working group in 2012 to address this goal. The group consisted of site-specific experts in cancer genetic sequencing. The group identified current next-generation sequencing (NGS)-based cancer tests and compiled them into a usable resource. The genes were annotated by the working group. The annotation process drew on published knowledge, including public databases and the medical literature. RESULTS: The compiled list includes NGS panels offered by 19 laboratories or vendors, accompanied by annotations. The list has 611 different genes for which NGS-based mutation testing is offered. Surprisingly, of these 611 genes, 0 genes were listed in every panel, 43 genes were listed in 4 panels, and 54 genes were listed in 3 panels. In addition, tests for 393 genes were offered by only 1 or 2 institutions. Table 1 provides an example of gene mutations offered for breast cancer genomic testing with the annotation as it appears in the CGRL 2014. CONCLUSIONS: The final product, referred to as the Cancer Genomics Resource List 2014, is available as supplemental digital content.


Subject(s)
Databases, Factual , High-Throughput Nucleotide Sequencing , Neoplasms/genetics , Pathology, Molecular , Humans , Pathology, Molecular/standards
6.
PLoS One ; 9(11): e114035, 2014.
Article in English | MEDLINE | ID: mdl-25415203

ABSTRACT

OBJECTIVE: Platelets express the α2ß1 integrin and the glycoprotein VI (GPVI)/FcRγ complex, both collagen receptors. Understanding platelet-collagen receptor function has been enhanced through use of genetically modified mouse models. Previous studies of GPVI/FcRγ-mediated collagen-induced platelet activation were perfomed with mice in which the FcRγ subunit was genetically deleted (FcRγ-/-) or the complex was depleted. The development of α2ß1-/- and GPVI-/- mice permits side-by-side comparison to address contributions of these collagen receptors in vivo and in vitro. APPROACH AND RESULTS: To understand the different roles played by the α2ß1 integrin, the GPVI receptor or FcRγ subunit in collagen-stimulated hemostasis and thrombosis, we compared α2ß1-/-, FcRγ-/-, and GPVI-/- mice in models of endothelial injury and intravascular thrombosis in vivo and their platelets in collagen-stimulated activation in vitro. We demonstrate that both the α2ß1 integrin and the GPVI receptor, but not the FcRγ subunit influence carotid artery occlusion in vivo. In contrast, the GPVI receptor and the FcRγ chain, but not the α2ß1 integrin, play similar roles in intravascular thrombosis in response to soluble Type I collagen. FcRγ-/- platelets showed less attenuation of tyrosine phosphorylation of several proteins including RhoGDI when compared to GPVI-/- and wild type platelets. The difference between FcRγ-/- and GPVI-/- platelet phosphotyrosine levels correlated with the in vivo thrombosis findings. CONCLUSION: Our data demonstrate that genetic deletion of GPVI receptor, FcRγ chain, or the α2ß1 integrin changes the thrombotic potentials of these platelets to collagen dependent on the stimulus mechanism. The data suggest that the FcRγ chain may provide a dominant negative effect through modulating signaling pathways in platelets involving several tyrosine phosphorylated proteins such as RhoGDI. In addition, these findings suggest a more complex signaling network downstream of the platelet collagen receptors than previously appreciated.


Subject(s)
Blood Platelets/metabolism , Collagen/pharmacology , Integrin alpha2beta1/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, IgG/metabolism , Thrombosis/metabolism , Animals , Collagen/adverse effects , Disease Models, Animal , Integrin alpha2beta1/genetics , Mice , Mice, Knockout , Platelet Activation/drug effects , Platelet Activation/genetics , Platelet Membrane Glycoproteins/genetics , Rats , Receptors, IgG/genetics , Thrombosis/chemically induced , Thrombosis/genetics
7.
Adv Exp Med Biol ; 819: 41-60, 2014.
Article in English | MEDLINE | ID: mdl-25023166

ABSTRACT

The α2ß1 integrin, also known as VLA-2, GPIa-IIa, CD49b, was first identified as an extracellular matrix receptor for collagens and/or laminins [55, 56]. It is now recognized that the α2ß1 integrin serves as a receptor for many matrix and nonmatrix molecules [35, 79, 128]. Extensive analyses have clearly elucidated the α2 I domain structural motifs required for ligand binding, and also defined distinct conformations that lead to inactive, partially active or highly active ligand binding [3, 37, 66, 123, 136, 137, 140]. The mechanisms by which the α2ß1 integrin plays a critical role in platelet function and homeostasis have been carefully defined via in vitro and in vivo experiments [76, 104, 117, 125]. Genetic and epidemiologic studies have confirmed human physiology and disease states mediated by this receptor in immunity, cancer, and development [6, 20, 21, 32, 43, 90]. The role of the α2ß1 integrin in these multiple complex biologic processes will be discussed in the chapter.


Subject(s)
Integrin alpha2beta1/physiology , Animals , Hemostasis , Humans , Immunity, Innate , Integrin alpha2beta1/chemistry , Neovascularization, Physiologic , Protein Structure, Tertiary , Signal Transduction , Wound Healing
8.
Invest Ophthalmol Vis Sci ; 55(7): 4338-47, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24917135

ABSTRACT

PURPOSE: The α2ß1 integrin plays an important but complex role in angiogenesis and vasculopathies. Published GWAS studies established a correlation between genetic polymorphisms of the α2ß1 integrin gene and incidence of diabetic retinopathy. Recent studies indicated that α2-null mice demonstrate superior vascularization in both the wound and diabetic microenvironments. The goal of this study was to determine whether the vasculoprotective effects of α2-integrin deficiency extended to the retina, using the oxygen-induced retinopathy (OIR) model for retinopathy of prematurity (ROP). METHODS: In the OIR model, wild-type (WT) and α2-null mice were exposed to 75% oxygen for 5 days (postnatal day [P] 7 to P12) and subsequently returned to room air for 6 days (P12-P18). Retinas were collected at postnatal day 7, day 13, and day 18 and examined via hematoxylin and eosin and Lectin staining. Retinas were analyzed for retinal vascular area, neovascularization, VEGF expression, and Müller cell activation. Primary Müller cell cultures from WT and α2-null mice were isolated and analyzed for hypoxia-induced VEGF-A expression. RESULTS: In the retina, the α2ß1 integrin was minimally expressed in endothelial cells and strongly expressed in activated Müller cells. Isolated α2-null primary Müller cells demonstrated decreased hypoxia-induced VEGF-A expression. In the OIR model, α2-null mice displayed reduced hyperoxia-induced vaso-attenuation, reduced pathological retinal neovascularization, and decreased VEGF expression as compared to WT counterparts. CONCLUSIONS: Our data suggest that the α2ß1 integrin contributes to the pathogenesis of retinopathy. We describe a newly identified role for α2ß1 integrin in mediating hypoxia-induced Müller cell VEGF-A production.


Subject(s)
Ependymoglial Cells/metabolism , Integrin alpha2beta1/genetics , RNA/genetics , Retinopathy of Prematurity/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Ependymoglial Cells/pathology , Gene Expression Regulation , Integrin alpha2beta1/biosynthesis , Integrin alpha2beta1/deficiency , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Oxygen/toxicity , Polymerase Chain Reaction , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
9.
Sci Signal ; 7(318): pe9, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24667375

ABSTRACT

Integrin switching plays a critical role in the progression to metastatic disease, but the mechanism by which it contributes remains poorly understood. In the 11 February 2014 issue of Science Signaling, Truong et al. identified a transforming growth factor-ß-mediated, prometastatic switch that is activated by ß1 integrin inhibition in triple-negative breast cancers (TNBCs). Their work provides insight into the complex signaling changes that arise from integrin switching. Further characterization of ß-integrin switching will require elucidation of the distribution of specific α-ß integrin heterodimers and the role of ligand binding. Identifying the nature of the molecular interactions and the influence of a specific oncogenic context, including the status of driver mutations such as those in Myc and p53, will define the next phase in integrin cancer biology.


Subject(s)
Extracellular Matrix/metabolism , Integrin beta1/metabolism , Lung Neoplasms/secondary , Neoplasm Metastasis/physiopathology , Signal Transduction/physiology , Triple Negative Breast Neoplasms/physiopathology , Animals , Humans
10.
Leuk Lymphoma ; 55(3): 601-5, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23876099

ABSTRACT

Fluorescence in situ hybridization for abnormalities common to myelodysplastic syndrome (MDS FISH) is often used with traditional karyotype in the diagnosis and monitoring of myeloid neoplasms. However, its value in these roles has been questioned. To evaluate its utility, we compared MDS FISH results with karyotype in 544 bone marrow specimens obtained for diagnosis (180 cases) or follow-up (364 cases) of myeloid neoplasia. We found excellent concordance between FISH and karyotype, such that FISH is rarely abnormal (1.7% at diagnosis and 3.0% at follow-up) in cases with normal karyotype. Even in the rare discordant cases, the abnormal FISH has little or no clinical value. Thus, we propose that this test should be limited to cases with inadequate karyotype only. Such guidelines could result in significant cost savings with no impact on patient diagnosis.


Subject(s)
Chromosome Aberrations , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , In Situ Hybridization, Fluorescence , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Bone Marrow/pathology , Disease Progression , Female , Follow-Up Studies , Hematologic Neoplasms/diagnosis , Humans , Karyotype , Karyotyping , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Young Adult
11.
Am J Clin Pathol ; 140(5): 643-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24124142

ABSTRACT

OBJECTIVES: To address the overuse of testing that complicates patient care, diminishes quality, and increases costs by implementing the diagnostic management team, a multidisciplinary system for the development and deployment of diagnostic testing guidelines for hematologic malignancies. METHODS: The team created evidence-based standard ordering protocols (SOPs) for cytogenetic and molecular testing that were applied by pathologists to bone marrow biopsy specimens on adult patients. Testing on 780 biopsy specimens performed during the six months before SOP implementation was compared with 1,806 biopsy specimens performed during the subsequent 12 months. RESULTS: After implementation, there were significant decreases in tests discordant with SOPs, omitted tests, and the estimated cost of testing to payers. The fraction of positive tests increased. Clinicians reported acceptance of the new procedures and perceived time savings. CONCLUSIONS: This process is a model for optimizing complex and personalized diagnostic testing.


Subject(s)
Bone Marrow Cells/pathology , Bone Marrow/pathology , Hematologic Neoplasms/diagnosis , Patient Care Team/organization & administration , Practice Guidelines as Topic , Precision Medicine/methods , Clinical Protocols , Evidence-Based Medicine , Humans , Patient Care Team/standards , Precision Medicine/standards , Reproducibility of Results
12.
J Innate Immun ; 3(5): 459-70, 2011.
Article in English | MEDLINE | ID: mdl-21502744

ABSTRACT

Mast cells, critical mediators of inflammation and anaphylaxis, are poised as one of the first lines of defense against external assault. Mast cells release several classes of preformed and de novo synthesized mediators. Cross-linking of the high-affinity FcεRI results in degranulation and the release of preformed, proinflammatory mediators including histamine and serotonin. We previously demonstrated that mast cell activation by Listeria monocytogenes requires the α2ß1 integrin for rapid IL-6 secretion both in vivo and in vitro. However, the mechanism of IL-6 release is unknown. Here, we demonstrate the Listeria- and α2ß1 integrin-mediated mast cell release of preformed IL-6 without the concomitant release of histamine or ß-hexosaminidase. α2ß1 integrin-dependent mast cell activation and IL-6 release is calcium independent. In contrast, IgE cross-linking-mediated degranulation is calcium dependent and does not result in IL-6 release, demonstrating that distinct stimuli result in the release of specific mediator pools. These studies demonstrate that IL-6 is presynthesized and stored in connective tissue mast cells and can be released from mast cells in response to distinct, α2ß1 integrin-dependent stimulation, providing the host with a specific innate immune response without stimulating an allergic reaction.


Subject(s)
Integrin alpha2beta1/metabolism , Interleukin-6 , Listeria monocytogenes/immunology , Mast Cells/metabolism , Animals , Antigens, Bacterial/immunology , Calcium Signaling/genetics , Calcium Signaling/immunology , Cell Degranulation , Cells, Cultured , Connective Tissue/pathology , Humans , Immunity, Innate/genetics , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Integrin alpha2beta1/genetics , Integrin alpha2beta1/immunology , Interleukin-6/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Aggregation
13.
Diabetes ; 60(2): 416-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21270253

ABSTRACT

OBJECTIVE: The hypothesis that high-fat (HF) feeding causes skeletal muscle extracellular matrix (ECM) remodeling in C57BL/6J mice and that this remodeling contributes to diet-induced muscle insulin resistance (IR) through the collagen receptor integrin α(2)ß(1) was tested. RESEARCH DESIGN AND METHODS: The association between IR and ECM remodeling was studied in mice fed chow or HF diet. Specific genetic and pharmacological murine models were used to study effects of HF feeding on ECM in the absence of IR. The role of ECM-integrin interaction in IR was studied using hyperinsulinemic-euglycemic clamps on integrin α(2)ß(1)-null (itga2(-/-)), integrin α(1)ß(1)-null (itga1(-/-)), and wild-type littermate mice fed chow or HF. Integrin α(2)ß(1) and integrin α(1)ß(1) signaling pathways have opposing actions. RESULTS: HF-fed mice had IR and increased muscle collagen (Col) III and ColIV protein; the former was associated with increased transcript, whereas the latter was associated with reduced matrix metalloproteinase 9 activity. Rescue of muscle IR by genetic muscle-specific mitochondria-targeted catalase overexpression or by the phosphodiesterase 5a inhibitor, sildenafil, reversed HF feeding effects on ECM remodeling and increased muscle vascularity. Collagen remained elevated in HF-fed itga2(-/-) mice. Nevertheless, muscle insulin action and vascularity were increased. Muscle IR in HF-fed itga1(-/-) mice was unchanged. Insulin sensitivity in chow-fed itga1(-/-) and itga2(-/-) mice was not different from wild-type littermates. CONCLUSIONS: ECM collagen expansion is tightly associated with muscle IR. Studies with itga2(-/-) mice provide mechanistic insight for this association by showing that the link between muscle IR and increased collagen can be uncoupled by the absence of collagen-integrin α(2)ß(1) interaction.


Subject(s)
Extracellular Matrix/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Integrin alpha2beta1/metabolism , Muscle, Skeletal/metabolism , Analysis of Variance , Animals , Blotting, Western , Collagen Type III/metabolism , Collagen Type V/metabolism , Diet , Extracellular Matrix/drug effects , Glucose Clamp Technique , Immunohistochemistry , Integrin alpha2beta1/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Purines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sildenafil Citrate , Sulfones/pharmacology
14.
J Clin Invest ; 121(1): 226-37, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21135504

ABSTRACT

Integrins regulate cell-cell and cell-matrix adhesion and thereby play critical roles in tumor progression and metastasis. Although work in preclinical models suggests that ß1 integrins may stimulate metastasis of a number of cancers, expression of the ß1 subunit alone has not been shown to be a useful prognostic indicator in human cancer patients. Here we have demonstrated that the α2ß1 integrin suppresses metastasis in a clinically relevant spontaneous mouse model of breast cancer. These data are consistent with previous studies indicating high expression of α2ß1 integrin in normal breast epithelium and loss of α2ß1 in poorly differentiated breast cancer. They are also consistent with our systematic analysis of microarray databases of human breast and prostate cancer, which revealed that decreased expression of the gene encoding α2 integrin, but not genes encoding α1, α3, or ß1 integrin, was predictive of metastatic dissemination and decreased survival. The predictive value of α2 expression persisted within both good-risk and poor-risk cohorts defined by estrogen receptor and lymph node status. Thus, the α2ß1 integrin functionally inhibits breast tumor metastasis, and α2 expression may serve as an important biomarker of metastatic potential and patient survival.


Subject(s)
Breast Neoplasms/physiopathology , Integrin alpha2beta1/physiology , Mammary Neoplasms, Experimental/physiopathology , Tumor Suppressor Proteins/physiology , Animals , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Primers/genetics , Female , Genes, erbB-2 , Humans , In Vitro Techniques , Integrin alpha2beta1/deficiency , Integrin alpha2beta1/genetics , Kaplan-Meier Estimate , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , Neoplasm Metastasis/prevention & control , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Tumor Stem Cell Assay , Tumor Suppressor Proteins/genetics
15.
J Biol Chem ; 284(50): 34640-7, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19815553

ABSTRACT

Thrombin and fibrillar collagen are potent activators of platelets at sites of vascular injury. Both agonists cause platelet shape change, granule secretion, and aggregation to form the primary hemostatic plug. Human platelets express two thrombin receptors, protease-activated receptors 1 and 4 (PAR1 and PAR4) and two collagen receptors, the alpha(2)beta(1) integrin (alpha(2)beta(1)) and the glycoprotein VI (GPVI)/FcRgamma chain complex. Although these receptors and their signaling mechanisms have been intensely studied, it is not known whether and how these receptors cooperate in the hemostatic function of platelets. This study examined cooperation between the thrombin and collagen receptors in platelet adhesion by utilizing a collagen-related peptide (alpha2-CRP) containing the alpha(2)beta(1)-specific binding motif, GFOGER, in conjunction with PAR-activating peptides. We demonstrate that platelet adhesion to alpha2-CRP is substantially enhanced by suboptimal PAR activation (agonist concentrations that do not stimulate platelet aggregation) using the PAR4 agonist peptide and thrombin. The enhanced adhesion induced by suboptimal PAR4 activation was alpha(2)beta(1)-dependent and GPVI/FcRgamma-independent as revealed in experiments with alpha(2)beta(1)- or FcRgamma-deficient mouse platelets. We further show that suboptimal activation of other platelet G(q)-linked G protein-coupled receptors (GPCRs) produces enhanced platelet adhesion to alpha2-CRP. The enhanced alpha(2)beta(1)-mediated platelet adhesion is controlled by phospholipase C (PLC), but is not dependent on granule secretion, activation of alpha(IIb)beta(3) integrin, or on phosphoinositol-3 kinase (PI3K) activity. In conclusion, we demonstrate a platelet priming mechanism initiated by suboptimal activation of PAR4 or other platelet G(q)-linked GPCRs through a PLC-dependent signaling cascade that promotes enhanced alpha(2)beta(1) binding to collagens containing GFOGER sites.


Subject(s)
Blood Platelets/metabolism , Collagen/metabolism , Integrin alpha2beta1/metabolism , Platelet Adhesiveness/physiology , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Mice , Mice, Inbred C57BL , Peptides/genetics , Peptides/metabolism , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Rats , Receptors, Collagen/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Signal Transduction/physiology , Thrombin/metabolism , Type C Phospholipases/metabolism
16.
Blood ; 111(7): 3562-70, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18198349

ABSTRACT

Data from several investigators suggest that the alpha2beta1 integrin, a receptor for collagens, laminins, decorin, E-cadherin, matrix metalloproteinase-1, endorepellin, and several viruses, is required for innate immunity and regulation of autoimmune/allergic disorders. We demonstrated that the innate immune response to Listeria monocytogenes required alpha2beta1 integrin expression by peritoneal mast cells (PMCs). Ligation of the alpha2beta1 integrin by C1q contained in immune complexes comprised of Listeria and antibody was required for PMC activation in vitro and in vivo. However, ligation of the alpha2beta1 integrin alone was insufficient to activate cytokine secretion, suggesting that one or more additional signals emanating from a coreceptor were required for PMC activation. Here, we demonstrate that C1q, but neither other complement proteins nor FcRgamma, is required for early innate immune response to Listeria. The binding of Listeria's Internalin B (InlB) to hepatocyte growth factor receptor (HGF-R)/c-met provides the costimulatory function required for PMC activation. Either HGF or Listeria InlB bound to c-met and either C1q or type I collagen bound to alpha2beta1 integrin stimulates PMC activation. These findings suggest that crosstalk between c-met and the alpha2beta1 integrin may contribute to mast-cell activation in autoimmune and inflammatory disorders.


Subject(s)
Immunity, Innate/physiology , Integrin alpha2/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Mast Cells/immunology , Proto-Oncogene Proteins c-met/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Complement C1q/genetics , Complement C1q/immunology , Cytokines/genetics , Cytokines/immunology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Immunologic Capping/immunology , Inflammation/genetics , Inflammation/immunology , Integrin alpha2/genetics , Leukocytes, Mononuclear/immunology , Listeriosis/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Protein Binding/immunology , Proto-Oncogene Proteins c-met/genetics , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, Virus/genetics , Receptors, Virus/immunology
17.
Blood ; 111(4): 1980-8, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18042800

ABSTRACT

To define the role of the alpha2beta1 integrin in pathologic angiogenesis, we investigated tumor-associated growth and angiogenesis in wild-type and alpha2-null mice. Our findings reveal that the alpha2beta1 integrin plays an important role in angiogenesis via regulation of VEGFR1 expression. When challenged with B16F10 melanoma cells, mice lacking alpha2beta1 integrin ex-pression exhibit increased tumor angiogenesis associated with up-regulated VEGFR1 expression. In contrast, there was no alpha2beta1 integrin-dependent difference in the angiogenic response to Lewis lung carcinoma (LLC) cells. Interestingly, whereas B16F10 cells secrete high levels of placental growth factor (PLGF), LLC cells produce high levels of VEGF, but low levels of PLGF. The alpha2beta1 integrin-dependent difference in angiogenesis was restored to LLC cells by expression of PLGF, strongly suggesting that the angiogenic phenotype and tumor growth in the alpha2-null host is dependent on specific interactions between the tumor cell and the genetically defined integrin repertoire of the host microenvironment. Thus integrin alpha2-null mice represent an example of genetic alterations of "the soil" determining response to the "seed."


Subject(s)
Integrin alpha2beta1/deficiency , Integrin alpha2beta1/genetics , Melanoma/blood supply , Neovascularization, Pathologic/genetics , Animals , Gene Expression Regulation, Neoplastic , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Vascular Endothelial Growth Factor Receptor-1/genetics
18.
Adv Exp Med Biol ; 608: 87-100, 2007.
Article in English | MEDLINE | ID: mdl-17993234

ABSTRACT

The integrin family of extracellular matrix receptors plays an important role in normal development, epithelial morphogenesis, angiogenesis, and in tumor progression and metastasis. Integrins cooperate with growth factor receptors to control many cellular functions including proliferation and cell survival. Integrin-mediated adhesion regulates many of the cell cycle checkpoints including activation of cyclin D/cdk4/6 complexes, expression of cyclin D genes, and regulation of levels of cyclin-dependent kinase inhibitors. In addition, integrin-mediated cell adhesion regulates apoptosis by modulating the activity of both the mitochondrial pathway and the death receptor pathways. Therefore, integrin-mediated adhesion modulates the decision of life or death. A role for tumor-matrix interactions in the acquisition of drug resistance has been reported for many cancers including breast cancer. Recent evidence suggests that integrin-mediated adhesion to the ECM may undermine the response of tumors to chemotherapeutic agents. Integrins have been shown to be readily accessible drug targets and are therefore attractive potential targets for combined modality chemotherapy.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Integrins/physiology , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Humans , Integrins/genetics
19.
Immunobiology ; 212(4-5): 343-53, 2007.
Article in English | MEDLINE | ID: mdl-17544819

ABSTRACT

Our laboratory focuses on the alpha2beta1 integrin, a receptor for a number of matrix and non-matrix ligands, including collagens, laminins, decorin, E-cadherin, matrix metalloproteinase-1 (MMP-1), endorepellin, and several viruses. The alpha2beta1 integrin is expressed on numerous different cell types, including epithelial cells, endothelial cells, fibroblasts, and hematopoietic elements, including platelets and specific subsets of leukocytes. Although alpha2beta1 integrin expression is widespread, it is not ubiquitous. Rather, it is expressed in a differentiation-dependent and activation-dependent manner. Interactions between the alpha2beta1 integrin and extracellular matrix ligands have been implicated in important biological processes including inflammation and immunity. Studies from a number of laboratories have demonstrated a role for the alpha2beta1 integrin during the immune response. Our laboratory generated an alpha2beta1 integrin-deficient mouse to define the role of the alpha2beta1 integrin in vivo. Our studies demonstrated that the alpha2-null mice have a profound defect in the innate immune response. We have recently reported the identification of a novel family of ligands for the alpha2beta1 integrin, which include C1q and the collectins. The goal of this article is to review the important role that the interaction between the alpha2beta1 integrin and C1q plays in the innate immune response. The identification of C1q and the collectins as ligands for the alpha2beta1 integrin suggests that the integrin may play important roles in a number of immunological responses.


Subject(s)
Collectins/metabolism , Integrin alpha2beta1/metabolism , Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , Animals , Humans , Immunity, Innate/immunology , Integrin alpha2beta1/deficiency , Integrin alpha2beta1/genetics , Interleukin-6/metabolism , Mast Cells/immunology , Mast Cells/metabolism
20.
J Invest Dermatol ; 127(2): 455-66, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17068473

ABSTRACT

The alpha2beta1 integrin, a collagen/laminin receptor, is expressed at high level in the basal cell layer of the epidermis. To define the role of the alpha2beta1 integrin in wound healing, wound repair was extensively evaluated in wild-type and alpha2-null mice in vivo. In addition, the impact of alpha2beta1 integrin-deficiency on the function of primary murine keratinocytes in vitro was analyzed. Our in vivo findings demonstrate that genetic deletion of the alpha2beta1 integrin does not significantly alter the rate of re-epithelialization, collagen deposition, or tensile strength during wound closure in mice. In marked contrast to the observed similarities in wound healing, deletion of the alpha2beta1 integrin resulted in a dramatic increase in neoangiogenesis in the wound microenvironment. In contrast to in vivo studies, primary keratinocytes from alpha2-null mice adhered poorly and displayed impaired migration on type I collagen in vitro. We demonstrate that alpha2beta1 integrin-ligation negatively regulates expression of genes including matrix metalloproteinases both in vivo and in vitro. Furthermore, the changes in gene expression could potentially account for relatively normal wound healing in the alpha2-deficient mouse and our recent observation that suggests an antiangiogenic role for the alpha2beta1 integrin in vivo.


Subject(s)
Integrin alpha2beta1/deficiency , Keratinocytes , Matrix Metalloproteinases/metabolism , Skin/injuries , Wound Healing , Wounds, Penetrating/physiopathology , Animals , Cell Adhesion , Cell Movement , Gene Expression , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Wound Healing/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...