Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 258
Filter
1.
Gene ; 815: 146137, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35007686

ABSTRACT

The extracellular matrix (ECM) is composed of a mesh of proteins, proteoglycans, growth factors, and other secretory components. It constitutes the tumor microenvironment along with the endothelial cells, cancer-associated fibroblasts, adipocytes, and immune cells. The proteins of ECM can be functionally classified as adhesive proteins and matricellular proteins (MCP). In the tumor milieu, the ECM plays a major role in tumorigenesis and therapeutic resistance. The current review encompasses thrombospondins, osteonectin, osteopontin, tenascin C, periostin, the CCN family, laminin, biglycan, decorin, mimecan, and galectins. The matrix metalloproteinases (MMPs) are also discussed as they are an integral part of the ECM with versatile functions in the tumor stroma. In this review, the role of these proteins in tumor initiation, growth, invasion and metastasis have been highlighted, with emphasis on their contribution to tumor therapeutic resistance. Further, their potential as biomarkers and therapeutic targets based on existing evidence are discussed. Owing to the recent advancements in protein targeting, the possibility of agents to modulate MCPs in cancer as therapeutic options are discussed.


Subject(s)
Biomarkers, Tumor , Extracellular Matrix Proteins/physiology , Neoplasms/etiology , Neoplasms/therapy , Cell Adhesion Molecules/physiology , Extracellular Matrix Proteins/analysis , Humans , Matrix Metalloproteinases/physiology , Osteonectin/analysis , Osteonectin/physiology , Osteopontin/physiology , Tenascin/physiology , Thrombospondin 1/physiology , Treatment Outcome
2.
Cell Rep ; 37(13): 110170, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965439

ABSTRACT

Osteopontin (OPN) has been considered a potential biomarker of graft-versus-host disease (GVHD). However, the function of OPN in GVHD is still elusive. Using a mouse model of acute GVHD (aGVHD), we report that OPN generated by CD4+ T cells is sufficient to exert a beneficial effect in controlling aGVHD through limiting gastrointestinal pathology, a major target organ of aGVHD. CD4+ T cell-derived OPN works on CD44 expressed in intestinal epithelial cells (IECs) and abates cell death of IECs. OPN also modulates gut microbiota with enhanced health-associated commensal bacteria Akkermansia. Importantly, we use our in vivo mouse mutant model to specifically express OPN isoforms and demonstrate that secreted OPN (sOPN), not intracellular OPN (iOPN), is solely responsible for the protective role of OPN. This study demonstrates that sOPN generated by CD4+ T cells is potent enough to limit aGVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Hyaluronan Receptors/metabolism , Osteopontin/physiology , T-Lymphocytes/metabolism , Animals , Epithelial Cells/metabolism , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Intestines/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
3.
Invest Ophthalmol Vis Sci ; 62(12): 18, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34546326

ABSTRACT

Purpose: The aim of this study is to evaluate the expression of osteopontin (OPN) and its relationship with relative cytokines in patients with Graves' ophthalmopathy (GO), and to observe the effect of OPN on orbital fibroblasts (OFs) proliferation, migration, and the expression of relative cytokines, as well as the signaling pathways involved in its effect. Methods: The orbital adipose connective tissue was obtained from 24 patients with GO (12 cases of active GO, and 12 cases of inactive GO) and 12 healthy controls. OFs were isolated from orbital tissues obtained from patients with active GO who were undergoing orbital decompression surgery. Quantitative PCR and Western blot were performed to detect RNA and protein expression. The proliferation and cell migration rates of OFs were measured by methylthiazol tetrazolium (MTT) and the cell scratch test. Signaling pathway inhibitors, such as OPN monoclonal antibody 1A12, ERK1/2 inhibitor PD98059, and PI3K inhibitor LY294002, were applied to determine the involved pathways. Results: The mRNA and protein levels of OPN were increased in orbital adipose connective tissue from patients with active GO than those from patients with inactive GO (2.83-fold increase, P < 0.001; 1.91-fold increase, P < 0.05). The OPN mRNA level was positively correlated with CD40 ligand (CD40L) and hyaluronan synthases 2 (HAS2) mRNA in patients with GO. OPN promoted proliferation and migration rate of OFs and induced vascular endothelial growth factor (VEGF) and collagen I mRNA expression, and the effects were inhibited by 1A12 or LY294002. Conclusions: OPN in orbital adipose connective tissues were significantly increase in active GO, and there were significant correlations of OPN with CD40L and HAS2 mRNA levels in patients with GO. OPN promoted proliferation and migration of OFs and induced VEGF and collagen I mRNA expression in OFs through PI3K/Akt signaling pathway. This suggested a role for OPN in the pathogenesis of GO through the activation of OFs.


Subject(s)
Graves Ophthalmopathy/etiology , Osteopontin/physiology , Adipose Tissue/pathology , Adult , Blotting, Western , Cell Movement/physiology , Cell Proliferation/physiology , Cells, Cultured , Cytokines/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/pathology , Humans , Male , Middle Aged , Orbit/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
4.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 45(9): 1015-1023, 2020.
Article in English, Chinese | MEDLINE | ID: mdl-33051414

ABSTRACT

OBJECTIVES: To investigate the effect of ligand-receptor interaction of osteopontin (OPN)-CD44 on the expression of hyaluronic acid (HA) in the cultured human knee osteoarthritis (OA) chondrocytes via interfering the reaction between OPN and CD44 ligand-receptor. METHODS: The OA chondrocytes and normal chondrocytes were obtained from knee joint cartilage tissues in the patients with knee OA and malignant tumor respectively. The normal chondrocytes and OA chondrocytes were detected and analyzed, and then the intervention analysis of OA chondrocytes was carried out. The OA chondrocytes were divided into 4 groups: a negative control group, which was cultured with complete medium without any molecular intervention reagent; an OPN intervention group, which was cultured with recombinant human OPN (rhOPN) for 24 hours; a CD44 blocking group, which were pretreated with CD44 receptor specific antagonist for 1 hour to block the binding of OPN-CD44, and then treated with rhOPN for 23 hours; a CD44 homotype group, which was pretreated with CD44 for 1 hour and then treated with rhOPN for 23 hours. In addition, the study for OPN-CD44 axis was also divided into 4 groups: an OA-negative control group (OA-NC group), a si-OPN intervention group, a rhOPN intervention group, and a rhOPN + CD44 antibody (Ab) group. Western blotting, real-time PCR, and enzyme linked immunosorbent assay (ELISA) were used to detect the protein and mRNA expression levels of OPN, CD44, hyaluronate synthase (HAS), and HA, respectively. RESULTS: The protein expression levels of OPN, CD44, and HAS1 and the secretion levels of HA in the OA chondrocytes were higher than those in the normal chondrocytes. Compared with the OPN intervention group, the expression levels of HAS1, HAS2, HAS3 and HA in the CD44 blocking group were lower than those in OPN intervention group (all P<0.05); but there was no significant difference in the expression levels of HAS1, HAS2, HAS3 and HA between the CD44 homotype group and the OPN intervention group (all P>0.05). The results of OPN-CD44 axis study showed that: compared with the OA-NC group, the expression of CD44 in the rhOPN intervention group was slightly lower, but the protein and mRNA levels of HAS1 were significantly increased (all P<0.05); compared with the OA-NC group, the expression of CD44 was up-regulated, but the protein and mRNA level of HAS1 were significantly inhibited in the si-OPN intervention group (all P<0.05); compared with the OA-NC group, the protein and mRNA levels of HAS1 in the rhOPN+CD44 Ab group were also significantly inhibited (all P<0.05). CONCLUSIONS: The OPN in OA chondrocytes can promote the expression of HAS1, and the OPN can stimulate the secretion of HAS and induce HA expression by reacting with CD44 ligand receptor. They constitute the axis of OPN/CD44/HAS1, which plays an important role in regulating the expression of HA in chondrocytes.


Subject(s)
Hyaluronic Acid , Osteoarthritis, Knee , Osteopontin , Chondrocytes , Humans , Hyaluronan Receptors/genetics , Knee Joint , Osteopontin/genetics , Osteopontin/physiology
5.
Int. j. morphol ; 38(5): 1398-1404, oct. 2020. tab, graf
Article in English | LILACS | ID: biblio-1134455

ABSTRACT

SUMMARY: The objective of this study were bone defect complications that occur due to traumas or infections. Bone grafts are required to provide support, fill gaps and improve biological repair in skeletal damage. Dexamethasone plays role in calcium signaling modulation and used in diseases. Aim of this study was to evaluate osteonectin and osteopontin expressions in new bone development after dexamethasone application on tibial bone defects. Rats were divided into defect, defect+graft and defect+graft+dexamethasone treated groups. Tibial bone defect created, and rats were kept immobile for 28 days. Alloplastic material was placed in defect area in second and group third groups. 2.5 mg/kg Dex and normal saline were injected to dexamethasone and defect groups twice a week for 56 days. Inflammation and congestion were increased in defect and defect+graft groups. Defect+graft+dexamethasone group; increased number of osteoblast and osteocyte cells, dense bone matrix, formation of new bone trabeculae was observed. Defect+graft group; osteonectin expression in graft regions, osteoblast cells, some connective tissue cells and fibers were seen whereas in defect+graft+dexamethasone group; osteopontin expression in osteoblast and osteocyte cells of new bone trabeculae were observed. Dexamethasone may lead to formation of new bone trabeculae into the graft material resulting in increased osteoconduction and osteoinductive effect for differentiation of osteon.


RESUMEN: Los defectos óseos son complicaciones que ocurren debido a traumas o infecciones. Se requieren injertos óseos para proporcionar apoyo, llenar los espacios y mejorar la reparación biológica en el hueso dañado. La dexametasona desempeña un papel importante en la modulación de la señalización del calcio y se usa en enfermedades. El objetivo de este estudio fue evaluar las expresiones de osteonectina y osteopontina en el desarrollo óseo después de la aplicación de dexametasona en defectos óseos tibiales. Las ratas se dividieron en grupos: defecto, defecto + injerto y defecto + injerto + grupos tratados con dexametasona. Se creó un defecto óseo tibial, y las ratas se mantuvieron inmóviles durante 28 días. El material aloplástico se colocó en el área del defecto en el segundo y tercer grupo. Se inyectaron 2,5 mg / kg de dexametasona y solución salina normal a grupos de defectos dos veces por semana durante 56 días. La inflamación y la congestión aumentaron en los grupos de defectos y defectos + injerto; En el grupo defecto + injerto + grupo tratado con dexametasona se observó un aumento en el número de osteoblastos y osteocitos, de matriz ósea densa y en la formación de nuevas trabéculas óseas. En el grupo defecto + grupo de injerto se observó la expresión de osteonectina en las áreas de injerto, osteoblastos, algunas células y fibras de tejido conectivo, mientras que en el grupo defecto + injerto + dexametasona se observó la expresión de osteopontina en osteoblastos y osteocitos y formación de nuevas trabéculas óseas . En conclusión la dexametasona puede conducir a la formación de nuevas trabéculas óseas en el material de injerto, lo que resulta en un aumento de la osteoconducción y un efecto osteoinductivo para la diferenciación del osteón.


Subject(s)
Animals , Male , Rats , Tibia/surgery , Tibia/drug effects , Dexamethasone/administration & dosage , Bone Transplantation , Tibia/pathology , Bone Regeneration , Immunohistochemistry , Osteonectin/physiology , Bone Remodeling , Rats, Wistar , Disease Models, Animal , Osteopontin/physiology
6.
J Am Heart Assoc ; 9(18): e017071, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32865099

ABSTRACT

Background We previously reported that osteopontin plays an essential role in accelerating both reparative fibrosis and clearance of dead cells (efferocytosis) during tissue repair after myocardial infarction (MI) and galectin-3hiCD206+ macrophages is the main source of osteopontin in post-MI heart. Interleukin-10- STAT3 (signal transducer and activator of transcription 3)-galectin-3 axis is essential for Spp1 (encoding osteopontin) transcriptional activation in cardiac macrophages after MI. Here, we investigated the more detailed mechanism responsible for functional maturation of osteopontin-producing macrophages. Methods and Results In post-MI hearts, Spp1 transcriptional activation occurred almost exclusively in MerTK (Mer tyrosine kinase)+ galectin-3hi macrophages. The induction of MerTK on galectin-3hi macrophages is essential for their functional maturation including efferocytosis and Spp1 transcriptional activity. MerTK+galectin-3hi macrophages showed a strong activation of both STAT3 and ERK (extracellular signal-regulated kinase). STAT3 inhibition suppressed the differentiation of osteopontin-producing MerTK+galectin-3hi macrophages, however, STAT3 activation was insufficient at inducing Spp1 transcriptional activity. ERK inhibition suppressed Spp1 transcriptional activation without affecting MerTK or galectin-3 expression. Concomitant activation of ERK is required for transcriptional activation of Spp1. In Il-10 knockout enhanced green fluorescent protein-Spp1 knock-in mice subjected to MI, osteopontin-producing macrophages decreased but did not disappear entirely. Interleukin-10 and macrophage colony-stimulating factor synergistically activated STAT3 and ERK and promoted the differentiation of osteopontin-producing MerTK+galectin-3hi macrophages in bone marrow-derived macrophages. Coadministration of anti-interleukin-10 plus anti-macrophage colony-stimulating factor antibodies substantially reduced the number of osteopontin-producing macrophages by more than anti-interleukin-10 antibody alone in post-MI hearts. Conclusions Interleukin-10 and macrophage colony-stimulating factor act synergistically to activate STAT3 and ERK in cardiac macrophages, which in turn upregulate the expression of galectin-3 and MerTK, leading to the functional maturation of osteopontin-producing macrophages.


Subject(s)
MAP Kinase Signaling System/physiology , Macrophages/pathology , Myocardial Infarction/pathology , Osteopontin/physiology , c-Mer Tyrosine Kinase/physiology , Animals , Disease Models, Animal , Flow Cytometry , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Osteopontin/metabolism , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/physiology , c-Mer Tyrosine Kinase/metabolism
7.
Life Sci ; 245: 117328, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31954162

ABSTRACT

AIMS: Atrial fibrosis is a common feature of atrial fibrillation (AF). Recently, it is reported that osteopontin (OPN) can induce fibrosis in lungs, livers and kidneys. However, its role in atrial fibrosis remains unclear. Here, we sought to determine the involvement of OPN in atrial fibrosis and the underlying mechanisms during this pathological remodeling process. MATERIALS AND METHODS: Protein expressions were determined by enzyme-linked immunosorbent assay (ELISA), immunohistochemical staining and immunoblotting. mRNA expressions were detected by qRT-PCR. Cell proliferation was assessed by CCK-8. Left atrial electroanatomical voltage maps were created using PentaRay catheters and a 3-dimensional mapping system. KEY FINDINGS: OPN was highly expressed in the circulation of AF patients and was further increased with the progression of AF. In addition, correlation analysis showed that circulating OPN positively related with low-voltage areas (LVAs, a marker of atrial fibrosis) in AF patients. Immunohistological staining and immunoblotting revealed an increased expression of OPN in AF patients who present a higher degree of atrial fibrosis. Furthermore, in vitro studies in cultured human atrial fibroblasts (hAFs) demonstrated that OPN promoted the proliferation of fibroblasts and increased production of collagen I and fibronectin. Mechanistically, the profibrotic effects of OPN on atrial fibroblasts were determined via activating Akt/GSK-3ß/ß-catenin signaling and suppressing autophagy. SIGNIFICANCE: This study uncovered a previously unrecognized profibrotic role of OPN in atrial fibrosis, which was achieved through activation of Akt/GSK-3ß/ß-catenin signaling pathway and suppression of autophagy, implying a promising therapeutic target in atrial fibrosis and AF.


Subject(s)
Atrial Fibrillation/pathology , Autophagy , Glycogen Synthase Kinase 3 beta/metabolism , Heart Atria/pathology , Osteopontin/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , beta Catenin/metabolism , Aged , Atrial Fibrillation/metabolism , Autophagy/physiology , Case-Control Studies , Collagen/metabolism , Female , Fibronectins/metabolism , Fibrosis , Heart Atria/metabolism , Humans , Male , Middle Aged , Polymerase Chain Reaction , Signal Transduction/physiology
8.
Cells ; 8(8)2019 08 02.
Article in English | MEDLINE | ID: mdl-31382483

ABSTRACT

The glycoprotein osteopontin (OPN) possesses multiple functions in health and disease. To this end, osteopontin has beneficial roles in wound healing, bone homeostasis, and extracellular matrix (ECM) function. On the contrary, osteopontin can be deleterious for the human body during disease. Indeed, osteopontin is a cardinal mediator of tumor-associated inflammation and facilitates metastasis. The purpose of this review is to highlight the importance of osteopontin in malignant processes, focusing on lung and pleural tumors as examples.


Subject(s)
Inflammation/pathology , Neoplasms/pathology , Osteopontin/physiology , Animals , Cell Line, Tumor , Humans
9.
Clin Exp Rheumatol ; 37(6): 899-905, 2019.
Article in English | MEDLINE | ID: mdl-31074728

ABSTRACT

OBJECTIVES: Kidney biopsy is the gold standard for the diagnosis of lupus nephritis (LN). Conventional biomarkers of disease activity or renal function, such as complement levels, anti-dsDNA, serum creatinine, urinary sediment and proteinuria, do not have a sensitive diagnostic and prognostic value, therefore new biomarkers are needed to help predict or monitor LN. Osteopontin (OPN) is a pro-inflammatory molecule detectable in serum and renal tissue. The aim of this study was to evaluate OPN as a biomarker of renal involvement in patients with systemic lupus erythematosus (SLE) and correlate its levels with disease activity and laboratory features. METHODS: OPN was measured in the serum and urine of SLE patients with active LN (n=14), LN in remission (n=20), SLE without kidney involvement (n=22) and age- and sex-matched healthy controls (HC, n=20). RESULTS: OPN levels were significantly higher in urine than in serum in both groups of patients and controls (p<0.001). Serum OPN levels were higher in the LN patients than in HC and in SLE patients without renal involvement (p<0.0001 and 0.0032, respectively), regardless of the phase of renal activity. SLE patients without renal involvement and controls showed similar serum levels. We detected a direct correlation between low complement levels and OPN serum levels in patients with LN (p=0.014; R=0.438). Moreover, a higher percentage of patients with LN, compared to SLE without LN and HC, showed abnormal serum OPN. CONCLUSIONS: Our data suggest that serum OPN could be considered a biomarker of renal involvement, without differentiating between active and remission LN.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Osteopontin/physiology , Biomarkers/blood , Humans , Kidney , Lupus Erythematosus, Systemic/blood , Lupus Nephritis/blood , Osteopontin/blood
10.
Cells ; 8(4)2019 04 25.
Article in English | MEDLINE | ID: mdl-31027317

ABSTRACT

Inducible cyclization recombinase (Cre) transgenic mouse strains are powerful tools for cell lineage tracing and tissue-specific knockout experiments. However, low efficiency or leaky expression can be important pitfalls. Here, we compared the efficiency and specificity of two commonly used cholangiocyte-specific Cre drivers, the Opn-iCreERT2 and Ck19-CreERT drivers, using a tdTomato reporter strain. We found that Opn-iCreERT2 triggered recombination of the tdTomato reporter in 99.9% of all cholangiocytes while Ck19-CreERT only had 32% recombination efficiency after tamoxifen injection. In the absence of tamoxifen, recombination was also induced in 2% of cholangiocytes for the Opn-iCreERT2 driver and in 13% for the Ck19-CreERT driver. For both drivers, Cre recombination was highly specific for cholangiocytes since recombination was rare in other liver cell types. Toxic liver injury ectopically activated Opn-iCreERT2 but not Ck19-CreERT expression in hepatocytes. However, ectopic recombination in hepatocytes could be avoided by applying a three-day long wash-out period between tamoxifen treatment and toxin injection. Therefore, the Opn-iCreERT2 driver is best suited for the generation of mutant bile ducts, while the Ck19-CreERT driver has near absolute specificity for bile duct cells and is therefore favorable for lineage tracing experiments.


Subject(s)
Genetic Engineering/methods , Keratin-19/metabolism , Osteopontin/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Bile Ducts/metabolism , Cell Lineage/drug effects , Female , Gene Expression/genetics , Gene Expression/physiology , Integrases/biosynthesis , Integrases/genetics , Integrases/metabolism , Keratin-19/genetics , Keratin-19/physiology , Liver/metabolism , Male , Mice , Mice, Transgenic/genetics , Osteopontin/genetics , Osteopontin/physiology , Recombinant Proteins/metabolism , Tamoxifen/pharmacology
11.
Brain Pathol ; 29(6): 793-802, 2019 11.
Article in English | MEDLINE | ID: mdl-30868685

ABSTRACT

In severe forms of cerebral amyloid angiopathy (CAA) pathology, vascular calcification has been observed in the cerebral cortex, both in vivo on MRI and CT, and post-mortem using histopathology. However, the pathomechanisms leading to calcification of CAA-laden arteries are unknown. Therefore, we investigated the correlation between calcification of cortical arterioles and several potential modulators of vascular calcification using immunohistochemistry in a unique collection of brain material of patients with a hereditary form of CAA, namely hereditary cerebral hemorrhage with amyloidosis-Dutch type (HCHWA-D or D-CAA). We show a topographical association of osteopontin (OPN) and TGFß signaling factor phospho-SMAD2/3 (pSMAD2/3) in calcified CAA vessel walls. OPN and pSMAD2/3 gradually accumulate in vessels prior to calcification. Moreover, we found that the vascular accumulation of Collagen 1 (Col1), OPN and pSMAD2/3 immunomarkers correlated with the CAA severity. This was independently of the vessel size, including capillaries in the most severe cases. We propose that calcification of CAA vessels in the observed HCHWA-D cases may be induced by extracellular OPN trapped in the fibrotic Col1 vessel wall, independently of the presence of vascular amyloid.


Subject(s)
Calcinosis/pathology , Cerebral Amyloid Angiopathy/pathology , Osteopontin/metabolism , Aged , Alzheimer Disease/pathology , Amyloid , Amyloid beta-Peptides/metabolism , Amyloidosis/pathology , Arterioles/pathology , Brain/pathology , Capillaries/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy, Familial/pathology , Cerebral Cortex/pathology , Collagen Type I/metabolism , Female , Humans , Male , Middle Aged , Osteopontin/physiology , Phosphorylation , Smad2 Protein/metabolism , Smad3 Protein/metabolism
12.
Article in English | MEDLINE | ID: mdl-30865978

ABSTRACT

Human milk (HM) contains hundreds of proteins with very diverse functions that likely contribute to the short- and long-term beneficial effects of breastfeeding. These functions include serving as a source of amino acids, improving the bioavailability of micronutrients, including vitamins, minerals, and trace elements, providing immunologic defense, stimulating intestinal growth and maturation, shaping the microbiome, and enhancing learning and memory. Human milk proteins can be broadly classified into 3 categories: caseins, whey proteins, and mucins, which are present in the milk fat globule membrane. HM is whey predominant; however, the whey/casein ratio of HM changes from 90/10 in colostrum to 60/40 in mature HM. The whey proteins present in significant quantities in the whey fraction are α-lactalbumin, lactoferrin, IgA, osteopontin, and lysozyme. Additionally, bioactive peptides are formed during digestion of casein and whey, and glycans from glycoproteins are bifidogenic, adding further complexity to the functional properties of HM proteins. Recent advances in dairy technology have enabled isolation of bioactive milk proteins from bovine milk in sufficient quantities for clinical studies and, in some cases, addition to commercially available infant formula. Herein, the current evidence on HM protein composition and bioactivity of HM proteins is reviewed.


Subject(s)
Milk Proteins/chemistry , Milk, Human/chemistry , Milk, Human/physiology , Caseins/analysis , Female , Glycolipids/physiology , Glycoproteins/physiology , Humans , Infant , Infant, Newborn , Lactoferrin/physiology , Lipid Droplets , Milk Proteins/metabolism , Mucins/analysis , Mucins/physiology , Nutritive Value , Osteopontin/physiology , Whey Proteins/analysis
13.
FASEB J ; 33(6): 7236-7251, 2019 06.
Article in English | MEDLINE | ID: mdl-30893567

ABSTRACT

Recent studies indicated that osteopontin (OPN) was involved in the genesis and progression of pulmonary arterial hypertension (PAH); however, its role in congenital heart disease-associated PAH (CHD/PAH) remains unknown. Our results showed that OPN was increased in lungs and plasma of patients with Eisenmenger syndrome; moreover, OPN and αVß3-integrin expression levels were augmented in rat lungs exposed to systemic-to-pulmonary shunt. Cell culture assay demonstrated that distal pulmonary arterial smooth muscle cells (PASMCs) from rat lungs suffering from volume and pressure overload exhibited enhanced proliferation compared with those from healthy rats. Mechanical stretch (20% at 1 Hz) increased OPN expression and activated ERK1/2 and protein kinase B (Akt) signal pathway in distal PASMCs from healthy rats. Interestingly, OPN enhanced the proliferation and migration of PASMCs while blocking αVß3-integrin with neutralizing antibody LM609 or Arg-Gly-Asp peptidomimetic antagonist cyclo(Ala-Arg-Gly-Asp-3-aminomethylbenzoyl) (XJ735), rectified the proliferative and migratory effects of OPN, which were partially mediated via ERK1/2 and Akt signaling pathways. Furthermore, surgical correction of systemic-to-pulmonary shunt, particularly XJ735 supplementation after surgical correction of systemic-to-pulmonary shunt, significantly alleviated the pulmonary hypertensive status in terms of pulmonary hemodynamic indices, pulmonary vasculopathy, and right ventricular hypertrophy. In summary, OPN alteration in lungs exposed to systemic-to-pulmonary shunt exerts a deteriorative role in pulmonary vascular remodeling through modulating the proliferation and migration of PASMCs, at least in part, via ανß3-ERK1/2 and ανß3-Akt signaling pathways. Antagonizing OPN receptor ανß3-integrin accelerated the regression of pulmonary vasculopathy after surgical correction of systemic-to-pulmonary shunt, indicating a potential therapeutic strategy for patients with CHD/PAH.-Meng, L., Liu, X., Teng, X., Gu, H., Yuan, W., Meng, J., Li, J., Zheng, Z., Wei, Y., Hu, S. Osteopontin plays important roles in pulmonary arterial hypertension induced by systemic-to-pulmonary shunt.


Subject(s)
Eisenmenger Complex/physiopathology , Hypertension, Pulmonary/physiopathology , Osteopontin/physiology , Adult , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Eisenmenger Complex/complications , Humans , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/physiopathology , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/biosynthesis , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/physiology , Lung/blood supply , Lung/pathology , MAP Kinase Signaling System , Male , Middle Aged , Myocytes, Smooth Muscle/metabolism , Osteopontin/biosynthesis , Osteopontin/genetics , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Young Adult
14.
Brain Res ; 1714: 52-64, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30790559

ABSTRACT

We previously reported that mRNA encoding secreted phosphoprotein 1 (SPP1), also known as osteopontin, is preferentially expressed in large neurons in layer V of the macaque motor cortex, most of which are presumed to be corticospinal tract neurons. As a first step to elucidating the cellular function of SPP1 in macaque neurons, we examined the localization of SPP1 in the primary motor cortex (M1) of the macaque by using immunohistochemistry. SPP1 immunoreactivity was found to be localized in the cell bodies of neurons, but not outside the cells, indicating that SPP1 was not secreted from these neurons. The results of electron microscope analysis and double-labeling analysis with marker proteins suggested that SPP1 was localized in the mitochondria of neurons. The distributions of SPP1 in the neurons corresponded to those of integrin αV, a putative receptor for SPP1. The distribution of SPP1 was also investigated in macaques whose M1 had been lesioned. We found that SPP1 was secreted by proliferated microglia in the lesioned area. Double-labeling analysis indicated that SPP1 immunoreactivity in the microglia was colocalized with CD44, another putative receptor for SPP1. Success rates in the small-object-retrieval task were positively correlated with SPP1 immunoreactivity in the neurons in the perilesional area. SPP1 has multiple roles in the macaque motor cortex, and it may be a key protein during recovery of hand movement after brain damage.


Subject(s)
Motor Cortex/metabolism , Neurons/metabolism , Osteopontin/metabolism , Animals , Female , Hyaluronan Receptors/immunology , In Situ Hybridization/methods , Macaca mulatta , Male , Microglia/metabolism , Motor Cortex/pathology , Osteopontin/genetics , Osteopontin/physiology , Pyramidal Tracts/metabolism , RNA, Messenger/metabolism
15.
Arterioscler Thromb Vasc Biol ; 39(4): 613-622, 2019 04.
Article in English | MEDLINE | ID: mdl-30727754

ABSTRACT

Inflammatory cytokines are necessary for an acute response to injury and the progressive healing process. However, when this acute response does not resolve and becomes chronic, the same proteins that once promoted healing then contribute to chronic inflammatory pathologies, such as atherosclerosis. OPN (Osteopontin) is a secreted matricellular cytokine that signals through integrin and CD44 receptors, is highly upregulated in acute and chronic inflammatory settings, and has been implicated in physiological and pathophysiologic processes. Evidence from the literature suggests that OPN may fit within the Goldilocks paradigm with respect to cardiovascular disease, where acute increases are protective, attenuate vascular calcification, and promote postischemic neovascularization. In contrast, chronic increases in OPN are clinically associated with an increased risk for a major adverse cardiovascular event, and OPN expression is a strong predictor of cardiovascular disease independent of traditional risk factors. With the recent finding that humans express multiple OPN isoforms as the result of alternative splicing and that these isoforms have distinct biologic functions, future studies are required to determine what OPN isoform(s) are expressed in the setting of vascular disease and what role each of these isoforms plays in vascular disease progression. This review aims to discuss our current understanding of the role(s) of OPN in vascular disease pathologies using evidence from in vitro, animal, and clinical studies. Where possible, we discuss what is known about OPN isoform expression and our understanding of OPN isoform contributions to cardiovascular disease pathologies.


Subject(s)
Inflammation/metabolism , Osteopontin/physiology , Vascular Diseases/metabolism , Alternative Splicing , Animals , Atherosclerosis/physiopathology , Calcinosis/physiopathology , Glycosylation , Humans , Hyaluronan Receptors/physiology , Inflammation/physiopathology , Integrins/physiology , Ischemia/physiopathology , Mice , Models, Cardiovascular , Neointima/pathology , Osteopontin/chemistry , Osteopontin/genetics , Phosphorylation , Protein Isoforms/physiology , Protein Processing, Post-Translational , Risk Factors
16.
FASEB J ; 33(2): 1681-1694, 2019 02.
Article in English | MEDLINE | ID: mdl-30199283

ABSTRACT

Osteopontin (OPN) is a pleiotropic protein and is abundantly present in milk. Its functions include immune modulation and cellular proliferation and differentiation. OPN is highly expressed in the brain. We investigated the effects of milk-derived OPN on brain development of mouse pups. Wild-type (WT) dams producing OPN+ milk and OPN knockout (KO) dams producing OPN- milk nursed WT pups (OPN+/+), yielding 2 pup treatment groups, OPN+ OPN+/+ and OPN- OPN+/+, for comparison. Preliminary studies supported use of this model by showing high concentrations of OPN in milk of WT dams and no OPN in milk of OPN KO dams, and production of similar amounts of milk by WT and KO dams. The ability of ingested milk OPN to enter the brain was revealed by appearance of orally gavaged [125I]-labeled and antibody-probed milk OPN in brains of pups. Brain OPN mRNA levels were similar in both nursed groups, but the brain OPN protein level was significantly lower in the OPN- OPN+/+ group at postnatal days 6 and 8. Behavior tests showed impaired memory and learning ability in OPN- OPN+/+ pups. In addition, our study revealed increased expression of myelination-related proteins and elevated proliferation and differentiation of NG-2 glia into oligodendrocytes in the brain of OPN+ OPN+/+ pups, accompanied by increased activation of ERK-1/2 and PI3K/Akt signaling. We concluded that milk OPN can play an important role in brain development and behavior in infancy by promoting myelination.-Jiang, R., Prell, C., Lönnerdal, B. Milk osteopontin promotes brain development by up-regulating osteopontin in the brain in early life.


Subject(s)
Brain/growth & development , Milk/metabolism , Osteopontin/physiology , Up-Regulation , Animals , Animals, Suckling , Behavior, Animal , Female , Learning , Memory , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/metabolism , Oligodendroglia/cytology , Osteopontin/genetics , Osteopontin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Protein Kinases/metabolism , RNA, Messenger/genetics , Signal Transduction
17.
Inflamm Res ; 68(2): 93-102, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30456594

ABSTRACT

BACKGROUND: Osteopontin (OPN) is a pleiotropic glycoprotein expressed in various cell types in animals and in humans, including bone, immune, smooth muscle, epithelial and endothelial cells. Moreover, OPN is found in kidneys (in the thick ascending limbs of the loop of Henle and in distal nephrons) and urine. The protein plays an important role in mineralization and bone resorption. In addition, OPN is involved in the regulation of immunity and inflammation, angiogenesis and apoptosis. It was demonstrated that OPN and some OPN gene polymorphic variants are associated with the pathogenesis and progression of multiple disorders, such as cancer, autoimmune, neurodegenerative and cardiovascular diseases. Moreover, recent studies suggested that OPN is associated with the pathogenesis of renal failure. METHODS: In this review, I briefly discussed the role of OPN and its gene polymorphisms in kidney physiology, as well as in various kidney diseases. FINDINGS AND CONCLUSION: Most studies reported that OPN expression is elevated in urolithiasis, and also in acute and chronic kidney diseases, and in renal allograft dysfunction. Moreover, it was demonstrated that polymorphic variants of the OPN gene may be associated with renal failure. However, some reports suggested that OPN is essential for tubulogenesis, and that it inhibits calcium oxalate crystal formation and retention, nitric oxide synthesis, cell apoptosis and promotes cell regeneration. Thus, further studies are required to fully understand the role of OPN in kidney physiology and pathology. Eventually, these studies may result in the identification of OPN as a valuable marker for renal dysfunction prognosis and treatment.


Subject(s)
Kidney Diseases/physiopathology , Osteopontin/physiology , Animals , Biomarkers/analysis , Humans , Kidney/metabolism , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Osteopontin/analysis , Osteopontin/genetics
18.
Lab Invest ; 99(3): 331-345, 2019 03.
Article in English | MEDLINE | ID: mdl-29959420

ABSTRACT

Osteopontin (OPN) is critical for ischemia-induced neovascularization. Unlike rodents, humans express three OPN isoforms (a, b, and c); however, the roles of these isoforms in post-ischemic neovascularization and cell migration remain undefined. Our objective was to determine if OPN isoforms differentially affect post-ischemic neovascularization and to elucidate the mechanisms underlying these differences. To investigate if human OPN isoforms exert divergent effects on post-ischemic neovascularization, we utilized OPN-/- mice and a loss-of-function/gain-of-function approach in vivo and in vitro. In this study OPN-/- mice underwent hindlimb ischemia surgery and 1.5 × 106 lentivirus particles were administered intramuscularly to overexpress OPNa, OPNb, or OPNc. OPNa and OPNc significantly improved limb perfusion 30.4% ± 0.8 and 70.9% ± 6.3, respectively, and this translated to improved functional limb use, as measured by voluntary running wheel utilization. OPNa- and OPNc-treated animals exhibited significant increases in arteriogenesis, defined here as the remodeling of existing arterioles into larger conductance arteries. Macrophages play a prominent role in the arteriogenesis process and OPNa- and OPNc-treated animals showed significant increases in macrophage accumulation in vivo. In vitro, OPN isoforms did not affect macrophage polarization, whereas all three isoforms increased macrophage survival and decreased macrophage apoptosis. However, OPN isoforms exert differential effects on macrophage migration, where OPNa and OPNc significantly increased macrophage migration, with OPNc serving as the most potent isoform. In conclusion, human OPN isoforms exert divergent effects on neovascularization through differential effects on arteriogenesis and macrophage accumulation in vivo and on macrophage migration and survival, but not polarization, in vitro. Altogether, these data support that human OPN isoforms may represent novel therapeutic targets to improve neovascualrization and preserve tissue function in patients with obstructive artery diseases.


Subject(s)
Ischemia/pathology , Ischemia/physiopathology , Macrophages/pathology , Macrophages/physiology , Neovascularization, Physiologic , Osteopontin/physiology , Animals , Apoptosis , Arterial Occlusive Diseases/pathology , Arterial Occlusive Diseases/physiopathology , Arterial Occlusive Diseases/therapy , Cell Movement , Cell Survival , Disease Models, Animal , Humans , Ischemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteopontin/deficiency , Osteopontin/genetics , Osteopontin/therapeutic use , Protein Isoforms/genetics , Protein Isoforms/physiology , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Vascular Remodeling/genetics , Vascular Remodeling/physiology
19.
J Biomech ; 80: 45-52, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30205977

ABSTRACT

Non-collagenous proteins are a vital component of bone matrix. Amongst them, osteocalcin (OC) and osteopontin (OPN) hold special significance due to their intimate interaction with the mineral and collagenous matrix in bone. Both proteins have been associated with microdamage and fracture, but their structural role in energy dissipation is unclear. This study used bone tissue from genetic deficient mice lacking OC and/or OPN and subjected them to a series of creep-fatigue-creep tests. To this end, whole tibiae were loaded in four-point bending to 70% stiffness loss which captured the three characteristic phases of fatigue associated with initiation, propagation, and coalescence of microdamage. Fatigue loading preceded and followed creep tests to determine creep and dampening parameters. Microdamage in the form of linear microcracks and diffuse damage were analyzed by histology. It was shown that OC and OPN were 'activated' following stiffness loss associated with fatigue damage where they facilitated creep and dampening parameters (i.e. increased energy dissipation). More specifically, post-fatigue creep rate and dampening were significantly greater in wild-types (WTs) than genetic deficient mice (p < 0.05). These results were supported by microdamage analysis which showed significant increase in creep-associated diffuse damage formation in WTs compared to genetic deficient groups (p < 0.05). Based on these findings, we propose that during local yield events, OC and OPN rely on ionic interactions of their charged side chains and on hydrogen bonding to dissipate energy in bone.


Subject(s)
Bone and Bones/pathology , Osteocalcin/physiology , Osteopontin/physiology , Animals , Fractures, Bone/pathology , Genotype , Hindlimb , Hydrogen Bonding , Male , Materials Testing , Mice , Mice, Inbred C57BL , Mice, Knockout , Minerals , Osteocalcin/chemistry , Osteocalcin/genetics , Osteopontin/chemistry , Osteopontin/genetics , Tibia/pathology
20.
Clin Biochem ; 59: 17-24, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30003880

ABSTRACT

Osteopontin (OPN) is a highly phosphorylated glycophosphoprotein having acidic characteristics and rich in aspartic acid. OPN, a multifunctional protein, has important functions on cardiovascular diseases, cancer, diabetes and kidney stone diseases and in the process of inflammation, biomineralization, cell viability and wound healing. Osteopontin acts on organisms by playing a key role in secretion levels of interleukin-10 (IL-10), interleukin-12 (IL-12), interleukin-3 (IL-3), interferon-γ (IFN-γ), integrin αvB3, nuclear factor kappa B (NF-kB), macrophage and T cells, regulating the osteoclast function and affecting CD44 receptors. The aim of the present review is to address majority of different functions of OPN protein which are known, suspected or suggested through the data obtained about this protein yet.


Subject(s)
Osteopontin/metabolism , Osteopontin/physiology , Animals , Biomarkers/blood , Biomarkers/urine , Calcinosis/metabolism , Fractures, Bone/metabolism , Humans , Joint Diseases/metabolism , Macrophages/physiology , Osteopontin/blood , Plaque, Atherosclerotic/metabolism , Signal Transduction , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Vascular Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...