1
|
Chavez MB, Andras NL, Tan MH, Kolli TN, Chu EY, Goldberg HA, Foster BL. Exogenous bone sialoprotein improves extraction socket healing in Ibsp knockout and wild-type mice. Bone 2025; 192:117381. [PMID: 39722365 PMCID: PMC11761379 DOI: 10.1016/j.bone.2024.117381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Bone sialoprotein (Ibsp/BSP) is a bone-associated extracellular matrix protein. Ibsp knockout (Ibsp-/-) mice exhibit defective alveolar bone formation, mineralization, and healing. We hypothesized BSP would rescue defective alveolar bone healing in a molar extraction model in Ibsp-/- mice. Collagen gel with or without native rat BSP (nBSP) or recombinant rat BSP (rBSP) was delivered to sockets after first maxillary molar extraction in Ibsp-/- and wild-type (WT) mice. Tissues were harvested 0, 1, 2, 7, and 14 days post-procedure (dpp) and analyzed by micro-computed tomography, histology, and immunohistochemistry (IHC). Histology and IHC demonstrated that collagen and BSP were retained within sockets. At 14 dpp, both bone volume fraction (BV/TV) and bone mineral density (BMD) were increased by both nBSP (over 50 %) and rBSP (over 60 %), compared to collagen alone in Ibsp-/- mice. In WT alveolar bone, BV/TV and BMD were also increased by nBSP (over 30 %) and rBSP (over 60 %) compared to collagen controls. Gene expression analyses revealed few changes from delivery of nBSP, while addition of rBSP resulted in regulation of cell signaling, ECM, mineralization, and osteoblast/osteoclast-associated genes. Exogenous BSP rescued alveolar bone healing defects in Ibsp-/- mice and enhanced bone healing in WT mice. Despite both forms of BSP improving bone healing, the substantial differences in how they regulate gene expression suggests that exogenous BSP acts in a complex, multifunctional manner to promote bone healing. These results support BSP as a novel approach to improve the quantity and quality of new bone in socket healing.
Collapse
Affiliation(s)
- M B Chavez
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA; College of Dentistry, University of Iowa, Iowa City, IA, USA
| | - N L Andras
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M H Tan
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - T N Kolli
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - E Y Chu
- Division of Operative Dentistry, Department of General Dentistry, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - H A Goldberg
- Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Abdolvahabi Z, Ezzati-Mobaser S, Hesari Z. The route of autophagy regulation by osteopontin: a review on the linking mechanisms. J Recept Signal Transduct Res 2023; 43:102-108. [PMID: 38082480 DOI: 10.1080/10799893.2023.2291563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/15/2023] [Indexed: 01/23/2024]
Abstract
Autophagy is a dynamic intracellular process of protein degradation, which is mostly triggered by nutrient deprivation. This process initiates with the formation of autophagosomes, which they capture cytosolic material that is then degraded upon fusion with the lysosome. Several factors have been found to be associated with autophagy modulation, of which extracellular matrix (ECM) components has attracted the attention of recent studies. Osteopontin (OPN) is an important extracellular matrix component that has been detected in a wide range of tumor cells, and is involved in cancer cell invasion and metastasis. Recently, a number of studies have focused on the relationship of OPN with autophagy, by delineating the intracellular signaling pathways that connect OPN to the autophagy process. We will summarize signaling pathways and cell surface receptors, through which OPN regulates the process of autophagy.
Collapse
Affiliation(s)
- Zohreh Abdolvahabi
- Cellular and Molecular Research Centre, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Samira Ezzati-Mobaser
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Hesari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
3
|
Sørensen ES, Christensen B. Milk Osteopontin and Human Health. Nutrients 2023; 15:nu15112423. [PMID: 37299387 DOI: 10.3390/nu15112423] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Osteopontin (OPN) is a multifunctional protein found in all vertebrates. OPN is expressed in many different cell types, and is consequently found in most tissues and physiological secretions. OPN is involved in a multitude of biological processes, such as activation and regulation of the immune system; biomineralization; tissue-transformative processes, including growth and development of the gut and brain; interaction with bacteria; and many more. OPN is found in the highest concentrations in milk, where it is believed to initiate and regulate developmental, immunological and physiological processes in infants who consume milk. Processes for the isolation of bovine OPN for use in infant formula have been developed, and in recent years, many studies have investigated the effects of the intake of milk OPN. The purpose of this article is to review and compare existing knowledge about the structure and function of milk OPN, with a particular focus on the effects of milk OPN on human health and disease.
Collapse
Affiliation(s)
- Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Brian Christensen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| |
Collapse
|
4
|
Tanner L, Bergwik J, Bhongir RKV, Puthia M, Lång P, Ali MN, Welinder C, Önnerfjord P, Erjefält JS, Palmberg L, Andersson G, Egesten A. Tartrate resistant acid phosphatase 5 (TRAP5) mediates immune cell recruitment in a murine model of pulmonary bacterial infection. Front Immunol 2022; 13:1079775. [PMID: 36569898 PMCID: PMC9779928 DOI: 10.3389/fimmu.2022.1079775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction During airway infection, upregulation of proinflammatory cytokines and subsequent immune cell recruitment is essential to mitigate bacterial infection. Conversely, during prolonged and non-resolving airway inflammation, neutrophils contribute to tissue damage and remodeling. This occurs during diseases including cystic fibrosis (CF) and COPD where bacterial pathogens, not least Pseudomonas aeruginosa, contribute to disease progression through long-lasting infections. Tartrate-resistant acid phosphatase (TRAP) 5 is a metalloenzyme expressed by alveolar macrophages and one of its target substrates is the phosphoglycoprotein osteopontin (OPN). Methods We used a knockout mouse strain (Trap5-/-) and BALB/c-Tg (Rela-luc)31Xen mice paired with siRNA administration or functional protein add-back to elucidate the role of Trap5 during bacterial infection. In a series of experiments, Trap5-/- and wild-type control mice received intratracheal administration of P.aerugniosa (Xen41) or LPS, with mice monitored using intravital imaging (IVIS). In addition, multiplex cytokine immunoassays, flow cytometry, multispectral analyses, histological staining were performed. Results In this study, we found that Trap5-/- mice had impaired clearance of P. aeruginosa airway infection and reduced recruitment of immune cells (i.e. neutrophils and inflammatory macrophages). Trap5 knockdown using siRNA resulted in a decreased activation of the proinflammatory transcription factor NF-κB in reporter mice and a subsequent decrease of proinflammatory gene expression. Add-back experiments of enzymatically active TRAP5 to Trap5-/- mice restored immune cell recruitment and bacterial killing. In human CF lung tissue, TRAP5 of alveolar macrophages was detected in proximity to OPN to a higher degree than in normal lung tissue, indicating possible interactions. Discussion Taken together, the findings of this study suggest a key role for TRAP5 in modulating airway inflammation. This could have bearing in diseases such as CF and COPD where excessive neutrophilic inflammation could be targeted by pharmacological inhibitors of TRAP5.
Collapse
Affiliation(s)
- Lloyd Tanner
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Jesper Bergwik
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Ravi K. V. Bhongir
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Manoj Puthia
- Department of Dermatology and Venereology, Lund University and Skåne University Hospital, Lund, Sweden,Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Pernilla Lång
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mohamad N. Ali
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Charlotte Welinder
- Swedish National Infrastructure for Biological Mass Spectrometry (BioMS), Lund University, Lund, Sweden
| | - Patrik Önnerfjord
- Molecular Skeletal Biology, Section for Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jonas S. Erjefält
- Unit of Airway Inflammation, Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Lena Palmberg
- Work Environment Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Göran Andersson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Arne Egesten
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden,*Correspondence: Arne Egesten,
| |
Collapse
|
5
|
Zhu J, Yu X, Wang Y, Bai S, Lai J, Tong X, Xing Y. Longitudinal changes of lactopontin (milk osteopontin) in term and preterm human milk. Front Nutr 2022; 9:962802. [PMID: 35967792 PMCID: PMC9372532 DOI: 10.3389/fnut.2022.962802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
Background Lactopontin (LPN) in breast milk, also known as milk osteopontin is thought to play a myriad of important roles in infants when they are immature. The purpose of the present study was to examine the longitudinal changes in LPN concentrations in term and preterm milk, and elucidate the links between maternal characteristics, LPN levels, and child growth in a birth cohort. Methods 131 mothers who delivered term, moderate-late preterm (MPT), very preterm (VPT), and extremely preterm (EPT) infants were included, milk samples were collected at 7, 14, 28, and 120 days postpartum. LPN concentration was determined by multiple reaction monitoring (MRM) using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). Results Our results indicated that LPN change over time of VPT (P = 0.024) and EPT (P = 0.003) were significantly different from term milk, although they all gradually decreased with lactation. In terms of LPN-related factors, maternal age was a significant contributor in late mature milk and pre-pregnancy BMI a significant contributor to colostrum and transitional milk. We further investigated relationships between LPN levels and infant weight and our results suggested that high levels of LPN in breast milk might be useful for the catch-up growth of infants. Conclusion LPN levels in breast milk are related to maternal factors, and differences in LPN levels may affect the growth of infants. As milk is a critical part in the mother-breastmilk-infant "triad," the association between maternal-infant factors and milk LPN levels warrants further study.
Collapse
Affiliation(s)
- Jing Zhu
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology, Beijing, China
| | - Xue Yu
- School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yiran Wang
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology, Beijing, China
| | - Shasha Bai
- Beijing Institute of Nutritional Resources, Beijing Academy of Science and Technology, Beijing, China
| | - Jianqiang Lai
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
6
|
Xu C, Wu Y, Liu N. Osteopontin in autoimmune disorders: current knowledge and future perspective. Inflammopharmacology 2022; 30:385-396. [PMID: 35235108 DOI: 10.1007/s10787-022-00932-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/03/2022] [Indexed: 11/30/2022]
Abstract
Osteopontin (OPN) is a multifunctional cytokine and adhesion molecule, as well as an unusual regulator for both innate and adaptive immune responses. Several immune cells can produce OPN, including dendritic cells (DCs), macrophages, and T lymphocytes. OPN expression is reported to be increased in a wide range of disorders, including autoimmunity, cancer, and allergy. The overexpression of OPN in several autoimmune disorders, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), Type 1 diabetes (T1D), inflammatory bowel disease (IBD), Sjögren's, and myasthenia gravis, have been shown to be correlated with disease severity. Regarding the important regulatory roles of OPN in the immune system, this study aimed to review the role of this molecule in autoimmune disorders and to provide a complete view of the current knowledge in this field.
Collapse
Affiliation(s)
- Canhua Xu
- Department of Spine Surgery, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Yaohong Wu
- Department of Spine Surgery, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Ning Liu
- Department of Spine Surgery, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
7
|
Phosphorylated Osteopontin Secreted from Cancer Cells Induces Cancer Cell Motility. Biomolecules 2021; 11:biom11091323. [PMID: 34572536 PMCID: PMC8470647 DOI: 10.3390/biom11091323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/30/2021] [Accepted: 09/04/2021] [Indexed: 12/22/2022] Open
Abstract
Osteopontin (OPN) plays a pivotal role in cancer cell invasion and metastasis. Although OPN has a large number of phosphorylation sites, the functional significance of OPN phosphorylation in cancer cell motility remains unclear. In this study, we attempted to investigate whether phosphorylated OPN secreted from cancer cells affect cancer cell migration. Quantitative PCR and Western blot analyses revealed that MDA-MB435S, A549, and H460 cells highly expressed OPN, whereas the OPN expression levels in H358, MIAPaca-2, and Panc-1 cells were quite low or were not detected. Compared with the cancer cell lines with a low OPN expression, the high OPN-expressing cancer cell lines displayed a higher cell migration, and the cell migration was suppressed by the anti-OPN antibody. This was confirmed by the OPN overexpression in H358 cancer cells with a low endogenous OPN. Phos-tag ELISA showed that phosphorylated OPN was abundant in the cell culture media of A549 and H460 cells, but not in those of MDA-MB435S cells. Moreover, the A549 and H460 cell culture media, as well as the MDA-MB435S cell culture media with a kinase treatment increased cancer cell motility, both of which were abrogated by phosphatase treatment or anti-OPN antibodies. These results suggest that phosphorylated OPN secreted from cancer cells regulates cancer cell motility.
Collapse
|
8
|
Hattori T, Iwasaki-Hozumi H, Bai G, Chagan-Yasutan H, Shete A, Telan EF, Takahashi A, Ashino Y, Matsuba T. Both Full-Length and Protease-Cleaved Products of Osteopontin Are Elevated in Infectious Diseases. Biomedicines 2021; 9:biomedicines9081006. [PMID: 34440210 PMCID: PMC8394573 DOI: 10.3390/biomedicines9081006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022] Open
Abstract
Circulating full-length osteopontin (FL-OPN) is elevated in plasma from patients with various infectious diseases, such as adult T-cell leukemia, Mycobacterium tuberculosis (TB), hepatitis virus infection, leptospirosis, acquired immune deficiency syndrome (AIDS), AIDS/TB, and coronavirus disease 2019 (COVID-19). Proteolysis of OPN by thrombin, matrix metalloproteases, caspase 8/3, cathepsin D, plasmin, and enterokinase generates various cleaved OPNs with a variety of bioactivities by binding to different target cells. Moreover, OPN is susceptible to gradual proteolysis. During inflammation, one of the cleaved fragments, N-terminal thrombin-cleaved OPN (trOPN or OPN-Arg168 [OPN-R]), induces dendritic cell (DC) adhesion. Further cleavage by carboxypeptidase B2 or carboxypeptidase N removes Arg168 from OPN-R to OPN-Leu167 (OPN-L). Consequently, OPN-L decreases DC adhesion. In particular, the differences in plasma level over time are observed between FL-OPN and its cleaved OPNs during inflammation. We found that the undefined OPN levels (mixture of FL-OPN and cleaved OPN) were elevated in plasma and reflected the pathology of TB and COVID-19 rather than FL-OPN. These infections are associated with elevated levels of various proteases. Inhibition of the cleavage or the activities of cleaved products may improve the outcome of the therapy. Research on the metabolism of OPN is expected to create new therapies against infectious diseases.
Collapse
Affiliation(s)
- Toshio Hattori
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (G.B.); (H.C.-Y.); (A.T.)
- Correspondence: ; Tel./Fax: +81-866-22-9469
| | - Hiroko Iwasaki-Hozumi
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (G.B.); (H.C.-Y.); (A.T.)
| | - Gaowa Bai
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (G.B.); (H.C.-Y.); (A.T.)
| | - Haorile Chagan-Yasutan
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (G.B.); (H.C.-Y.); (A.T.)
- Mongolian Psychosomatic Medicine Department, International Mongolian Medicine Hospital of Inner Mongolia, Hohhot 010065, China
| | - Ashwnini Shete
- ICMR-National AIDS Research Institute, 73 G-Block, MIDC, Bhosari, Pune 411026, India;
| | - Elizabeth Freda Telan
- STD AIDS Cooperative Central Laboratory, San Lazaro Hospital, Manila 1003, Philippines;
| | - Atsushi Takahashi
- Research Institute of Health and Welfare, Kibi International University, Takahashi 716-8508, Japan; (H.I.-H.); (G.B.); (H.C.-Y.); (A.T.)
| | - Yugo Ashino
- Department of Respiratory Medicine, Sendai City Hospital, Sendai 982-8502, Japan;
| | - Takashi Matsuba
- Department of Animal Pharmaceutical Science, School of Pharmaceutical Science, Kyusyu University of Health and Welfare, Nobeoka 882-8508, Japan;
| |
Collapse
|
9
|
Amilca-Seba K, Sabbah M, Larsen AK, Denis JA. Osteopontin as a Regulator of Colorectal Cancer Progression and Its Clinical Applications. Cancers (Basel) 2021; 13:cancers13153793. [PMID: 34359694 PMCID: PMC8345080 DOI: 10.3390/cancers13153793] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/29/2022] Open
Abstract
Simple Summary The mortality of colorectal cancer is principally related to metastatic disease at the time of diagnosis or to the growth of initially undetectable micro-metastasis. Current therapeutic strategies are efficient in patients with locally advanced cancer, but are rarely able to cure patients with metastatic disease. Therapeutic failure is mainly associated with drug resistance and an aggressive phenotype. The identification of new biomarkers for micro-metastasis and tumor progression remains an unmet clinical need that should allow for improved patient stratification for optimal treatment and may lead to the identification of novel therapeutic targets. Osteopontin (OPN), a multifunctional protein, has emerged as a potentially valuable biomarker in several cancer types. This review principally describes the molecular mechanisms of OPN that are associated with colorectal cancer (CRC) progression and metastasis, as well as the use of OPN as a clinical biomarker. This review identifies a role for OPN as a biomarker ready for extended clinical application and discusses its use as a therapeutic target. Abstract A high expression of the phosphoprotein osteopontin (OPN) has been associated with cancer progression in several tumor types, including breast cancer, hepatocarcinoma, ovarian cancer, and colorectal cancer (CRC). Interestingly, OPN is overexpressed in CRC and is associated with a poor prognosis linked to invasion and metastasis. Here, we review the regulation and functions of OPN with an emphasis on CRC. We examine how epigenetic and genetic regulators interact with the key signaling pathways involved in this disease. Then, we describe the role of OPN in cancer progression, including proliferation, survival, migration, invasion, and angiogenesis. Furthermore, we outline the interest of using OPN as a clinical biomarker, and discuss if and how osteopontin can be implemented as a routine assay in clinical laboratories for monitoring CRC patients. Finally, we discuss the use of OPN an attractive, but challenging, therapeutic target.
Collapse
Affiliation(s)
- Katyana Amilca-Seba
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
| | - Michèle Sabbah
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Annette K. Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - Jérôme A. Denis
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France; (K.A.-S.); (M.S.); (A.K.L.)
- Institut National de la Santé et de la Recherche Médicale (INSERM) U938, 75012 Paris, France
- Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, 75005 Paris, France
- Department of Endocrinology and Oncology Biochemistry, Pitié-Salpetrière Hospital, 75013 Paris, France
- Correspondence: ; Tel.: +33-(0)1-42-16-20-39
| |
Collapse
|
10
|
Jia Q, Wang Y, Zhu J, Yu H, Tong X. A literature review on lactopontin and its roles in early life. Transl Pediatr 2021; 10:1924-1931. [PMID: 34430441 PMCID: PMC8349962 DOI: 10.21037/tp-21-293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/14/2021] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Our study aims to review the functions and possible mechanisms of lactopontin (LPN) in early life. BACKGROUND Human milk proteins provide a variety of protection and health benefits in early life. One of these multifunctional proteins is LPN, which is osteopontin (OPN) derived from milk. METHODS Information used to write this paper was collected from Uniprot, PubMed, and Google Scholar, including in vitro, in vivo, and clinical studies. CONCLUSIONS LPN is a highly phosphorylated, O-glycosylated acidic protein and a unique type of OPN, as it presents at the highest concentration and a higher degree of posttranslational modifications (PTMs) in human milk than other tissues and excretions. LPN is present in milk and the intestinal tracts of infants after consumption as a mixture of intact protein and peptides, which can bind diverse integrin and receptors in the target cell and drive downstream signaling pathways. LPN is found to play important roles in developing the immune, intestinal and nervous systems in early life. Moreover, LPN has also shown to support preterm infants' health when they are especially vulnerable after delivery via animal studies. Additionally, LPN can form protein complex with another milk bioactive protein, lactoferrin (LF), to withstand proteolysis and perform more efficient biological activity. Therefore, LPN showed great potential for early life while more clinical trials and evidence are still emergying.
Collapse
Affiliation(s)
- Qiong Jia
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yiran Wang
- Department of Nutritional and Functional Assessment, Beijing Institute of Nutritional Resources, Beijing, China
| | - Jing Zhu
- Department of Nutritional and Functional Assessment, Beijing Institute of Nutritional Resources, Beijing, China
| | - Huanling Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing, China
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
11
|
Schytte GN, Christensen B, Bregenov I, Kjøge K, Scavenius C, Petersen SV, Enghild JJ, Sørensen ES. FAM20C phosphorylation of the RGDSVVYGLR motif in osteopontin inhibits interaction with the αvβ3 integrin. J Cell Biochem 2020; 121:4809-4818. [PMID: 32115754 DOI: 10.1002/jcb.29708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/10/2020] [Indexed: 01/13/2023]
Abstract
Osteopontin (OPN) is a ubiquitously expressed, multifunctional, and highly phosphorylated protein. OPN contains two neighboring integrin-binding motifs, RGD and SVVYGLR, which mediate interaction with cells. Phosphorylation and proteolytic processing affect the integrin-binding activities of OPN. Here we report that the kinase, FAM20C, phosphorylates Ser146 in the 143 RGDSVVYGLR152 motif of OPN and that Ser146 is phosphorylated in vivo in human and bovine milk. Ser146 is located right next to the RGD motif and close by the regulatory thrombin and plasmin cleavage sites in the OPN sequence. Phosphorylation of Ser146 could potentially affect the proteolytic processing and the integrin-binding activities of OPN. We show that phosphorylation of Ser146 does not affect the susceptibility of OPN for thrombin or plasmin cleavage. However, phosphorylation of Ser146 significantly reduces the RGD-mediated interaction with the αv β3 integrin in MDA-MB-435 and Moαv cells. This suggests a new mechanism by which specific phosphorylation of OPN can regulate interaction with the αv β3 integrin and thereby affect OPN-cell interaction.
Collapse
Affiliation(s)
- Gitte N Schytte
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Brian Christensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Ida Bregenov
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Katarzyna Kjøge
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | | | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Schytte GN, Christensen B, Bregenov I, Sørensen ES. Ras-transformation reduce FAM20C expression and osteopontin phosphorylation. Biosci Rep 2020; 40:BSR20194378. [PMID: 32830861 PMCID: PMC7494989 DOI: 10.1042/bsr20194378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 07/07/2020] [Accepted: 08/19/2020] [Indexed: 12/30/2022] Open
Abstract
Family with sequence similarity 20, member C (FAM20C) is the main kinase of secreted phosphoproteins, including the multifunctional protein and cytokine, osteopontin (OPN). The phosphorylation of OPN varies greatly among cell types, tissues and species, and the different phospho-isoforms contribute to the multifunctionality of the protein. Expression of OPN is increased in human malignancies, and less phosphorylated isoforms of the protein have been associated with this phenotype. Here, we compared OPN from ras-transformed fibroblasts with that from their non-transformed parental cells, and found that OPN was less phosphorylated after ras-transformation. Furthermore, we demonstrated that expression of FAM20C mRNA was reduced five-fold in ras-transformed fibroblasts compared with non-transformed fibroblasts. Transfection with FAM20C of the ras-transformed fibroblasts restored the FAM20C mRNA expression but the phosphorylation of OPN was not increased proportionally. Likewise, the mRNA level of FAM20C was reduced in the malignant ras-transformed mammary cell line MCF10ACA1a compared with its non-transformed parental cell line MCF10A. These results suggest that expression of the FAM20C kinase is reduced after oncogenic ras-transformation, which potentially affects the phosphorylation of secreted phosphoproteins.
Collapse
Affiliation(s)
- Gitte N Schytte
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Aarhus, Denmark
| | - Brian Christensen
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Aarhus, Denmark
| | - Ida Bregenov
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Aarhus, Denmark
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
Melanitou E. Investigation of type 1 diabetes in NOD mice knockout for the osteopontin gene. Gene 2020; 753:144785. [PMID: 32445922 DOI: 10.1016/j.gene.2020.144785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 05/07/2020] [Accepted: 05/15/2020] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Type 1 diabetes onset is preceded by a pre-inflammatory stage leading to insulitis and followed by targeted destruction of the insulin-producing beta cells of the pancreas. Osteopontin (OPN) is a secreted phosphoprotein with cytokine properties, implicated in many physiological and pathological processes, including infection and autoimmunity. We have previously identified up-regulated osteopontin transcripts in the pancreatic lymph nodes of the NOD (Non-Obese Diabetic) mouse at the pre-diabetic stages. Investigating the underlined disease initiating mechanisms may well contribute to the development of novel preventive therapies. Our aim was to construct opn null mice in a NOD autoimmune-prone genetic background and address the pathogenic or protective role of the osteopontin molecule in the early stages of type 1 diabetes. METHODS We generated opn null mutant mice in a NOD genetic background by serial backcrossing to the existing C57BL/6 opn knockout strain. The presence of opn wild type or null alleles in the congenic lines was evaluated by PCR amplification. We used NOD opn-null mice to assess the phenotypic evolution of type 1 diabetes. The presence of OPN in the serum was evaluated by ELISA and by immunostaining on the mouse tissues. The primary gene structure of the NOD opn encoding gene and protein sequences were compared to the known alleles of other mouse strains. Evaluation of Single Nucleotide Polymorphisms (SNPs) variation between opn alleles of the opn gene is reported. RESULTS In the absence of OPN, type 1 diabetes is accelerated, suggesting a protective role of this cytokine on the insulin-producing cells of the pancreatic islets. Conversely, in the presence of the opn gene, an increase of the OPN protein in the serum of young NOD mice indicates that this molecule might be involved in the immune regulatory events taking place at early stages, prior to disease onset. Our data support that OPN acts as a positive regulator of the early islet autoimmune damage, possibly by a shift of the steady-state of T1D pathogenesis. We report that the OPN protein structure of the NOD/ShiLtJ strain corresponds to the a-type allele of the osteopontin gene. Comparative analysis of the single nucleotide polymorphisms between the a-type and b-type alleles indicates that the majority of variations are within the non-coding regions of the gene. CONCLUSIONS The construction of opn null mice in an autoimmune genetic background (NOD.B6.Cg-spp1-/-) provides important tools for the study of the implication of the OPN in type 1 diabetes, offering the possibility to address the significance of this molecule as an early marker of the disease and as a therapeutic agent in preclinical studies.
Collapse
Affiliation(s)
- Evie Melanitou
- Department of Parasites and Insect Vectors, Institut Pasteur, 25-28 rue du Dr Roux, 75724 Paris, Cedex 15, France.
| |
Collapse
|
14
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
15
|
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.
Collapse
Affiliation(s)
- Zoe Shin Yee Lok
- Department of Surgery, School of Clinical Sciences, Monash Health, Clayton, Australia (Z.S.Y.L.)
| | - Alicia N Lyle
- From the Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (A.N.L.)
| |
Collapse
|
16
|
Anan G, Yoneyama T, Noro D, Tobisawa Y, Hatakeyama S, Sutoh Yoneyama M, Yamamoto H, Imai A, Iwamura H, Kohada Y, Mikami J, Ito J, Kaiho Y, Yoneyama T, Hashimoto Y, Sato M, Ohyama C. The Impact of Glycosylation of Osteopontin on Urinary Stone Formation. Int J Mol Sci 2019; 21:ijms21010093. [PMID: 31877766 PMCID: PMC6982307 DOI: 10.3390/ijms21010093] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/31/2023] Open
Abstract
Osteopontin (OPN) is a matrix glycoprotein of urinary calculi. This study aims to identify the role of aberrant glycosylation of OPN in urolithiasis. We retrospectively measured urinary glycosylated OPN normalized by urinary full-length-OPN levels in 110 urolithiasis patients and 157 healthy volunteers and 21 patients were prospectively longitudinal follow-up during stone treatment. The urinary full-length-OPN levels were measured using enzyme-linked immunosorbent assay and glycosylated OPN was measured using a lectin array and lectin blotting. The assays were evaluated using the area under the receiver operating characteristics curve to discriminate stone forming urolithiasis patients. In the retrospective cohort, urinary Gal3C-S lectin reactive- (Gal3C-S-) OPN/full-length-OPN, was significantly higher in the stone forming urolithiasis patients than in the healthy volunteers (p < 0.0001), with good discrimination (AUC, 0.953), 90% sensitivity, and 92% specificity. The Lycopersicon esculentum lectin analysis of urinary full-length-OPN showed that urinary full-length-OPN in stone forming urolithiasis patients had a polyLacNAc structure that was not observed in healthy volunteers. In the prospective longitudinal follow-up study, 92.8% of the stone-free urolithiasis group had Gal3C-S-OPN/full-length-OPN levels below the cutoff value after ureteroscopic lithotripsy (URS), whereas 71.4% of the residual-stone urolithiasis group did not show decreased levels after URS. Therefore, Gal3C-S-OPN/full-length-OPN levels could be used as a urolithiasis biomarker.
Collapse
Affiliation(s)
- Go Anan
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Tohru Yoneyama
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
- Correspondence: ; Tel.: +81-172-39-5091
| | - Daisuke Noro
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Yuki Tobisawa
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Shingo Hatakeyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Mihoko Sutoh Yoneyama
- Department of Cancer Immunology and Cell Biology, Oyokyo Kidney Research Institute, Hirosaki, Aomori 036-8243, Japan
| | - Hayato Yamamoto
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Atsushi Imai
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Hiromichi Iwamura
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Yuki Kohada
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Jotaro Mikami
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Jun Ito
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Yasuhiro Kaiho
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Takahiro Yoneyama
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Yasuhiro Hashimoto
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
| | - Makoto Sato
- Department of Urology, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan;
- Department of Advanced Transplant and Regenerative Medicine, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| |
Collapse
|
17
|
Christensen B, Karlsen NJ, Jørgensen SDS, Jacobsen LN, Ostenfeld MS, Petersen SV, Müllertz A, Sørensen ES. Milk osteopontin retains integrin-binding activity after in vitro gastrointestinal transit. J Dairy Sci 2019; 103:42-51. [PMID: 31733850 DOI: 10.3168/jds.2019-17212] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/30/2019] [Indexed: 11/19/2022]
Abstract
Osteopontin (OPN) is a multifunctional protein highly expressed in milk, where it is hypothesized to be involved in immunological signaling via the conserved Arg-Gly-Asp (RGD) integrin-binding sequence. Intervention studies have indicated beneficial effects of orally administered OPN in animal and human infants, but the mechanisms underlying these effects are not well described. To induce physiological effects, OPN must resist gastrointestinal transit in a bioactive form. In this study, we subjected bovine milk OPN to in vitro gastrointestinal transit, and characterized the generated fragments using monoclonal antibody and mass spectrometric analyses. We found that the fragment Trp27-Phe151 containing the integrin-binding RGD sequence resisted in vitro gastric digestion. This resistance was dependent on glycosylation of threonine residues near the integrin-binding sequence in both human and bovine milk OPN. Furthermore, the fragment Trp27-Phe151 retained the ability to interact with integrins in an RGD-dependent process. These results suggest a mechanism for how ingested milk OPN can induce physiological effects via integrin signaling in the intestine.
Collapse
Affiliation(s)
- B Christensen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark; iFood, Aarhus University Center for Innovative Food Research, DK-8000 Aarhus, Denmark
| | - N J Karlsen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - S D S Jørgensen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - L N Jacobsen
- Arla Foods Ingredients Group P/S, DK-8260 Viby J, Denmark
| | - M S Ostenfeld
- Arla Foods Ingredients Group P/S, DK-8260 Viby J, Denmark
| | - S V Petersen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - A Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - E S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark; iFood, Aarhus University Center for Innovative Food Research, DK-8000 Aarhus, Denmark.
| |
Collapse
|
18
|
Jiang R, Lönnerdal B. Osteopontin in human milk and infant formula affects infant plasma osteopontin concentrations. Pediatr Res 2019; 85:502-505. [PMID: 30636771 DOI: 10.1038/s41390-018-0271-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Osteopontin (OPN), a multifunctional protein, is present abundantly in human milk, but not in bovine milk and infant formulas. A recent randomized clinical trial showed that supplementing infant formula with bovine milk OPN (bOPN) resulted in better immune outcomes. METHODS Human milk OPN (hOPN) concentrations were analyzed by ELISA. Plasma samples were obtained from infants receiving one of four treatments: breast milk (BF), unsupplemented formula (F0), formula supplemented with 65 mg/L bOPN (F65), or with 130 mg/L bOPN (F130). Plasma samples were analyzed for hOPN and bOPN by ELISA. RESULTS The hOPN concentration was high in early lactation (D1 to D8), decreased gradually after D9, and deceased significantly after 1 month. At 4 and 6 months, higher levels of hOPN were found in plasma samples from the BF, F65, and F130 groups than in samples from the F0 group; the plasma bOPN concentration in the F130 group was greater than that in the F65 group. CONCLUSION Dynamic changes in the concentration of milk OPN may reflect infant needs for different amounts of milk OPN for various functions at different developmental stages. Supplemental bOPN in infant formula may exert its beneficial effects by increasing endogenous OPN in plasma.
Collapse
Affiliation(s)
- Rulan Jiang
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
19
|
The Phylogeny of Osteopontin-Analysis of the Protein Sequence. Int J Mol Sci 2018; 19:ijms19092557. [PMID: 30154395 PMCID: PMC6164354 DOI: 10.3390/ijms19092557] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022] Open
Abstract
Osteopontin (OPN) is important for tissue remodeling, cellular immune responses, and calcium homeostasis in milk and urine. In pathophysiology, the biomolecule contributes to the progression of multiple cancers. Phylogenetic analysis of 202 osteopontin protein sequences identifies a core block of integrin-binding sites in the center of the protein, which is well conserved. Remarkably, the length of this block varies among species, resulting in differing distances between motifs within. The amino acid sequence SSEE is a candidate phosphorylation site. Two copies of it reside in the far N-terminus and are variably affected by alternative splicing in humans. Between those motifs, birds and reptiles have a histidine-rich domain, which is absent from other species. Just downstream from the thrombin cleavage site, the common motif (Q/I)(Y/S/V)(P/H/Y)D(A/V)(T/S)EED(L/E)(-/S)T has been hitherto unrecognized. While well preserved, it is yet without assigned function. The far C-terminus, although very different between Reptilia/Aves on the one hand and Mammals on the other, is highly conserved within each group of species, suggesting important functional roles that remain to be mapped. Taxonomic variations in the osteopontin sequence include a lack of about 20 amino acids in the downstream portion, a small unique sequence stretch C-terminally, a lack of six amino acids just upstream of the RGD motifs, and variable length insertions far C-terminally.
Collapse
|
20
|
Biological role of site-specific O-glycosylation in cell adhesion activity and phosphorylation of osteopontin. Biochem J 2018; 475:1583-1595. [PMID: 29626154 DOI: 10.1042/bcj20170205] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 02/08/2018] [Accepted: 04/03/2018] [Indexed: 01/13/2023]
Abstract
Osteopontin (OPN) is an extracellular glycosylated phosphoprotein that promotes cell adhesion by interacting with several integrin receptors. We previously reported that an OPN mutant lacking five O-glycosylation sites (Thr134/Thr138/Thr143/Thr147/Thr152) in the threonine/proline-rich region increased cell adhesion activity and phosphorylation compared with the wild type. However, the role of O-glycosylation in cell adhesion activity and phosphorylation of OPN remains to be clarified. Here, we show that site-specific O-glycosylation in the threonine/proline-rich region of OPN affects its cell adhesion activity and phosphorylation independently and/or synergistically. Using site-directed mutagenesis, we found that OPN mutants with substitution sets of Thr134/Thr138 or Thr143/Thr147/Thr152 had decreased and increased cell adhesion activity, respectively. In contrast, the introduction of a single mutation into the O-glycosylation sites had no effect on OPN cell adhesion activity. An adhesion assay using function-blocking antibodies against αvβ3 and β1 integrins, as well as αvβ3 integrin-overexpressing A549 cells, revealed that site-specific O-glycosylation affected the association of OPN with the two integrins. Phosphorylation analyses using phos-tag and LC-MS/MS indicated that phosphorylation levels and sites were influenced by the O-glycosylation status, although the number of O-glycosylation sites was not correlated with the phosphorylation level in OPN. Furthermore, a correlation analysis between phosphorylation level and cell adhesion activity in OPN mutants with the site-specific O-glycosylation showed that they were not always correlated. These results provide conclusive evidence of a novel regulatory mechanism of cell adhesion activity and phosphorylation of OPN by site-specific O-glycosylation.
Collapse
|
21
|
Foster BL, Ao M, Salmon CR, Chavez MB, Kolli TN, Tran AB, Chu EY, Kantovitz KR, Yadav M, Narisawa S, Millán JL, Nociti FH, Somerman MJ. Osteopontin regulates dentin and alveolar bone development and mineralization. Bone 2018; 107:196-207. [PMID: 29313816 PMCID: PMC5803363 DOI: 10.1016/j.bone.2017.12.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/09/2017] [Accepted: 12/03/2017] [Indexed: 01/09/2023]
Abstract
The periodontal complex is essential for tooth attachment and function and includes the mineralized tissues, cementum and alveolar bone, separated by the unmineralized periodontal ligament (PDL). To gain insights into factors regulating cementum-PDL and bone-PDL borders and protecting against ectopic calcification within the PDL, we employed a proteomic approach to analyze PDL tissue from progressive ankylosis knock-out (Ank-/-) mice, featuring reduced PPi, rapid cementogenesis, and excessive acellular cementum. Using this approach, we identified the matrix protein osteopontin (Spp1/OPN) as an elevated factor of interest in Ank-/- mouse molar PDL. We studied the role of OPN in dental and periodontal development and function. During tooth development in wild-type (WT) mice, Spp1 mRNA was transiently expressed by cementoblasts and strongly by alveolar bone osteoblasts. Developmental analysis from 14 to 240days postnatal (dpn) indicated normal histological structures in Spp1-/- comparable to WT control mice. Microcomputed tomography (micro-CT) analysis at 30 and 90dpn revealed significantly increased volumes and tissue mineral densities of Spp1-/- mouse dentin and alveolar bone, while pulp and PDL volumes were decreased and tissue densities were increased. However, acellular cementum growth was unaltered in Spp1-/- mice. Quantitative PCR of periodontal-derived mRNA failed to identify potential local compensators influencing cementum in Spp1-/- vs. WT mice at 26dpn. We genetically deleted Spp1 on the Ank-/- mouse background to determine whether increased Spp1/OPN was regulating periodontal tissues when the PDL space is challenged by hypercementosis in Ank-/- mice. Ank-/-; Spp1-/- double deficient mice did not exhibit greater hypercementosis than that in Ank-/- mice. Based on these data, we conclude that OPN has a non-redundant role regulating formation and mineralization of dentin and bone, influences tissue properties of PDL and pulp, but does not control acellular cementum apposition. These findings may inform therapies targeted at controlling soft tissue calcification.
Collapse
Affiliation(s)
- B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| | - M Ao
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - C R Salmon
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - M B Chavez
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - T N Kolli
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - A B Tran
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - E Y Chu
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - K R Kantovitz
- Department of Dental Materials, São Leopoldo Mandic Research Center, Campinas, São Paulo, Brazil
| | - M Yadav
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - S Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - J L Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - F H Nociti
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
22
|
Hoac B, Susan-Resiga D, Essalmani R, Marcinkiweicz E, Seidah NG, McKee MD. Osteopontin as a novel substrate for the proprotein convertase 5/6 (PCSK5) in bone. Bone 2018; 107:45-55. [PMID: 29126984 DOI: 10.1016/j.bone.2017.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 11/05/2017] [Accepted: 11/06/2017] [Indexed: 12/30/2022]
Abstract
Seven proprotein convertases cleave the basic amino acid consensus sequence K/R-Xn-K/R↓ (where n=0, 2, 4 or 6 variable amino acids) to activate precursor proteins. Despite similarities in substrate specificity, basic amino acid-specific proprotein convertases have a distinct tissue distribution allowing for enzymatic actions on tissue-resident substrates. Proprotein convertase 5/6 (PC5/6) has two splice variants - soluble PC5/6A and membrane-bound PC5/6B - and is expressed during mouse development in many tissues including bone and tooth, but little is known about the substrates for PC5/6 therein. Osteopontin (OPN) is an abundant bone extracellular matrix protein with roles in mineralization, cell adhesion and cell migration, and it has putative consensus sequence sites for cleavage by PC5/6, which may modify its function in bone. Since PC5/6-knockout mouse embryos show developmental abnormalities, and reduced overall mineralization, we designed this study to determine whether OPN is a substrate of PC5/6. In silico analysis of OPN protein sequences identified four potential PC5/6 consensus cleavage sites in human OPN, and three sites - including a noncanonical sequence - in mouse OPN. Ex vivo co-transfections with human OPN revealed complete OPN cleavage reducing full-length OPN (~70kDa) to an N-terminal fragment migrating at ~50kDa and two C-terminal fragments at ~18kDa and ~16kDa. Direct cleavage of OPN by PC5/6A - the predominant isoform expressed in human osteoblast cells - was confirmed by cell-free enzyme-substrate assays and by mass spectrometry. The latter was also used to investigate potential cleavage sites. Co-transfections of PC5/6 and mouse OPN showed partial cleavage of OPN into a C-terminal OPN fragment migrating at ~30kDa and an N-terminal fragment migrating at ~29kDa. Micro-computed tomography of PC5/6-knockout embryos at E18.5 confirmed a reduction in mineralized bone, and in situ hybridization performed on cryo-sections of normal mouse bone using Pcsk5 and Opn anti-sense and control-sense cRNA probes indicated the co-localization of the expression of these genes in bone cells. This mRNA expression profile was supported by semi-quantitative RT-PCR using osteoblast primary cultures, and cultured MC3T3-E1 osteoblast and MLO-Y4 osteocyte cell lines. Immunoblotting for OPN from mouse bone extracts showed altered OPN processing in PC5/6-knockout mice compared to wildtype mice. OPN fragments migrated at ~25kDa and ~16kDa in wildtype bone and were not present in PC5/6-deficient bone. In conclusion, this study demonstrates that Pcsk5 is expressed in bone-forming cells, and that OPN is a novel substrate for PC5/6. Cleavage of OPN by PC5/6 may modify the function of OPN in bone and/or modulate other enzymatic cleavages of OPN, leading to alterations in the bone phenotype.
Collapse
Affiliation(s)
- Betty Hoac
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Delia Susan-Resiga
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute, Affiliated with the University of Montreal, Montreal, QC, Canada
| | - Rachid Essalmani
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute, Affiliated with the University of Montreal, Montreal, QC, Canada
| | - Edwige Marcinkiweicz
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute, Affiliated with the University of Montreal, Montreal, QC, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute, Affiliated with the University of Montreal, Montreal, QC, Canada
| | - Marc D McKee
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
23
|
Ao M, Chavez MB, Chu EY, Hemstreet KC, Yin Y, Yadav MC, Millán JL, Fisher LW, Goldberg HA, Somerman MJ, Foster BL. Overlapping functions of bone sialoprotein and pyrophosphate regulators in directing cementogenesis. Bone 2017; 105:134-147. [PMID: 28866368 PMCID: PMC5730356 DOI: 10.1016/j.bone.2017.08.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
Although acellular cementum is essential for tooth attachment, factors directing its development and regeneration remain poorly understood. Inorganic pyrophosphate (PPi), a mineralization inhibitor, is a key regulator of cementum formation: tissue-nonspecific alkaline phosphatase (Alpl/TNAP) null mice (increased PPi) feature deficient cementum, while progressive ankylosis protein (Ank/ANK) null mice (decreased PPi) feature increased cementum. Bone sialoprotein (Bsp/BSP) and osteopontin (Spp1/OPN) are multifunctional extracellular matrix components of cementum proposed to have direct and indirect effects on cell activities and mineralization. Studies on dentoalveolar development of Bsp knockout (Bsp-/-) mice revealed severely reduced acellular cementum, however underlying mechanisms remain unclear. The similarity in defective cementum phenotypes between Bsp-/- mice and Alpl-/- mice (the latter featuring elevated PPi and OPN), prompted us to examine whether BSP is operating by modulating PPi-associated genes. Genetic ablation of Bsp caused a 2-fold increase in circulating PPi, altered mRNA expression of Alpl, Spp1, and Ank, and increased OPN protein in the periodontia. Generation of a Bsp knock-out (KO) cementoblast cell line revealed significantly decreased mineralization capacity, 50% increased PPi in culture media, and increased Spp1 and Ank mRNA expression. While addition of 2μg/ml recombinant BSP altered Spp1, Ank, and Enpp1 expression in cementoblasts, changes resulting from this dose were not dependent on the integrin-binding RGD motif or MAPK/ERK signaling pathway. Decreasing PPi by genetic ablation of Ank on the Bsp-/- mouse background reestablished cementum formation, allowing >3-fold increased acellular cementum volume compared to wild-type (WT). However, deleting Ank did not fully compensate for the absence of BSP. Bsp-/-; Ank-/- double-deficient mice exhibited mean 20-27% reduced cementum thickness and volume compared to Ank-/- mice. From these data, we conclude that the perturbations in PPi metabolism are not solely driving the cementum pathology in Bsp-/- mice, and that PPi is more potent than BSP as a cementum regulator, as shown by the ability to override loss of BSP by lowering PPi. We propose that BSP and PPi work in concert to direct mineralization in cementum and likely other mineralized tissues.
Collapse
Affiliation(s)
- M Ao
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M B Chavez
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - E Y Chu
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - K C Hemstreet
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Y Yin
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M C Yadav
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - J L Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - L W Fisher
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - H A Goldberg
- Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - B L Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
24
|
Hamias R, Rudich A, Greenberg G, Szendro G, Wolak T. Angiotensin 1–7, but not the thrombin-cleaved osteopontin C-terminal fragment, attenuates osteopontin-mediated macrophage-induced endothelial-cell inflammation. Inflamm Res 2017; 67:265-275. [DOI: 10.1007/s00011-017-1120-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 11/11/2017] [Accepted: 11/22/2017] [Indexed: 12/22/2022] Open
|
25
|
Osteopontin at the Crossroads of Inflammation and Tumor Progression. Mediators Inflamm 2017; 2017:4049098. [PMID: 28769537 PMCID: PMC5523273 DOI: 10.1155/2017/4049098] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/04/2017] [Indexed: 12/13/2022] Open
Abstract
Complex interactions between tumor and host cells regulate systemic tumor dissemination, a process that begins early at the primary tumor site and goes on until tumor cells detach themselves from the tumor mass and start migrating into the blood or lymphatic vessels. Metastatic cells colonize the target organs and are capable of surviving and growing at distant sites. In this context, osteopontin (OPN) appears to be a key determinant of the crosstalk between cancer cells and the host microenvironment, which in turn modulates immune evasion. OPN is overexpressed in several human carcinomas and has been implicated in inflammation, tumor progression, and metastasis. Thus, it represents one of the most attracting targets for cancer therapy. Within the tumor mass, OPN is secreted in various forms either by the tumor itself or by stroma cells, and it can exert either pro- or antitumorigenic effects according to the cell type and tumor microenvironment. Thus, targeting OPN for therapeutic purposes needs to take into account the heterogeneous functions of the multiple OPN forms with regard to cancer formation and progression. In this review, we will describe the role of systemic, tumor-derived, and stroma-derived OPN, highlighting its pivotal role at the crossroads of inflammation and tumor progression.
Collapse
|
26
|
An J, Briggs TA, Dumax-Vorzet A, Alarcón-Riquelme ME, Belot A, Beresford M, Bruce IN, Carvalho C, Chaperot L, Frostegård J, Plumas J, Rice GI, Vyse TJ, Wiedeman A, Crow YJ, Elkon KB. Tartrate-Resistant Acid Phosphatase Deficiency in the Predisposition to Systemic Lupus Erythematosus. Arthritis Rheumatol 2016; 69:131-142. [PMID: 27390188 DOI: 10.1002/art.39810] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 06/30/2016] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Mutations in the ACP5 gene, which encodes tartrate-resistant acid phosphatase (TRAP), cause the immuno-osseous disorder spondyloenchondrodysplasia, which includes as disease features systemic lupus erythematosus (SLE) and a type I interferon (IFN) signature. Our aims were to identify TRAP substrates, determine the consequences of TRAP deficiency in immune cells, and assess whether ACP5 mutations are enriched in sporadic cases of SLE. METHODS Interaction between TRAP and its binding partners was tested by a yeast 2-hybrid screening, confocal microscopy, and immunoprecipitation/Western blotting. TRAP knockdown was performed using small interfering RNA. Phosphorylation of osteopontin (OPN) was analyzed by mass spectrometry. Nucleotide sequence analysis of ACP5 was performed by Sanger sequencing or next-generation sequencing. RESULTS TRAP and OPN colocalized and interacted in human macrophages and plasmacytoid dendritic cells (PDCs). TRAP dephosphorylated 3 serine residues on specific OPN peptides. TRAP knockdown resulted in increased OPN phosphorylation and increased nuclear translocation of IRF7 and P65, with resultant heightened expression of IFN-stimulated genes and IL6 and TNF following Toll-like receptor 9 stimulation. An excess of heterozygous ACP5 missense variants was observed in SLE compared to controls (P = 0.04), and transfection experiments revealed a significant reduction in TRAP activity in a number of variants. CONCLUSION Our findings indicate that TRAP and OPN colocalize and that OPN is a substrate for TRAP in human immune cells. TRAP deficiency in PDCs leads to increased IFNα production, providing at least a partial explanation for how ACP5 mutations cause lupus in the context of spondyloenchondrodysplasia. Detection of ACP5 missense variants in a lupus cohort suggests that impaired TRAP functioning may increase susceptibility to sporadic lupus.
Collapse
Affiliation(s)
- Jie An
- University of Washington, Seattle
| | - Tracy A Briggs
- University of Manchester and St. Mary's Hospital, Central Manchester Foundation Trust, Manchester, UK
| | | | - Marta E Alarcón-Riquelme
- Universidad de Granada-Junta de Andalucía, Granada, Spain, and Oklahoma Medical Research Foundation, Oklahoma City
| | - Alexandre Belot
- Pediatric Rheumatology Unit, Femme Mère Enfant Hospital, Hospices Civils de Lyon, INSERM U1111, University of Lyon, Lyon, France
| | - Michael Beresford
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Ian N Bruce
- University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Claudia Carvalho
- Universidade do Porto, Abel Salazar Institute of Biomedical Sciences, Porto, Portugal
| | - Laurence Chaperot
- INSERM U823/UJF/EFS, UGA, INSERM U1209, CNRS 5309, Immunobiology and Immunotherapy of Cancers and Chronic Diseases, Grenoble, France
| | | | - Joel Plumas
- INSERM U823/UJF/EFS, UGA, INSERM U1209, CNRS 5309, Immunobiology and Immunotherapy of Cancers and Chronic Diseases, Grenoble, France
| | | | | | | | - Yanick J Crow
- University of Manchester, Manchester, UK, and Institut Imagine, Laboratory of Neurogenetics and Neuroinflammation, Paris, France
| | | |
Collapse
|
27
|
|
28
|
Christensen B, Zachariae ED, Scavenius C, Kløverpris S, Oxvig C, Petersen SV, Enghild JJ, Sørensen ES. Transglutaminase 2-Catalyzed Intramolecular Cross-Linking of Osteopontin. Biochemistry 2016; 55:294-303. [PMID: 26678563 DOI: 10.1021/acs.biochem.5b01153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Osteopontin (OPN) is a multifunctional integrin-binding protein present in several tissues and body fluids. OPN is a substrate for the enzyme transglutaminase 2 (TG2), which catalyzes inter- and intramolecular cross-linking affecting the biological activity of the protein. Polymerization of OPN by intermolecular cross-linking has mostly been studied using relatively high TG2 concentrations, whereas the effect of lower concentrations of TG2 has remained unexplored. Here we show that TG2 at physiologically relevant concentrations predominantly catalyzes the formation of intramolecular cross-links in OPN. By site-directed mutagenesis and mass spectrometry, we demonstrate that Gln(42) and Gln(193) serve as the primary amine acceptor sites for isopeptide bond formation. We find that Gln(42) predominantly is linked to Lys(4) and that Gln(193) participates in a cross-link with Lys(154), Lys(157), or Lys(231). The formation of specific isopeptide bonds was not dependent on OPN phosphorylation, and similar patterns of cross-linking were observed in human and mouse OPN. Furthermore, we find that OPN purified from human urine contains the Lys(154)-Gln(193) isopeptide bond, indicating that intramolecular cross-linking of OPN occurs in vivo. Collectively, these data suggest that specific intramolecular cross-linking in the N- and C-terminal parts of OPN is most likely the dominant step in TG2-catalyzed modification of OPN.
Collapse
Affiliation(s)
- Brian Christensen
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Elias D Zachariae
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Søren Kløverpris
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark
| | - Steen V Petersen
- Department of Biomedicine, Aarhus University , 8000 Aarhus C, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University , 8000 Aarhus C, Denmark
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University , 8000 Aarhus C, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University , 8000 Aarhus C, Denmark
| |
Collapse
|
29
|
Abstract
Since its initial identification as one of the genes most highly upregulated upon T-cell activation, osteopontin (or Eta-1, as it was designated then) has been demonstrated to have many roles in the regulation of the immune response on multiple levels. It contributes to the development of immune-mediated and inflammatory diseases, and it regulates the host response to infection. In some cases, the mechanisms of these effects have been elucidated, while other mechanistic functions of the protein remain obscure. The protein itself makes these analyses complex, since it binds to a series of different integrins, and in addition to its classically secreted form, an intracellular form of osteopontin has been identified, which participates in several aspects of immune regulation. In this review, we focus on the role of osteopontin in a series of immune-related diseases, particularly those where significant advances have been made in recent years: multiple sclerosis, rheumatoid arthritis, lupus and related diseases, Sjögren's disease, colitis, and 1 area of inflammatory pathology, alcoholic and nonalcoholic liver diseases. A recurring theme in these diseases is a link between osteopontin and pathogenic T cells, particularly T helper 17 cells, where osteopontin produced by dendritic cells supports IL-17 expression, contributing to pathology. In addition, a role for osteopontin in B-cell differentiation is becoming clear. In general, osteopontin contributes to pathology in these diseases, but there are examples where it has a protective role; deciphering the mechanisms underlying these differences and the specific receptors for osteopontin will be a research challenge for the future. Aside from its newly discovered role in the development of Sjögren's disease, the role of osteopontin in inflammatory conditions in the oral cavity is still poorly understood. Elucidation of this role will be of interest.
Collapse
Affiliation(s)
- S R Rittling
- The Forsyth Institute, Cambridge, MA, USA Harvard School of Dental Medicine, Boston, MA, USA
| | - R Singh
- The Forsyth Institute, Cambridge, MA, USA
| |
Collapse
|
30
|
Martín-Alonso M, García-Redondo AB, Guo D, Camafeita E, Martínez F, Alfranca A, Méndez-Barbero N, Pollán Á, Sánchez-Camacho C, Denhardt DT, Seiki M, Vázquez J, Salaices M, Redondo JM, Milewicz D, Arroyo AG. Deficiency of MMP17/MT4-MMP proteolytic activity predisposes to aortic aneurysm in mice. Circ Res 2015; 117:e13-26. [PMID: 25963716 DOI: 10.1161/circresaha.117.305108] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 05/08/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Aortic dissection or rupture resulting from aneurysm causes 1% to 2% of deaths in developed countries. These disorders are associated with mutations in genes that affect vascular smooth muscle cell differentiation and contractility or extracellular matrix composition and assembly. However, as many as 75% of patients with a family history of aortic aneurysms do not have an identified genetic syndrome. OBJECTIVE To determine the role of the protease MMP17/MT4-MMP in the arterial wall and its possible relevance in human aortic pathology. METHODS AND RESULTS Screening of patients with inherited thoracic aortic aneurysms and dissections identified a missense mutation (R373H) in the MMP17 gene that prevented the expression of the protease in human transfected cells. Using a loss-of-function genetic mouse model, we demonstrated that the lack of Mmp17 resulted in the presence of dysfunctional vascular smooth muscle cells and altered extracellular matrix in the vessel wall; and it led to increased susceptibility to angiotensin-II-induced thoracic aortic aneurysm. We also showed that Mmp17-mediated osteopontin cleavage regulated vascular smooth muscle cell maturation via c-Jun N-terminal kinase signaling during aorta wall development. Some features of the arterial phenotype were prevented by re-expression of catalytically active Mmp17 or the N-terminal osteopontin fragment in Mmp17-null neonates. CONCLUSIONS Mmp17 proteolytic activity regulates vascular smooth muscle cell phenotype in the arterial vessel wall, and its absence predisposes to thoracic aortic aneurysm in mice. The rescue of part of the vessel-wall phenotype by a lentiviral strategy opens avenues for therapeutic intervention in these life-threatening disorders.
Collapse
MESH Headings
- Adult
- Amino Acid Substitution
- Aortic Dissection/genetics
- Angiotensin II
- Animals
- Aorta/embryology
- Aorta/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/therapy
- Aortic Rupture/etiology
- Extracellular Matrix/pathology
- Extracellular Matrix Proteins/metabolism
- Genetic Predisposition to Disease
- Genetic Therapy
- Genetic Vectors/therapeutic use
- HEK293 Cells
- Humans
- Lentivirus/genetics
- Male
- Matrix Metalloproteinases, Membrane-Associated/chemistry
- Matrix Metalloproteinases, Membrane-Associated/deficiency
- Matrix Metalloproteinases, Membrane-Associated/genetics
- Matrix Metalloproteinases, Membrane-Associated/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/pathology
- Mutation, Missense
- Osteopontin/metabolism
- Protein Conformation
Collapse
Affiliation(s)
- Mara Martín-Alonso
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Ana B García-Redondo
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Dongchuan Guo
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Emilio Camafeita
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Fernando Martínez
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Arántzazu Alfranca
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Nerea Méndez-Barbero
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Ángela Pollán
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Cristina Sánchez-Camacho
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - David T Denhardt
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Motoharu Seiki
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Jesús Vázquez
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Mercedes Salaices
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Juan Miguel Redondo
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Dianna Milewicz
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.)
| | - Alicia G Arroyo
- From the Department of Vascular Biology and Inflammation (M.M.-A., A.A., N.M.-B., A.P., J.M.R., A.G.A.), Proteomics Unit (E.C., J.V.) and Bioinformatics Unit (F.M.), Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Department of Pharmacology/Nephrology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R., M.S.); Department of Internal Medicine, University of Texas Health Science Center at Houston, TX (D.G., D.M.); Department of Basic Biomedical Sciences, Universidad Europea de Madrid, Villaviciosa de Odón, Madrid, Spain (C.S.-C.); Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (D.T.D.); and Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan (M.S.).
| |
Collapse
|
31
|
Kläning E, Christensen B, Bajic G, Hoffmann SV, Jones NC, Callesen MM, Andersen GR, Sørensen ES, Vorup-Jensen T. Multiple low-affinity interactions support binding of human osteopontin to integrin αXβ2. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:930-8. [PMID: 25839998 DOI: 10.1016/j.bbapap.2015.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 03/18/2015] [Accepted: 03/22/2015] [Indexed: 01/11/2023]
Abstract
Integrin α(X)β(2) (also known as complement receptor 4, p150,95, or CD11c/CD18) is expressed in the cell membrane of myeloid leukocytes. α(X)β(2) has been reported to bind a large number of structurally unrelated ligands, often with a shared molecular character in the presence of polyanionic stretches in poorly folded proteins or glucosaminoglycans. Nevertheless, it is unclear what chemical sources of polyanionicity enable the binding by α(X)β(2). Osteopontin (OPN) is an intrinsically disordered protein, which facilitates phagocytosis via the integrin α(X)β(2). Unlike for other integrins, neither the RGD nor the SVVYGLR motifs account for this binding, and the molecular basis of OPN binding by α(X)β(2) remains uncharacterized. Here, we show that the monovalent interactions between the ligand-binding domain of α(X)β(2) and OPN, its fragments, or caseins are weak, with dissociation constants higher than 10(-5)M but with high apparent stoichiometries. From comparison with cell adhesion studies, the discrimination between α(X)β(2) ligands and non-ligands appears to rely on these apparent stoichiometries in a way, which involves glutamate rather than aspartate side chains. Surprisingly, the extensive, negatively charged phosphorylation of OPN is not contributing to α(X)β(2) binding. Furthermore, synchrotron radiation circular spectroscopy excludes that the phosphorylation affects the general folding of OPN. Taken together, our quantitative analyses reveal a mode of ligand recognition by integrin α(X)β(2), which seem to differ in principles considerably from other OPN receptors.
Collapse
Affiliation(s)
- Eva Kläning
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark; Dept. of Biomedicine, Denmark
| | - Brian Christensen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Goran Bajic
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Søren V Hoffmann
- Institute for Storage Ring Facilities Aarhus (ISA), Dept. of Physics and Astronomy & Center for Storage Ring Facilities Aarhus, Denmark
| | - Nykola C Jones
- Institute for Storage Ring Facilities Aarhus (ISA), Dept. of Physics and Astronomy & Center for Storage Ring Facilities Aarhus, Denmark
| | - Morten M Callesen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Gregers R Andersen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark
| | - Esben S Sørensen
- Dept. of Molecular Biology and Genetics Aarhus University, Aarhus, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus Denmark
| | - Thomas Vorup-Jensen
- Dept. of Biomedicine, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus Denmark; MEMBRANES Research Center, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
32
|
Osteopontin-induced brown adipogenesis from white preadipocytes through a PI3K-AKT dependent signaling. Biochem Biophys Res Commun 2015; 459:553-9. [PMID: 25749339 DOI: 10.1016/j.bbrc.2015.02.153] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 02/25/2015] [Indexed: 11/20/2022]
Abstract
Recent studies have shown that OPN (osteopontin) plays critical roles in cell survival, differentiation, bio-mineralization, cancer and cardiovascular remodeling. However, its roles in the differentiation of brown adipocytes and the underlying mechanisms remain unclear. Therefore, the aim of this study was to investigate the roles of OPN in the brown adipogenesis and the underlying mechanisms. It was shown that the OPN successfully induced the differentiation of 3T3-L1 white preadipocytes into the PRDM16(+) (PRD1-BF1-RIZ1 homologous domain containing 16) and UCP-1(+) (uncoupling protein-1) brown adipocytes in a concentration and time-dependent manner. Also, activation of PI3K (phosphatidylinositol 3-kinase)-AKT pathway was required for the OPN-induced brown adipogenesis. The findings suggest OPN plays an important role in promoting the differentiation of the brown adipocytes and might provide a potential novel therapeutic approach for the treatment of obesity and related disorders.
Collapse
|
33
|
Osteopontin binding to the alpha 4 integrin requires highest affinity integrin conformation, but is independent of post-translational modifications of osteopontin. Matrix Biol 2014; 41:19-25. [PMID: 25446551 DOI: 10.1016/j.matbio.2014.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/20/2014] [Accepted: 11/22/2014] [Indexed: 12/17/2022]
Abstract
Osteopontin (OPN) is a ligand for the α4ß1 integrin, but the physiological importance of this binding is not well understood. Here, we have assessed the effect of post-translational modifications on OPN binding to the α4 integrin on cultured human leukocyte cell lines and compared OPN interaction with α4 integrin to that of VCAM and fibronectin. Jurkat cells, whose α4 integrins are inherently activated, adhered to different preparations of OPN in the presence of Mn(2+): the EC50 of adhesion was not affected by phosphorylation or glycosylation status. Thrombin cleavage of OPN at the C-terminus of the α4 integrin-binding site also did not affect binding affinity. THP-1 cells express a low-affinity conformation of the integrin and adhered to OPN only in the presence of Mn(2+) plus PMA or an activating antibody. This was in contrast to VCAM and fibronectin: THP-1 cells adhered to these ligands without integrin activation. Studies with ligand-induced binding site antibodies demonstrated that the SVVYGLR peptide of OPN bound to the α4 integrin with a similar affinity as the LDV peptide of fibronectin, suggesting that a high off-rate is responsible for the reduced binding of OPN to the low-affinity forms of this integrin. Together, the results suggest OPN has very low affinity for the α4 integrin on human leukocytes under physiological conditions.
Collapse
|
34
|
Identification of transglutaminase reactive residues in human osteopontin and their role in polymerization. PLoS One 2014; 9:e113650. [PMID: 25419572 PMCID: PMC4242673 DOI: 10.1371/journal.pone.0113650] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/26/2014] [Indexed: 01/21/2023] Open
Abstract
Osteopontin (OPN) is a highly posttranslationally modified protein present in several tissues where it is implicated in numerous physiological processes. OPN primarily exerts its functions through interaction with integrins via the Arg-Gly-Asp and Ser-Val-Val-Tyr-Gly-Leu-Arg sequences located in the N-terminal part of the protein. OPN can be polymerized by the cross-linking enzyme transglutaminase 2 (TG2), and polymerization has been shown to enhance the biological activity of OPN. However, little is known about the reactivity and location of the glutamine and lysine residues involved in the TG2-mediated modification of OPN. Here we show that TG2 catalyses the incorporation of 5-(Biotinamido)pentylamine at glutamines in both the N- and C-terminal parts of OPN, whereas TG2 primarily incorporated the glutamine-donor peptide biotinyl-TVQQEL-OH into the C-terminal part of OPN. By mass spectrometric analyses we identified Gln34, Gln42, Gln193 and Gln248 as the major TG2 reactive glutamines in OPN. The distribution of reactive Gln and Lys residues in OPN proved to be important, as the full-length protein but not the physiologically highly active integrin-binding N-terminal part of OPN were able to polymerize in a TG2-mediated reaction. Collectively, these data provide important new molecular knowledge about the mechanism of OPN polymerization.
Collapse
|
35
|
Osteopontin O-glycosylation contributes to its phosphorylation and cell-adhesion properties. Biochem J 2014; 463:93-102. [PMID: 25000122 DOI: 10.1042/bj20140060] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OPN (osteopontin) is a multiphosphorylated extracellular glycoprotein, which has important roles in bone remodelling, inflammation and cancer metastasis. OPN regulates cell spreading and adhesion primarily through its association with several integrins such as αvβ3, and its phosphorylation affects these processes. However, the mechanism by which OPN O-glycosylation affects these processes is not completely understood. In the present study, we demonstrated that OPN O-glycosylation self-regulates its biological activities and also affects its phosphorylation status. We prepared two recombinant OPNs, WT (wild-type)-OPN and mutant OPN (ΔO-OPN), which lacks five O-glycosylation sites at a threonine/proline-rich region. O-glycan defects in OPN increased its phosphorylation level, as observed by dephosphorylation assays. Moreover, compared with WT-OPN, ΔO-OPN exhibited enhanced cell spreading and adhesion activities and decreased associations with β1 integrins. This suggested that defects in O-glycans in OPN altered these activities, and that β1 integrins have a less important role in adhesion to ΔO-OPN. The cell-adhesion activity of dephosphorylated ΔO-OPN was higher than the cell-adhesion activities of ΔO-OPN and dephosphorylated WT-OPN. This suggested that some of the phosphorylation in ΔO-OPN caused by O-glycan defects and O-glycans of OPN suppressed the OPN cell-adhesion activity. Thus functional activities of OPN can be determined by the combined glycosylation and phosphorylation statuses and not by either status alone.
Collapse
|
36
|
Shao Z, Morser J, Leung LLK. Thrombin cleavage of osteopontin disrupts a pro-chemotactic sequence for dendritic cells, which is compensated by the release of its pro-chemotactic C-terminal fragment. J Biol Chem 2014; 289:27146-27158. [PMID: 25112870 DOI: 10.1074/jbc.m114.572172] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Thrombin cleavage alters the function of osteopontin (OPN) by exposing an integrin binding site and releasing a chemotactic C-terminal fragment. Here, we examined thrombin cleavage of OPN in the context of dendritic cell (DC) migration to define its functional domains. Full-length OPN (OPN-FL), thrombin-cleaved N-terminal fragment (OPN-R), thrombin- and carboxypeptidase B2-double-cleaved N-terminal fragment (OPN-L), and C-terminal fragment (OPN-CTF) did not have intrinsic chemotactic activity, but all potentiated CCL21-induced DC migration. OPN-FL possessed the highest potency, whereas OPNRAA-FL had substantially less activity, indicating the importance of RGD. We identified a conserved (168)RSKSKKFRR(176) sequence on OPN-FL that spans the thrombin cleavage site, and it demonstrated potent pro-chemotactic effects on CCL21-induced DC migration. OPN-FLR168A had reduced activity, and the double mutant OPNRAA-FLR168A had even lower activity, indicating that these functional domains accounted for most of the pro-chemotactic activity of OPN-FL. OPN-CTF also possessed substantial pro-chemotactic activity, which was fully expressed upon thrombin cleavage and its release from the intact protein, because OPN-CTF was substantially more active than OPNRAA-FLR168A containing the OPN-CTF sequence within the intact protein. OPN-R and OPN-L possessed similar potency, indicating that the newly exposed C-terminal SVVYGLR sequence in OPN-R was not involved in the pro-chemotactic effect. OPN-FL and OPN-CTF did not directly bind to the CD44 standard form or CD44v6. In conclusion, thrombin cleavage of OPN disrupts a pro-chemotactic sequence in intact OPN, and its loss of pro-chemotactic activity is compensated by the release of OPN-CTF, which assumes a new conformation and possesses substantial activity in enhancing chemokine-induced migration of DCs.
Collapse
Affiliation(s)
- Zhifei Shao
- Division of Hematology, Stanford University School of Medicine, Stanford, California 94305 and; Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - John Morser
- Division of Hematology, Stanford University School of Medicine, Stanford, California 94305 and; Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304
| | - Lawrence L K Leung
- Division of Hematology, Stanford University School of Medicine, Stanford, California 94305 and; Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304.
| |
Collapse
|
37
|
Wolak T. Osteopontin - a multi-modal marker and mediator in atherosclerotic vascular disease. Atherosclerosis 2014; 236:327-37. [PMID: 25128758 DOI: 10.1016/j.atherosclerosis.2014.07.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/26/2014] [Accepted: 07/07/2014] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory process of the vessel wall with systemic correlates. It is now well established that patients' outcome is tightly linked to atherosclerotic plaque stability, potentially more so than to the mere plaque size. Osteopontin (OPN) is an integrin-binding ligand, N-linked glycoprotein, which was recognized as a significant participant in the atherosclerotic inflammatory milieu. Evidence from several genetic mouse models suggests that OPN is an enhancer of atherosclerosis. This may be mediated by its capacity to enhance inflammation in the atherosclerotic plaque. Interestingly, OPN may also possess potentially protective vascular effects, such as attenuation of vascular calcification. In humans circulating levels of OPN were found to be independently associated with the severity of coronary atherosclerosis. Moreover, several studies report that high plasma OPN levels were associated with increased risk for major adverse cardiac events. This review aims to critically assess current understanding of the role of OPN in the atherosclerotic process, from animal models to clinical practice. Specific focus is given to evaluating whether OPN could serve as a marker for monitoring coronary atherosclerosis severity, and in parallel, assess the evidence for its role as a mediator in the pathogenic pathways leading to atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Talya Wolak
- Hypertension Unit Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
| |
Collapse
|
38
|
Osteopontin: A novel regulator at the cross roads of inflammation, obesity and diabetes. Mol Metab 2014; 3:384-93. [PMID: 24944898 PMCID: PMC4060362 DOI: 10.1016/j.molmet.2014.03.004] [Citation(s) in RCA: 294] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 02/06/2023] Open
Abstract
Since its first description more than 20 years ago osteopontin has emerged as an active player in many physiological and pathological processes, including biomineralization, tissue remodeling and inflammation. As an extracellular matrix protein and proinflammatory cytokine osteopontin is thought to facilitate the recruitment of monocytes/macrophages and to mediate cytokine secretion in leukocytes. Modulation of immune cell response by osteopontin has been associated with various inflammatory diseases and may play a pivotal role in the development of adipose tissue inflammation and insulin resistance. Here we summarize recent findings on the role of osteopontin in metabolic disorders, particularly focusing on diabetes and obesity.
Collapse
|
39
|
Zawadzka AM, Schilling B, Cusack MP, Sahu AK, Drake P, Fisher SJ, Benz CC, Gibson BW. Phosphoprotein secretome of tumor cells as a source of candidates for breast cancer biomarkers in plasma. Mol Cell Proteomics 2014; 13:1034-49. [PMID: 24505115 PMCID: PMC3977182 DOI: 10.1074/mcp.m113.035485] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is a heterogeneous disease whose molecular diversity is not well reflected in clinical and pathological markers used for prognosis and treatment selection. As tumor cells secrete proteins into the extracellular environment, some of these proteins reach circulation and could become suitable biomarkers for improving diagnosis or monitoring response to treatment. As many signaling pathways and interaction networks are altered in cancerous tissues by protein phosphorylation, changes in the secretory phosphoproteome of cancer tissues could reflect both disease progression and subtype. To test this hypothesis, we compared the phosphopeptide-enriched fractions obtained from proteins secreted into conditioned media (CM) derived from five luminal and five basal type breast cancer cell lines using label-free quantitative mass spectrometry. Altogether over 5000 phosphosites derived from 1756 phosphoproteins were identified, several of which have the potential to qualify as phosphopeptide plasma biomarker candidates for the more aggressive basal and also the luminal-type breast cancers. The analysis of phosphopeptides from breast cancer patient plasma and controls allowed us to construct a discovery list of phosphosites under rigorous collection conditions, and second to qualify discovery candidates generated from the CM studies. Indeed, a set of basal-specific phosphorylation CM site candidates derived from IBP3, CD44, OPN, FSTL3, LAMB1, and STC2, and luminal-specific candidates derived from CYTC and IBP5 were selected and, based on their presence in plasma, quantified across all cell line CM samples using Skyline MS1 intensity data. Together, this approach allowed us to assemble a set of novel cancer subtype specific phosphopeptide candidates for subsequent biomarker verification and clinical validation.
Collapse
Affiliation(s)
- Anna M Zawadzka
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, California 94945
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Christensen B, Sørensen E. Osteopontin is highly susceptible to cleavage in bovine milk and the proteolytic fragments bind the αVβ3-integrin receptor. J Dairy Sci 2014; 97:136-46. [DOI: 10.3168/jds.2013-7223] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/06/2013] [Indexed: 11/19/2022]
|
41
|
Nich C, Takakubo Y, Pajarinen J, Ainola M, Salem A, Sillat T, Rao AJ, Raska M, Tamaki Y, Takagi M, Konttinen YT, Goodman SB, Gallo J. Macrophages-Key cells in the response to wear debris from joint replacements. J Biomed Mater Res A 2013; 101:3033-45. [PMID: 23568608 PMCID: PMC3775910 DOI: 10.1002/jbm.a.34599] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/16/2012] [Accepted: 01/12/2013] [Indexed: 12/14/2022]
Abstract
The generation of wear debris is an inevitable result of normal usage of joint replacements. Wear debris particles stimulate local and systemic biological reactions resulting in chronic inflammation, periprosthetic bone destruction, and eventually, implant loosening, and revision surgery. The latter may be indicated in up to 15% patients in the decade following the arthroplasty using conventional polyethylene. Macrophages play multiple roles in both inflammation and in maintaining tissue homeostasis. As sentinels of the innate immune system, they are central to the initiation of this inflammatory cascade, characterized by the release of proinflammatory and pro-osteoclastic factors. Similar to the response to pathogens, wear particles elicit a macrophage response, based on the unique properties of the cells belonging to this lineage, including sensing, chemotaxis, phagocytosis, and adaptive stimulation. The biological processes involved are complex, redundant, both local and systemic, and highly adaptive. Cells of the monocyte/macrophage lineage are implicated in this phenomenon, ultimately resulting in differentiation and activation of bone resorbing osteoclasts. Simultaneously, other distinct macrophage populations inhibit inflammation and protect the bone-implant interface from osteolysis. Here, the current knowledge about the physiology of monocyte/macrophage lineage cells is reviewed. In addition, the pattern and consequences of their interaction with wear debris and the recent developments in this field are presented.
Collapse
Affiliation(s)
- Christophe Nich
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California; Laboratoire de Biomécanique et Biomatériaux Ostéo-Articulaires-UMR CNRS 7052, Faculté de Médecine-Université Paris 7, Paris, France; Department of Orthopaedic Surgery, European Teaching Hospital, Assistance Publique-Hôpitaux de Paris-Université Paris 5, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lu Y, Zhao X, Li K, Luo G, Nie Y, Shi Y, Zhou Y, Ren G, Feng B, Liu Z, Pan Y, Li T, Guo X, Wu K, Miranda-Vizuete A, Wang X, Fan D. Thioredoxin-like protein 2 is overexpressed in colon cancer and promotes cancer cell metastasis by interaction with ran. Antioxid Redox Signal 2013; 19:899-911. [PMID: 23311631 PMCID: PMC3763228 DOI: 10.1089/ars.2012.4736] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS Our previous work identified thioredoxin-like protein 2 (Txl-2) as the target of the monoclonal antibody MC3 associated with colon cancer, but its underlying mechanisms remain poorly understood. Txl-2, a novel thioredoxin (Trx) and nucleoside diphosphate kinase family member, is alternatively spliced and gives rise to three different Txl-2 isoforms. In this study, Txl-2 expression in colon cancer, differential functions for Txl-2 isoforms in cell invasion and metastasis, and the downstream signaling were investigated. RESULTS Txl-2 expression was elevated in colon cancer tissues compared to normal colonic tissues, with a high correlation between the histological grade and prognosis. Knockdown of Txl-2 expression significantly inhibited cancer cell motility, and the invasive and metastatic abilities of colon cancer cells. Interestingly, Txl-2 isoforms showed differential effects on cancer cell invasion and metastasis. Cell invasion and metastasis were significantly promoted by Txl-2b but inhibited by Txl-2c, while no obvious effect was observed for Txl-2a. Furthermore, a direct interaction was identified between Txl-2b and Ran, a Ras-related protein, by yeast two-hybrid assay and coimmunoprecipitation. PI3K pathway was found to be a major pathway mediating Txl-2b induced tumor invasion and metastasis. INNOVATION The current study provides a novel biomarker and target molecule for the diagnosis and treatment of colon cancer and provides a novel paradigm to understand how alternative splicing functions in human cancer. CONCLUSION Our findings demonstrate an elevated Txl-2 expression in colon cancer and that Txl-2b promotes cell invasion and metastasis through interaction with Ran and PI3K signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Lu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ge X, Lu Y, Leung TM, Sørensen ES, Nieto N. Milk osteopontin, a nutritional approach to prevent alcohol-induced liver injury. Am J Physiol Gastrointest Liver Physiol 2013; 304:G929-39. [PMID: 23518682 PMCID: PMC3652071 DOI: 10.1152/ajpgi.00014.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcohol consumption is a leading cause of liver disease worldwide; thus, there is an urgent need to develop novel therapeutic interventions. Key events for the onset and progression of alcoholic liver disease result in part from the gut-to-liver interaction. Osteopontin is a cytokine present at high concentration in human milk, umbilical cord, and infants' plasma with beneficial potential. We hypothesized that dietary administration of milk osteopontin could prevent alcohol-induced liver injury perhaps by maintaining gut integrity and averting hepatic inflammation and steatosis. Wild-type mice were fed either the control or the ethanol Lieber-DeCarli diets alone or in combination with milk osteopontin for 3 wk, and parameters of gut and liver damage were measured. Milk osteopontin protected the stomach and the gut by increasing gland height, crypt cell plus enterocyte proliferation, and mucin content in addition to lowering macrophages, plasmacytes, lymphocytes, and neutrophils in the mucosa and submucosa in alcohol-fed mice. Milk osteopontin targeted the gut-liver axis, preserving the expression of tight-junction proteins in alcohol-fed mice thus maintaining intestinal integrity and permeability. There was protection from liver injury since transaminases, the activity scores, triglyceride levels, neutrophil infiltration, 3-nitrotyrosine residues, lipid peroxidation end products, translocation of gram-negative bacteria, lipopolysaccharide levels, and tumor necrosis factor-α were lower in cotreated than in ethanol-fed mice. Furthermore, milk osteopontin diminished ethanol-mediated liver injury in OPN knockout mice. Milk osteopontin could be a simple effective nutritional therapeutic strategy to prevent alcohol hepatotoxicity due, among others, to gut protective, anti-inflammatory, and anti-steatotic actions.
Collapse
Affiliation(s)
- Xiaodong Ge
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| | - Yongke Lu
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| | - Tung-Ming Leung
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| | - Esben S. Sørensen
- 2Department of Molecular Biology and Genetics, Aarhus University, Aarhus Science Park, Denmark
| | - Natalia Nieto
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| |
Collapse
|
44
|
Phosphorylation of osteopontin in osteoarthritis degenerative cartilage and its effect on matrix metalloprotease 13. Rheumatol Int 2012; 33:1313-9. [PMID: 23132541 DOI: 10.1007/s00296-012-2548-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 10/21/2012] [Indexed: 12/25/2022]
Abstract
The purpose of this study is to observe the differences of osteopontin (OPN) phosphorylation in osteoarthritis (OA) cartilage and normal cartilage, and evaluate the possible correlations between the OPN phosphorylation and MMP-13 expression. Degenerative cartilage (n = 29) and normal cartilage (n = 10) were identified by hematoxylin-eosin, safranin-O staining and modified Mankin score. The phosphorylation level of OPN in OA cartilage and normal cartilage was detected by immunoprecipitation. Chondrocytes were treated with phospho-OPN, OPN or buffer. Quantitative reverse transcription polymerase chain reaction (qPCR) and ELISA were used to assess the expression of MMP-13 in different treatments. The OD values of phosphorylation of OPN in normal cartilage and OA cartilage were 137.89 ± 10.59 and 153.52 ± 8.80, respectively, (P = 0.000). Chondrocytes treated with OPN showed a higher MMP-13 expression at gene and protein level compared with control group. Chondrocytes treated with phospho-OPN showed the highest MMP-13 expression in gene and protein. In conclusion, our results revealed a higher phosphorylation level of OPN in OA cartilage than in normal cartilage. We found OPN leads to elevated expression of MMP-13 (both at gene level and protein level), and phospho-OPN had a more obvious upregulation effect on MMP-13 expression than nonphospho-OPN. Further studies are needed to reveal the mechanism of OPN phosphorylation on cartilage degeneration.
Collapse
|