Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 134(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34704600

ABSTRACT

Osteoclasts form special integrin-mediated adhesion structures called sealing zones that enable them to adhere to and resorb bone. Sealing zones consist of densely packed podosomes tightly interconnected by actin fibers. Their formation requires the presence of the hematopoietic integrin regulator kindlin-3 (also known as Fermt3). In this study, we investigated osteoclasts and their adhesion structures in kindlin-3 hypomorphic mice expressing only 5-10% of the kindlin-3 level of wild-type mice. Low kindlin-3 expression reduces integrin activity, results in impaired osteoclast adhesion and signaling, and delays cell spreading. Despite these defects, in vitro-generated kindlin-3-hypomorphic osteoclast-like cells arrange their podosomes into adhesion patches and belts, but their podosome and actin organization is abnormal. Remarkably, kindlin-3-hypomorphic osteoclasts form sealing zones when cultured on calcified matrix in vitro and on bone surface in vivo. However, functional assays, immunohistochemical staining and electron micrographs of bone sections showed that they fail to seal the resorption lacunae properly, which is required for secreted proteinases to digest bone matrix. This results in mild osteopetrosis. Our study reveals a new, hitherto understudied function of kindlin-3 as an essential organizer of integrin-mediated adhesion structures, such as sealing zones.


Subject(s)
Cytoskeletal Proteins , Osteoclasts , Osteopetrosis , Animals , Bone Matrix , Bone and Bones , Cytoskeletal Proteins/genetics , Integrins , Mice , Osteopetrosis/genetics
2.
Bone ; 153: 116150, 2021 12.
Article in English | MEDLINE | ID: mdl-34400384

ABSTRACT

Osteoblasts are not only responsible for bone formation. They also support hematopoiesis. This requires responding to cues originating from several signaling pathways, a task performed by Rho GTPases. We therefore examined several transgenic mouse models and used inhibitors of Cdc42 in vitro. Deletion of Cdc42 in vivo using the Osterix promoter suppressed osteoblast function, while its deletion in differentiating osteoblasts using the Collagen-α1(I) promoter decreased osteoblast numbers. In both cases, bone mineral density diminished confirming the importance of Cdc42. Evaluation of hematopoiesis revealed that deletion of Cdc42 using the Osterix, but not the Collagen-α1(I) promoter increased the common myeloid progenitors (CMPs) in the bone marrow as well as the erythrocytes and the thrombocytes/platelets in peripheral blood. Causality between Cdc42 loss in early osteoblasts and increased myelopoiesis was confirmed in vitro. Work in vitro supported the conclusion that interleukin-4 mediated the increase in myelopoiesis. Thus, Cdc42 is required for healthy bone through regulation of bone formation in Osterix-expressing osteoblasts and the number of osteoblasts in differentiating osteoblasts. In addition, its expression in early osteoblasts/stromal cells modulates myelopoiesis. This highlights the importance of osteoblasts in regulating hematopoiesis.


Subject(s)
Osteoblasts , Osteogenesis , Animals , Bone and Bones , Cell Differentiation , Cell Lineage/genetics , Mice , Osteogenesis/genetics
3.
Neoplasia ; 23(9): 837-850, 2021 09.
Article in English | MEDLINE | ID: mdl-34298233

ABSTRACT

Understanding how the extracellular matrix affects cancer development constitutes an emerging research field. Fibronectin and collagen are two intriguing matrix components found in cancer. Large concentrations of fibronectin or collagen type I have been implicated in poor prognosis in patients. In a mouse model, we had shown that genetically decreasing circulating fibronectin resulted in smaller tumors. We therefore aimed to manipulate fibronectin pharmacologically and determine how cancer development is affected. Deletion of fibronectin in human breast cancer cells (MDA-MB-231) using shRNA (knockdown: Kd) improved survival and diminished tumor burden in a model of metastatic lesions and in a model of local growth. Based on these findings, it seemed reasonable to attempt to prevent fibronectin accumulation using a bacterial derived peptide called pUR4. Treatment with this peptide for 10 days in the breast cancer local growth model or for 5 days in a melanoma skin cancer model (B16) was associated with a significant suppression of cancer growth. Treatment aimed at inhibiting collagen type I accumulation without interfering with fibronectin could not affect any changes in vivo. In the absence of fibronectin, diminished cancer progression was due to inhibition of proliferation, even though changes in blood vessels were also detected. Decreased proliferation could be attributed to decreased ERK phosphorylation and diminished YAP expression. In summary, manipulating fibronectin diminishes cancer progression, mostly by suppressing cell proliferation. This suggests that matrix modulation could be used as an adjuvant to conventional therapy as long as a decrease in fibronectin is obtained.


Subject(s)
Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Neoplasms/metabolism , Neoplasms/prevention & control , Amino Acid Sequence , Animals , Fibronectins/genetics , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Nude , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Tumor Burden/drug effects , Tumor Burden/physiology , Xenograft Model Antitumor Assays/methods
4.
Cells ; 9(12)2020 12 18.
Article in English | MEDLINE | ID: mdl-33353083

ABSTRACT

Fibronectin is a ubiquitous extracellular matrix protein that is produced by many cell types in the bone marrow and distributed throughout it. Cells of the stem cell niche produce the various isoforms of this protein. Fibronectin not only provides the cells a scaffold to bind to, but it also modulates their behavior by binding to receptors on the adjacent hematopoietic stem cells and stromal cells. These receptors, which include integrins such as α4ß1, α9ß1, α4ß7, α5ß1, αvß3, Toll-like receptor-4 (TLR-4), and CD44, are found on the hematopoietic stem cell. Because the knockout of fibronectin is lethal during embryonal development and because fibronectin is produced by almost all cell types in mammals, the study of its role in hematopoiesis is difficult. Nevertheless, strong and direct evidence exists for its stimulation of myelopoiesis and thrombopoiesis using in vivo models. Other reviewed effects can be deduced from the study of fibronectin receptors, which showed their activation modifies the behavior of hematopoietic stem cells. Erythropoiesis was only stimulated under hemolytic stress, and mostly late stages of lymphocytic differentiation were modulated. Because fibronectin is ubiquitously expressed, these interactions in health and disease need to be taken into account whenever any molecule is evaluated in hematopoiesis.


Subject(s)
Fibronectins/physiology , Hematopoiesis , Receptors, Fibronectin/physiology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Erythropoiesis , Hematopoietic Stem Cells/cytology , Hemolysis , Humans , Hyaluronan Receptors/metabolism , Integrins/metabolism , Mice , Myelopoiesis , Stem Cell Niche , Stem Cells/cytology , Thrombopoiesis , Toll-Like Receptor 4/metabolism
5.
Int J Mol Sci ; 21(2)2020 01 08.
Article in English | MEDLINE | ID: mdl-31936261

ABSTRACT

The regulation of the differentiation of the bone-forming cells, the osteoblasts, is complex. Many signaling pathways converge on the master regulator of osteoblast differentiation Runx2. The role of molecules that integrate several signaling pathways such as the Rho GTPases need to be better understood. We, therefore, asked at which stage Rac1, one of the Rho GTPase, is needed for osteoblast differentiation and whether it is involved in two pathways, the anabolic response to parathyroid hormone and the stimulatory effect of fibronectin isoforms on integrins. Genetic deletion of Rac1 in preosteoblasts using the osterix promoter diminished osteoblast differentiation in vitro. This effect was however similar to the presence of the promoter by itself. We, therefore, applied a Rac1 inhibitor and confirmed a decrease in differentiation. In vivo, Rac1 deletion using the osterix promoter decreased bone mineral density as well as histomorphometric measures of osteoblast function. In contrast, deleting Rac1 in differentiating osteoblasts using the collagen α1(I) promoter had no effects. We then evaluated whether intermittent parathyroid hormone (PTH) was able to affect bone mineral density in the absence of Rac1 in preosteoblasts. The increase in bone mineral density was similar in control animals and in mice in which Rac1 was deleted using the osterix promoter. Furthermore, stimulation of integrin by integrin isoforms was able to enhance osteoblast differentiation, despite the deletion of Rac1. In summary, Rac1 in preosteoblasts is required for normal osteoblast function and bone density, but it is neither needed for PTH-mediated anabolic effects nor for integrin-mediated enhancement of differentiation.


Subject(s)
Bone Density/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Neuropeptides/genetics , Parathyroid Hormone/genetics , rac1 GTP-Binding Protein/genetics , Animals , Cell Differentiation/genetics , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Fibronectins/genetics , Humans , Integrins/genetics , Mice , Osteoblasts/metabolism , Osteogenesis/genetics , Signal Transduction/genetics , Sp7 Transcription Factor/genetics , rho GTP-Binding Proteins/genetics
6.
Cancer Res ; 78(1): 129-142, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29066511

ABSTRACT

Breast and prostate cancer cells home to the bone marrow, where they presumably hijack the hematopoietic stem cell niche. We characterize here the elusive premetastatic niche by examining the role of mesenchymal stromal cells (MSC) in cancer cell homing. Decreasing the number of MSC pharmacologically enhanced cancer cell homing to the bone marrow in mice. In contrast, increasing the number of these MSCs by various interventions including G-CSF administration diminished cancer cell homing. The MSC subpopulation that correlated best with cancer cells expressed stem, endothelial, and pericytic cell markers, suggesting these cells represent an undifferentiated component of the niche with vascular commitment. In humans, a MSC subpopulation carrying markers for endothelial and pericytic cells was lower in the presence of cytokeratin+ cells in bone marrow. Taken together, our data show that a subpopulation of MSC with both endothelial and pericytic cell surface markers suppresses the homing of cancer cells to the bone marrow. Similar to the presence of cytokeratin+ cells in the bone marrow, this MSC subpopulation could prove useful in determining the risk of metastatic disease, and its manipulation might offer a new possibility for diminishing bone metastasis formation.Significance: These findings establish an inverse relationship between a subpopulation of mesenchymal stromal cells and cancer cells in the bone marrow. Cancer Res; 78(1); 129-42. ©2017 AACR.


Subject(s)
Bone Marrow/pathology , Breast Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Prostatic Neoplasms/pathology , Animals , Bone Marrow/drug effects , Cell Line, Tumor , Diphosphonates/pharmacology , Female , Humans , Imidazoles/pharmacology , Male , Mice, Mutant Strains , Parathyroid Hormone/pharmacology , Prenylation , Stem Cell Niche , Xenograft Model Antitumor Assays , Zoledronic Acid
7.
J Biol Chem ; 292(19): 7745-7760, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28325836

ABSTRACT

Fibronectin is a multidomain protein secreted by various cell types. It forms a network of fibers within the extracellular matrix and impacts intracellular processes by binding to various molecules, primarily integrin receptors on the cells. Both the presence of several isoforms and the ability of the various domains and isoforms to bind to a variety of integrins result in a wide range of effects. In vivo findings suggest that fibronectin isoforms produced by the osteoblasts enhance their differentiation. Here we report that the isoform characterized by the presence of extradomain A activates α4ß1 integrin and augments osteoblast differentiation. In addition, the isoform containing extradomain B enhances the binding of fibronectin through the RGD sequence to ß3-containing integrin, resulting in increased mineralization by and differentiation of osteoblasts. Our study thus reveals novel functions for two fibronectin isoforms and the mediating receptors in osteoblast differentiation.


Subject(s)
Cell Differentiation , Fibronectins/metabolism , Integrin alpha4beta1/metabolism , Integrin alphaVbeta3/metabolism , Osteoblasts/cytology , 3T3 Cells , Animals , Animals, Newborn , Cell Adhesion , DNA, Complementary/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , Green Fluorescent Proteins/metabolism , Humans , Mice , Oligopeptides/metabolism , Osteoblasts/metabolism , Osteogenesis , Protein Binding , Protein Domains , Protein Isoforms , RNA, Small Interfering/metabolism , Signal Transduction
8.
Kidney Int ; 91(6): 1374-1385, 2017 06.
Article in English | MEDLINE | ID: mdl-28159318

ABSTRACT

Fibronectin is ubiquitously expressed in the extracellular matrix, and its accumulation in the glomerular mesangium in diabetic nephropathy is associated with deterioration of renal function in these patients. However, the exact role of fibronectin in the pathogenesis of diabetic nephropathy remains unknown. To clarify this, we administered fluorescent-labeled plasma fibronectin to wild-type mice and found it to accumulate in the mesangium. Using liver-specific conditional-knockout mice to decrease circulating fibronectin, we reduced circulating fibronectin by more than 90%. In streptozotocin-induced diabetes of these knockout mice, the pronounced fall in circulating fibronectin resulted in a decrease in mesangial expansion by 25% and a decline in albuminuria by 30% compared to diabetic control mice. Indeed, the amount of fibronectin in the kidney was reduced, as was the total amount of collagen. In vitro experiments confirmed that matrix accumulation of fibronectin was enhanced by increasing fibronectin only, glucose only, or the combination of both. Thus, circulating fibronectin contributes to mesangial expansion and exacerbation of albuminuria in a murine model of type 1 diabetes.


Subject(s)
Cell Proliferation , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/etiology , Fibronectins/blood , Mesangial Cells/metabolism , Albuminuria/blood , Albuminuria/etiology , Animals , Blood Glucose/metabolism , Cells, Cultured , Collagen/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetic Nephropathies/blood , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Fibronectins/deficiency , Fibronectins/genetics , Genetic Predisposition to Disease , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Liver/metabolism , Male , Mesangial Cells/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , Time Factors
9.
J Bone Miner Res ; 32(1): 70-81, 2017 01.
Article in English | MEDLINE | ID: mdl-27427791

ABSTRACT

Patients with cholestatic liver disease experience increased fracture risk. Higher circulating levels of a fibronectin isoform called oncofetal fibronectin (oFN) were detected in a subset of such patients. Administering this isoform to mice suppresses osteoblast differentiation and diminishes bone mineral density in vivo, suggesting it is responsible for bone loss in cholestatic liver disease. The aim of this study was to define the mechanism by which oFN affects osteoblast function and evaluate possible modifiers in experimental hepatic osteodystrophy. The fibronectin isoform oFN is characterized by the presence of various glycosylations. In line with this, adding oFN that underwent enzymatic O-deglycosylation to osteoblasts normalized nodule formation in vitro. Of three possible O-glycosylation sites in oFN, only a mutation at AA 33 of the variable region or binding of this glycosylated site with an antibody normalized osteoblast differentiation. Because the responsible site is located in the variable region of fibronectin, which binds to α4ß1 or α4ß7 integrins, these integrins were evaluated. We show that integrin α4ß1 mediates the inhibitory effect of oFN both in vitro as well as in vivo. In a hepatic osteodystrophy mouse model, we demonstrate that liver fibrosis is associated with increased circulating oFN and diminished BMD. In addition, trabecular bone loss induced by oFN injection or fibrosis induction could be prevented by either administering an antibody that binds to α4 integrin (PS/2) or the CS1 peptide, which contains a binding site for α4ß1 integrin. In summary, oFN inhibits osteoblast activity. This is because of an O-glycosylation in the variable region that results in decreased integrin-mediated signaling. This deleterious effect can be thwarted by binding α4ß1 integrin. Thus, we have characterized the defect and the receptor mediating bone loss in patients with hepatic osteodystrophy and evaluated possible therapeutic interventions in a murine model. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Diseases/complications , Bone Diseases/metabolism , Fibronectins/metabolism , Integrin alpha4beta1/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Fibronectins/administration & dosage , Glycosylation , Humans , Mice , Osteoblasts/metabolism , Peptides/metabolism
10.
PLoS Biol ; 14(9): e1002562, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27653627

ABSTRACT

Osteoblasts lining the inner surface of bone support hematopoietic stem cell differentiation by virtue of proximity to the bone marrow. The osteoblasts also modify their own differentiation by producing various isoforms of fibronectin (FN). Despite evidence for immune regulation by osteoblasts, there is limited knowledge of how osteoblasts modulate cells of the immune system. Here, we show that extra domain A (EDA)-FN produced by osteoblasts increases arginase production in myeloid-derived cells, and we identify α5ß1 as the mediating receptor. In different mouse models of cancer, osteoblasts or EDA-FN was found to up-regulate arginase-1 expression in myeloid-derived cells, resulting in increased cancer growth. This harmful effect can be reduced by interfering with the integrin α5ß1 receptor or inhibiting arginase. Conversely, in tissue injury, the expression of arginase-1 is normally beneficial as it dampens the immune response to allow wound healing. We show that EDA-FN protects against excessive fibrotic tissue formation in a liver fibrosis model. Our results establish an immune regulatory function for EDA-FN originating from the osteoblasts and identify new avenues for enhancing the immune reaction against cancer.

11.
J Mol Med (Berl) ; 94(5): 567-81, 2016 05.
Article in English | MEDLINE | ID: mdl-26637426

ABSTRACT

UNLABELLED: Plasma fibronectin is a circulating protein that facilitates phagocytosis by connecting bacteria to immune cells. A fibronectin isoform, which includes a sequence of 90 AA called extra-domain B (EDB), is synthesized de novo at the messenger RNA (mRNA) level in immune cells, but the reason for its expression remains elusive. We detected an 80-fold increase in EDB-containing fibronectin in the cerebrospinal fluid of patients with bacterial meningitis that was most pronounced in staphylococcal infections. A role for this isoform in phagocytosis was further suggested by enhanced EDB fibronectin release after internalization of Staphylococcus aureus in vitro. Using transgenic mouse models, we established that immune cell production of fibronectin contributes to phagocytosis, more so than circulating plasma fibronectin, and that accentuated release of EDB-containing fibronectin by immune cells improved phagocytosis. In line with this, administration of EDB fibronectin enhanced in vitro phagocytosis to a larger extent than plasma fibronectin. This enhancement was mediated by αvß3 integrin as shown using inhibitors or cells from ß3 integrin knockout mice. Thus, we identified both a novel function for EDB fibronectin in augmenting phagocytosis over circulating plasma fibronectin, as well as the mediating receptor. Our data also establish for the first time, a direct role for ß3 integrin in bacterial phagocytosis in mammals. KEY MESSAGES: • Fibronectin containing an extra domain called EDB is released in bacterial meningitis. • EDB-containing fibronectin enhances phagocytosis more than plasma fibronectin. • The enhancement is mediated by activation of αvß3 integrin in the presence of EDB.


Subject(s)
Fibronectins/metabolism , Phagocytosis , Protein Interaction Domains and Motifs , Actins/chemistry , Actins/metabolism , Animals , Case-Control Studies , Fibronectins/chemistry , Fibronectins/genetics , Gene Expression , Humans , Immunohistochemistry , Integrin beta3/metabolism , Meningitis, Bacterial/diagnosis , Meningitis, Bacterial/genetics , Meningitis, Bacterial/immunology , Meningitis, Bacterial/metabolism , Mice , Mice, Transgenic , Phagocytosis/immunology , Protein Binding , Protein Interaction Domains and Motifs/genetics , Protein Multimerization , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
J Hepatol ; 62(3): 625-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24946284

ABSTRACT

BACKGROUND & AIMS: Common pathogenic steps in liver fibrosis are inflammation and accumulation of extracellular matrix proteins including collagen, which lead to disruption of tissue microarchitecture and liver dysfunction. Adequate fibronectin fibril formation is required for collagen matrix deposition in several cell types in vitro. We therefore hypothesized that preventing fibronectin fibril assembly will result in decreased collagen matrix accumulation, and hence diminish liver injury associated with fibrosis. METHODS: In vitro studies on hepatic stellate cells and in vivo studies in mice were performed. RESULTS: In vitro studies on hepatic stellate cells confirmed that a fibronectin assembly inhibitor, pUR4 diminishes the amount of both fibronectin and collagen, accumulating in the extracellular matrix, without affecting their production. Induction of fibrosis using CCl4 or DMN was therefore combined with pUR4-treatment. pUR4 normalized the amount of fibrotic tissue that accumulated with injury, and improved liver function. Specifically, pUR4-treatment decreased collagen accumulation, without changing its mRNA expression. Most interestingly, we did not detect any changes in Kupffer cell numbers (F4/80+) or α-smooth muscle actin expressing hepatic stellate cell numbers. Further, there was no impact on TGF-ß or TNF-α. Thus, in line with the in vitro findings, decreased fibrosis is due to inhibition of matrix accumulation and not a direct effect on these cells. CONCLUSIONS: In summary, a peptide that blocks fibronectin deposition results in decreased collagen accumulation and improved liver function during liver fibrogenesis. Thus, fibronectin matrix modulation offers a therapeutic benefit in preclinical models of liver fibrosis.


Subject(s)
Fibronectins/antagonists & inhibitors , Liver Cirrhosis, Experimental/prevention & control , Animals , Collagen/metabolism , Disease Models, Animal , Disease Progression , Fibronectins/genetics , Fibronectins/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mutant Proteins/genetics , Mutant Proteins/metabolism , Peptides/pharmacology , Protein Multimerization/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
13.
World J Gastroenterol ; 20(28): 9427-38, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25071337

ABSTRACT

It is being increasingly recognized that patients with liver disease develop bone loss that can be severe enough to lead to atraumatic fractures and thus markedly diminish life quality and expectancy. The estimated prevalence for liver-related osteoporosis is between 20-420/100000 of the general population, and fractures between 60-880/100000. It should be kept in mind that up to 40% of patients with chronic liver disease may experience a fracture. The pathogenic mediators include fibronectin, insulin like growth factor-I, and various cytokines, but decreased vitamin D and/or treatment with corticosteroids contribute to worsening bone health. Despite the advances in bone biology that have shed some light on the pathogenesis of this bone loss, treatment options remain nonspecific and tightly linked to treatments of other forms of osteoporosis. Thus, treatment should include calcium and vitamin D supplementation in all patients with chronic liver disease. Therapy with bisphosphonates should be considered, especially in patients receiving corticosteroids. This review focuses on the prevalence of this entity as well as the evidence available with regard to the pathogenesis of bone loss in liver disease, the diagnostic steps required in all patients, and the therapeutic options available.


Subject(s)
Bone and Bones/metabolism , Liver Diseases/epidemiology , Liver/metabolism , Osteoporosis/epidemiology , Osteoporotic Fractures/epidemiology , Animals , Bone Density Conservation Agents/therapeutic use , Bone Remodeling , Bone and Bones/pathology , Calcium/therapeutic use , Dietary Supplements , Humans , Liver Diseases/diagnosis , Liver Diseases/metabolism , Liver Diseases/therapy , Osteoporosis/diagnosis , Osteoporosis/metabolism , Osteoporosis/therapy , Osteoporotic Fractures/diagnosis , Osteoporotic Fractures/metabolism , Osteoporotic Fractures/prevention & control , Predictive Value of Tests , Prevalence , Signal Transduction , Treatment Outcome , Vitamin D/therapeutic use
14.
PLoS One ; 9(4): e94922, 2014.
Article in English | MEDLINE | ID: mdl-24740307

ABSTRACT

Cancer is associated with increased fracture risk, due either to metastasis or associated osteoporosis. After a fracture, blood clots form. Because proteins of the coagulation cascade and activated platelets promote cancer development, a fracture in patients with cancer often raises the question whether it is a pathologic fracture or whether the fracture itself might promote the formation of metastatic lesions. We therefore examined whether blood clot formation results in increased metastasis in a murine model of experimental breast cancer metastasis. For this purpose, a clot was surgically induced in the bone marrow of the left tibia of immundeficient mice. Either one minute prior to or five minutes after clot induction, human cancer cells were introduced in the circulation by intracardiac injection. The number of cancer cells that homed to the intervention site was determined by quantitative real-time PCR and flow cytometry. Metastasis formation and longitudinal growth were evaluated by bioluminescence imaging. The number of cancer cells that homed to the intervention site after 24 hours was similar to the number of cells in the opposite tibia that did not undergo clot induction. This effect was confirmed using two more cancer cell lines. Furthermore, no difference in the number of macroscopic lesions or their growth could be detected. In the control group 72% developed a lesion in the left tibia. In the experimental groups with clot formation 79% and 65% developed lesions in the left tibia (p = ns when comparing each experimental group with the controls). Survival was similar too. In summary, the growth factors accumulating in a clot/hematoma are neither enough to promote cancer cell homing nor support growth in an experimental model of breast cancer bone metastasis. This suggests that blood clot formation, as occurs in traumatic fractures, surgical interventions, and bruises, does not increase the risk of metastasis formation.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Disease Models, Animal , Thrombosis/pathology , Animals , Biomarkers, Tumor/genetics , Blood Coagulation , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Hematoma/genetics , Hematoma/metabolism , Hematoma/pathology , Humans , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements , Mice , Mice, Knockout , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Tibia , Transplantation, Heterologous , Tumor Burden/genetics
15.
Clin Immunol ; 150(2): 149-56, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24412908

ABSTRACT

Chronic hepatitis C viral infection modulates complement. The aim of this study was to determine whether complement analysis predicts liver inflammation and fibrosis in patients with chronic hepatitis C. 50 chronic hepatitis C patients who underwent a liver biopsy were compared to 50 healthy controls and 35 patients with various liver diseases. Total plasma complement activity (CH50) in plasma was diminished in hepatitis C patients suggesting complement activation. This decrease correlated with increased necrosis (r = -0.24, p < 0.05), and patients with levels below the normal range had a higher METAVIR activity score reflecting enhanced inflammation. SC5b-9, a marker of complement activation, correlated with inflammation (r = 0.40, p < 0.05), activity (r = 0.42, p < 0.05), and fibrosis scores (r = 0.49, p < 0.05). Finally, the prevalence of C1q auto-antibodies was higher in hepatitis C patients, and their presence was associated with increased inflammation and seemed to affect fibrosis. We conclude that complement-induced liver inflammation contributes to fibrosis in patients with chronic hepatitis C.


Subject(s)
Complement Activation/immunology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Necrosis , Adult , Autoantibodies/immunology , Biomarkers/metabolism , Complement C1q/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Female , Fibronectins/metabolism , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/diagnosis , Humans , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/diagnosis , Male , Middle Aged , Prognosis
16.
Neoplasia ; 15(8): 925-38, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23908593

ABSTRACT

Fibronectin is ubiquitously expressed in the extracellular matrix, and experimental evidence has shown that it modulates blood vessel formation. The relative contribution of local and circulating fibronectin to blood vessel formation in vivo remains unknown despite evidence for unexpected roles of circulating fibronectin in various diseases. Using transgenic mouse models, we established that circulating fibronectin facilitates the growth of bone metastases by enhancing blood vessel formation and maturation. This effect is more relevant than that of fibronectin produced by endothelial cells and pericytes, which only exert a small additive effect on vessel maturation. Circulating fibronectin enhances its local production in tumors through a positive feedback loop and increases the amount of vascular endothelial growth factor (VEGF) retained in the matrix. Both fibronectin and VEGF then cooperate to stimulate blood vessel formation. Fibronectin content in the tumor correlates with the number of blood vessels and tumor growth in the mouse models. Consistent with these results, examination of three separate arrays from patients with breast and prostate cancers revealed that a high staining intensity for fibronectin in tumors is associated with increased mortality. These results establish that circulating fibronectin modulates blood vessel formation and tumor growth by modifying the amount of and the response to VEGF. Furthermore, determination of the fibronectin content can serve as a prognostic biomarker for breast and prostate cancers and possibly other cancers.


Subject(s)
Endothelial Cells/metabolism , Fibronectins/blood , Mammary Neoplasms, Experimental/blood , Neovascularization, Pathologic/blood , Animals , Apoptosis , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Tissue Array Analysis , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
PLoS One ; 6(11): e28181, 2011.
Article in English | MEDLINE | ID: mdl-22140539

ABSTRACT

Fibrotic tissue in the liver is mainly composed of collagen. Fibronectin, which is also present in fibrotic matrices, is required for collagen matrix assembly in vitro. It also modulates the amount of growth factors and their release from the matrix. We therefore examined the effects of the absence of fibronectin on the development of fibrosis in mice.Conditional deletion of fibronectin in the liver using the Mx promoter to drive cre expression resulted in increased collagen production and hence a more pronounced fibrosis in response to dimethylnitrosamine in mice. Exclusive deletion of fibronectin in hepatocytes or normalization of circulating fibronectin in Mx-cKO mice did not affect the development of fibrosis suggesting a role for fibronectin production by other liver cell types. The boosted fibrosis in fibronectin-deficient mice was associated with enhanced stellate cell activation and proliferation, elevated concentrations of active TGF-ß, and increased TGF-ß-mediated signaling.In vitro experiments revealed that collagen-type-I production by fibronectin-deficient hepatic stellate cells stimulated with TGF-ß was more pronounced, and was associated with augmented Smad3-mediated signaling. Interfering with TGF-ß signaling using SB431542 normalized collagen-type-I production in fibronectin-deficient hepatic stellate cells. Furthermore, precoating culture plates with fibronectin, but not collagen, or providing fibronectin fibrils unable to interact with RGD binding integrins via the RGD domain significantly diminished the amount of active TGF-ß in fibronectin-deficient stellate cells and normalized collagen-type-I production in response to TGF-ß stimulation. Thus, excessive stellate cell activation and production of collagen results from increased active TGF-ß and TGF-ß signaling in the absence of fibronectin.In conclusion, our data indicate that fibronectin controls the availability of active TGF-ß in the injured liver, which impacts the severity of the resulting fibrosis. We therefore propose a novel role for locally produced fibronectin in protecting the liver from an excessive TGF-ß-mediated response.


Subject(s)
Fibronectins/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/prevention & control , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Collagen/biosynthesis , Collagen/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Matrix/metabolism , GTP-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myxovirus Resistance Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
18.
J Cell Biol ; 194(2): 307-22, 2011 Jul 25.
Article in English | MEDLINE | ID: mdl-21768292

ABSTRACT

The morphogenetic and differentiation events required for bone formation are orchestrated by diffusible and insoluble factors that are localized within the extracellular matrix. In mice, the deletion of ICAP-1, a modulator of ß1 integrin activation, leads to severe defects in osteoblast proliferation, differentiation, and mineralization and to a delay in bone formation. Deposition of fibronectin and maturation of fibrillar adhesions, adhesive structures that accompany fibronectin deposition, are impaired upon ICAP-1 loss, as are type I collagen deposition and mineralization. Expression of ß1 integrin with a mutated binding site for ICAP-1 recapitulates the ICAP-1-null phenotype. Follow-up experiments demonstrated that ICAP-1 negatively regulates kindlin-2 recruitment onto the ß1 integrin cytoplasmic domain, whereas an excess of kindlin-2 binding has a deleterious effect on fibrillar adhesion formation. These results suggest that ICAP-1 works in concert with kindlin-2 to control the dynamics of ß1 integrin-containing fibrillar adhesions and, thereby, regulates fibronectin deposition and osteoblast mineralization.


Subject(s)
Calcification, Physiologic , Fibronectins/metabolism , Integrin beta1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Osteoblasts/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cytoskeletal Proteins/metabolism , Extracellular Matrix/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Muscle Proteins/metabolism , Osteoblasts/cytology , Protein Binding
20.
J Bone Miner Res ; 25(4): 706-15, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19821765

ABSTRACT

The bone matrix is composed mostly of collagen, but the initial and continuous presence of fibronectin was found to be crucial for collagen matrix integrity in vitro. It has been assumed that osteoblasts produce the fibronectin required for bone matrix formation. Using transgenic mice, we conditionally deleted fibronectin in the osteoblasts and in the liver using the cre-loxP system. We also used mice with mutated fibronectin and conditionally deleted beta(1)-integrin in osteoblasts to identify the receptor involved in fibronectin effects on osteoblasts. Conditional deletion of fibronectin in the differentiating osteoblasts [using the 2.3 kb collagen-alpha1(I) promoter] failed to show a decrease in fibronectin amount in the bone matrix despite evidence of successful deletion. Using these mice we established that osteoblast-derived fibronectin solely affects osteoblast function. This effect was not mediated by integrins that bind to the RGD motif. Conditional deletion of fibronectin in the liver showed a marked decrease in fibronectin content in the matrix associated with decreased mineral-to-matrix ratio and changed biomechanical properties but had no effect on osteoblasts or osteoclasts. In conclusion, osteoblast fibronectin affects osteoblasts function. This does not seem to be mediated by the RGD motif on fibronectin. In contrast, liver-derived fibronectin affects bone matrix properties without affecting osteoblast or osteoclast function. A novel role for liver-derived circulating fibronectin thus was defined and delineated from that of locally produced fibronectin.


Subject(s)
Bone Matrix/metabolism , Fibronectins/blood , Osteoblasts/metabolism , Animals , Bone Density , Cell Differentiation , Collagen/metabolism , Extracellular Matrix/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Integrin beta1/metabolism , Liver/metabolism , Male , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...