1
|
Lu J, Xu X, Sun X, Du Y. Protein and peptide-based renal targeted drug delivery systems. J Control Release 2024; 366:65-84. [PMID: 38145662 DOI: 10.1016/j.jconrel.2023.12.036] [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: 07/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Renal diseases have become an increasingly concerned public health problem in the world. Kidney-targeted drug delivery has profound transformative potential on increasing renal efficacy and reducing extra-renal toxicity. Protein and peptide-based kidney targeted drug delivery systems have garnered more and more attention due to its controllable synthesis, high biocompatibility and low immunogenicity. At the same time, the targeting methods based on protein/peptide are also abundant, including passive renal targeting based on macromolecular protein and active targeting mediated by renal targeting peptide. Here, we review the application and the drug loading strategy of different proteins or peptides in targeted drug delivery, including the ferritin family, albumin, low molecular weight protein (LMWP), different peptide sequence and antibodies. In addition, we summarized the factors influencing passive and active targeting in drug delivery system, the main receptors related to active targeting in different kidney diseases, and a variety of nano forms of proteins based on the controllable synthesis of proteins.
Collapse
Affiliation(s)
- Jingyi Lu
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiaoling Xu
- College of Medical Sciences, Zhejiang Shuren University, 8 Shuren Street, Hangzhou, Zhejiang 310015, China.
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China.
| | - Yongzhong Du
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou, Zhejiang 310014, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321299, China.
| |
Collapse
|
2
|
Messex JK, Byrd CJ, Thomas MU, Liou GY. Macrophages Cytokine Spp1 Increases Growth of Prostate Intraepithelial Neoplasia to Promote Prostate Tumor Progression. Int J Mol Sci 2022; 23:4247. [PMID: 35457063 PMCID: PMC9027984 DOI: 10.3390/ijms23084247] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 11/27/2022] Open
Abstract
Prostate cancer development and progression are associated with increased infiltrating macrophages. Prostate cancer is derived from prostatic intraepithelial neoplasia (PIN) lesions. However, the effects macrophages have on PIN progression remain unclear. Here, we showed that the recruited macrophages adjacent to PIN expressed M2 macrophage markers. In addition, high levels of Spp1 transcripts, also known as osteopontin, were identified in these macrophages. Extraneously added Spp1 accelerated PIN cell proliferation through activation of Akt and JNK in a 3D culture setting. We also showed that PIN cells expressed CD44, integrin αv, integrin β1, and integrin β3, all of which have been previously reported as receptors for Spp1. Finally, blockade of Akt and JNK activation through their specific inhibitor completely abolished macrophage Spp1-induced cell proliferation of PIN. Hence, our data revealed Spp1 as another macrophage cytokine/growth factor and its mediated mechanism to upregulate PIN cell growth, thus promoting prostate cancer development.
Collapse
Affiliation(s)
- Justin K. Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA;
| | - Crystal J. Byrd
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA; (C.J.B.); (M.U.T.)
| | - Mikalah U. Thomas
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA; (C.J.B.); (M.U.T.)
| | - Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA;
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA; (C.J.B.); (M.U.T.)
| |
Collapse
|
3
|
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
|
4
|
Suri A, Singh N, Bansal SK. A Study on the Serum γ-Glutamyltranspeptidase and Plasma Osteopontin in Alcoholic Liver Disease. J Lab Physicians 2021; 14:101-108. [PMID: 36032990 PMCID: PMC9417738 DOI: 10.1055/s-0041-1729479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background
Alcoholic liver disease (ALD) is a major source of alcohol-related morbidity and mortality. Heavy drinkers and alcoholics may progress from fatty liver to alcoholic hepatitis to cirrhosis. The enzyme γ-glutamyltranspeptidase (GGT) is a membrane-bound glycoprotein which catalyzes the transfer of the γ-glutamyl group from γ-glutamyl peptides to other peptides, amino acids, and water. Serum GGT activity mainly attributed to hepatobiliary system and thus is an important marker of ALD. Hence the present study is conducted to estimate and correlate the levels of GGT and osteopontin (OPN) in ALD.
Aims and Objectives
The objective of this study is to estimate and correlate the levels of GGT and OPN in ALD.
Materials and Methods
Sixty clinically diagnosed cases of ALD and sixty age- and gender-matched healthy controls were recruited for the study. Blood samples were collected from them and serum aspartate aminotransferase, serum alanine transaminases (ALTs), serum ALP levels, and plasma OPN levels were measured. Estimation of serum aspartate transaminases (AST), ALTs, and alkaline phosphatase (ALP) was assayed by standard photometric methods in autoanalyzer ERBA-XL (EM-200) using commercially available kits. OPN was estimated by using commercial kit based on enzyme-linked immunosorbent assay.
Results
The parameters of the liver function tests such as AST, ALT, and ALP were significantly increased in patients with ALD (
p
< 0.001) when compared with the healthy control subjects. In the present study, significantly increased levels of γ-glutamyl transferases and OPN were found in patients with ALD (
p
< 0.001) when compared with the control subjects. OPN showed significant positive correlations with AST (
r
= 0.76,
p
< 0.001), ALT (
r
= 0.64,
p
< 0.001), ALP (
r
= 0.68,
p
< 0.001), and GGT (
r
= 0.61,
p
< 0.001).
Conclusion
The present study focuses on the role of GGT and OPN that are sensitive indicators of liver cell injury and are most helpful in recognizing hepatocellular diseases such as ALD, hepatitis, and liver cirrhosis. Hence, the pattern of the GGT and OPN levels elevation can be helpful diagnostically.
Collapse
Affiliation(s)
- Arpita Suri
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT University, Gurugram, Haryana, India
| | - Naveen Singh
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT University, Gurugram, Haryana, India
| | - Sanjiv Kumar Bansal
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT University, Gurugram, Haryana, India
| |
Collapse
|
5
|
Nephronectin as a Matrix Effector in Cancer. Cancers (Basel) 2021; 13:cancers13050959. [PMID: 33668838 PMCID: PMC7956348 DOI: 10.3390/cancers13050959] [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: 01/31/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The extracellular matrix provides an important scaffold for cells and tissues of multicellular organisms. The scaffold not only provides a secure anchorage point, but also functions as a reservoir for signalling molecules, sequestered and released when necessary. A dysregulated extracellular matrix may therefore modulate cellular behaviour, as seen during cancer progression. The extracellular matrix protein nephronectin was discovered two decades ago and found to regulate important embryonic developmental processes. Loss of either nephronectin or its receptor, integrin α8β1, leads to underdeveloped kidneys. Recent findings show that nephronectin is also dysregulated in breast cancer and plays a role in promoting metastasis. To enable therapeutic intervention, it is important to fully understand the role of nephronectin and its receptors in cancer progression. In this review, we summarise the literature on nephronectin, analyse the structure and domain-related functions of nephronectin and link these functions to potential roles in cancer progression. Abstract The extracellular matrix protein nephronectin plays an important regulatory role during embryonic development, controlling renal organogenesis through integrin α8β1 association. Nephronectin has three main domains: five N-terminal epidermal growth factor-like domains, a linker region harbouring two integrin-binding motifs (RGD and LFEIFEIER), and a C-terminal MAM domain. In this review, we look into the domain-related functions of nephronectin, and tissue distribution and expression. During the last two decades it has become evident that nephronectin also plays a role during cancer progression and in particular metastasis. Nephronectin is overexpressed in both human and mouse breast cancer compared to normal breast tissue where the protein is absent. Cancer cells expressing elevated levels of nephronectin acquire increased ability to colonise distant organs. In particular, the enhancer-motif (LFEIFEIER) which is specific to the integrin α8β1 association induces viability via p38 MAPK and plays a role in colonization. Integrins have long been desired as therapeutic targets, where low efficiency and receptor redundancy have been major issues. Based on the summarised publications, the enhancer-motif of nephronectin could present a novel therapeutic target.
Collapse
|
6
|
Ji Y, Garland MA, Sun B, Zhang S, Reynolds K, McMahon M, Rajakumar R, Islam MS, Liu Y, Chen Y, Zhou CJ. Cellular and developmental basis of orofacial clefts. Birth Defects Res 2020; 112:1558-1587. [PMID: 32725806 DOI: 10.1002/bdr2.1768] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/21/2020] [Accepted: 06/27/2020] [Indexed: 12/11/2022]
Abstract
During craniofacial development, defective growth and fusion of the upper lip and/or palate can cause orofacial clefts (OFCs), which are among the most common structural birth defects in humans. The developmental basis of OFCs includes morphogenesis of the upper lip, primary palate, secondary palate, and other orofacial structures, each consisting of diverse cell types originating from all three germ layers: the ectoderm, mesoderm, and endoderm. Cranial neural crest cells and orofacial epithelial cells are two major cell types that interact with various cell lineages and play key roles in orofacial development. The cellular basis of OFCs involves defective execution in any one or several of the following processes: neural crest induction, epithelial-mesenchymal transition, migration, proliferation, differentiation, apoptosis, primary cilia formation and its signaling transduction, epithelial seam formation and disappearance, periderm formation and peeling, convergence and extrusion of palatal epithelial seam cells, cell adhesion, cytoskeleton dynamics, and extracellular matrix function. The latest cellular and developmental findings may provide a basis for better understanding of the underlying genetic, epigenetic, environmental, and molecular mechanisms of OFCs.
Collapse
Affiliation(s)
- Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, California, USA
| | - Michael A Garland
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, California, USA
| | - Moira McMahon
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Ratheya Rajakumar
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Mohammad S Islam
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - Yue Liu
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Chengji J Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, California, USA.,Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, School of Medicine, University of California at Davis, Sacramento, California, USA.,Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, California, USA
| |
Collapse
|
7
|
Extracellular matrix, integrins, and focal adhesion signaling in polycystic kidney disease. Cell Signal 2020; 72:109646. [PMID: 32311505 DOI: 10.1016/j.cellsig.2020.109646] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022]
Abstract
In autosomal dominant polycystic kidney disease (ADPKD), the inexorable growth of numerous fluid-filled cysts leads to massively enlarged kidneys, renal interstitial damage, inflammation, and fibrosis, and progressive decline in kidney function. It has long been recognized that interstitial fibrosis is the most important manifestation associated with end-stage renal disease; however, the role of abnormal extracellular matrix (ECM) production on ADPKD pathogenesis is not fully understood. Early evidence showed that cysts in end-stage human ADPKD kidneys had thickened and extensively laminated cellular basement membranes, and abnormal regulation of gene expression of several basement membrane components, including collagens, laminins, and proteoglycans by cyst epithelial cells. These basement membrane changes were also observed in dilated tubules and small cysts of early ADPKD kidneys, indicating that ECM alterations were early features of cyst development. Renal cystic cells were also found to overexpress several integrins and their ligands, including ECM structural components and soluble matricellular proteins. ECM ligands binding to integrins stimulate focal adhesion formation and can promote cell attachment and migration. Abnormal expression of laminin-332 (laminin-5) and its receptor α6β4 stimulated cyst epithelial cell proliferation; and mice that lacked laminin α5, a component of laminin-511 normally expressed by renal tubules, had an overexpression of laminin-332 that was associated with renal cyst formation. Periostin, a matricellular protein that binds αVβ3- and αVβ5-integrins, was found to be highly overexpressed in the kidneys of ADPKD and autosomal recessive PKD patients, and several rodent models of PKD. αVβ3-integrin is also overexpressed by cystic epithelial cells, and the binding of periostin to αVβ3-integrin activates the integrin-linked kinase and downstream signal transduction pathways involved in tissue repair promoting cyst growth, ECM synthesis, and tissue fibrosis. This chapter reviews the roles of the ECM, integrins, and focal adhesion signaling in cyst growth and fibrosis in PKD.
Collapse
|
8
|
Kon S, Honda M, Ishikawa K, Maeda M, Segawa T. Antibodies against nephronectin ameliorate anti-type II collagen-induced arthritis in mice. FEBS Open Bio 2019; 10:107-117. [PMID: 31705832 PMCID: PMC6943231 DOI: 10.1002/2211-5463.12758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/25/2019] [Accepted: 11/07/2019] [Indexed: 12/23/2022] Open
Abstract
The extracellular matrix protein nephronectin (Npnt) is known to be critical for kidney development, but its function in inflammatory diseases is unknown. Here, we developed a new enzyme‐linked immunosorbent assay system to detect Npnt in various autoimmune diseases, which revealed that plasma Npnt levels are increased in various mouse autoimmune models. We also report that antibodies against the α8β1 integrin‐binding region of Npnt protect mice from anti‐type II collagen‐induced arthritis, suggesting that Npnt may be a potential therapeutic target molecule for the prevention of autoimmune arthritis.
Collapse
Affiliation(s)
- Shigeyuki Kon
- Department of Molecular Immunology, Faculty of Pharmaceutical Sciences, Fukuyama University, Japan
| | - Machiko Honda
- Department of Molecular Immunology, Faculty of Pharmaceutical Sciences, Fukuyama University, Japan
| | | | | | | |
Collapse
|
9
|
Chimento S, Billero V, Cavallin L, Romanelli M, Nadji M, Romanelli P. Evaluation of osteopontin expression in chronic wounds: a potential prognostic and therapeutic biomarker. J Wound Care 2019; 26:S4-S8. [PMID: 28880752 DOI: 10.12968/jowc.2017.26.sup9.s4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Osteopontin (OPN) is abundantly expressed during tissue repair, acting as a powerful chemokine that recruits inflammatory cells such as neutrophils, macrophages, and Langerhans cells. The role of OPN in chronic wounds has not been explored. In this study, we assess the expression levels of OPN in chronic wounds to assess its potential contribution to the exacerbated inflammation seen in chronic ulcers, which is thought to contribute to poor healing. METHODS This retrospective study included archived biopsies of chronic wounds from several aetiologies. Immunohistochemical staining and blind analysis of OPN expression were carried out. RESULTS We assessed biopsies from venous leg ulcers (n=5), diabetic foot ulcers (n=5), pyoderma gangrenosum (n=5), squamous cell carcinoma ulcers (n=4), and calciphylaxis ulcers (n=3). The data revealed that all these sets of chronic ulcers expressed high levels of OPN. CONCLUSION This study provides strong histopathologic evidence that OPN expression is significantly increased in chronic wounds, suggesting that its upregulation could contribute to the exacerbated inflammation. Furthermore, further characterisation of the role of OPN in wound healing could aid the development of specific and efficient anti-OPN therapies for the treatment of chronic wounds.
Collapse
Affiliation(s)
- S Chimento
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology, University of Pisa, Pisa, Italy.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US
| | - V Billero
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology, University of Pisa, Pisa, Italy.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US
| | - L Cavallin
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology, University of Pisa, Pisa, Italy.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US
| | - M Romanelli
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology, University of Pisa, Pisa, Italy.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US
| | - M Nadji
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology, University of Pisa, Pisa, Italy.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US
| | - P Romanelli
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology, University of Pisa, Pisa, Italy.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, US.,Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, US
| |
Collapse
|
10
|
Osteopontin mediates murine transfusion-related acute lung injury via stimulation of pulmonary neutrophil accumulation. Blood 2019; 134:74-84. [PMID: 31076444 DOI: 10.1182/blood.2019000972] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/02/2019] [Indexed: 01/18/2023] Open
Abstract
Transfusion-related acute lung injury (TRALI) is one of the leading causes of transfusion-related fatalities and is characterized by the onset of acute respiratory distress within 6 hours upon blood transfusion. Specific therapies are unavailable. Preexisting inflammation is a risk factor for TRALI and neutrophils (polymorphonuclear neutrophils [PMNs]) are considered to be the major pathogenic cells. Osteopontin (OPN) is a multifunctional protein expressed at sites of inflammation and, for example, is involved in pulmonary disorders, can regulate cellular migration, and can function as a PMN chemoattractant. We investigated whether OPN is involved in TRALI induction by promoting PMN recruitment to the lungs. Using a previously established murine TRALI model, we found that in contrast to wild-type (WT) mice, OPN knockout (KO) mice were resistant to antibody-mediated PMN-dependent TRALI induction. Administration of purified OPN to the OPN KO mice, however, restored the TRALI response and pulmonary PMN accumulation. Alternatively, blockade of OPN in WT mice using an anti-OPN antibody prevented the onset of TRALI induction. Using pulmonary immunohistochemistry, OPN could be specifically detected in the lungs of mice that suffered from TRALI. The OPN-mediated TRALI response seemed dependent on macrophages, likely the cellular source of OPN and OPN polymerization, and independent from the OPN receptor CD44, interleukin 6 (IL-6), and other PMN chemoattractants including macrophage inflammatory protein-2 (MIP-2). These data indicate that OPN is critically required for induction of antibody-mediated murine TRALI through localization to the lungs and stimulation of pulmonary PMN recruitment. This suggests that anti-OPN antibody therapy may be a potential therapeutic strategy to explore in TRALI patients.
Collapse
|
11
|
Theisen DJ, Ferris ST, Briseño CG, Kretzer N, Iwata A, Murphy KM, Murphy TL. Batf3-Dependent Genes Control Tumor Rejection Induced by Dendritic Cells Independently of Cross-Presentation. Cancer Immunol Res 2019; 7:29-39. [PMID: 30482745 DOI: 10.1158/2326-6066.cir-18-0138] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 09/12/2018] [Accepted: 11/21/2018] [Indexed: 11/16/2022]
Abstract
The BATF3-dependent cDC1 lineage of conventional dendritic cells (cDC) is required for rejection of immunogenic sarcomas and for rejection of progressive sarcomas during checkpoint blockade therapy. One unique function of the cDC1 lineage is the efficient cross-presentation of tumor-derived neoantigens to CD8+ T cells, but it is not clear that this is the only unique function of cDC1 required for tumor rejection. We previously showed that BATF3 functions during cDC1 lineage commitment to maintain IRF8 expression in the specified cDC1 progenitor. However, since cDC1 progenitors do not develop into mature cDC1s in Batf3 -/- mice, it is still unclear whether BATF3 has additional functions in mature cDC1 cells. A transgenic Irf8-Venus reporter allele increases IRF8 protein concentration sufficiently to allow autonomous cDC1 development in spleens of Batf3 -/- mice. These restored Batf3 -/- cDC1s are transcriptionally similar to control wild-type cDC1s but have reduced expression of a restricted set of cDC1-specific genes. Restored Batf3 -/- cDC1s are able to cross-present cell-associated antigens both in vitro and in vivo However, Batf3 -/- cDC1 exhibit altered characteristics in vivo and are unable to mediate tumor rejection. These results show that BATF3, in addition to regulating Irf8 expression to stabilize cDC1 lineage commitment, also controls expression of a small set of genes required for cDC1-mediated tumor rejection. These BATF3-regulated genes may be useful targets in immunotherapies aimed at promoting tumor rejection.
Collapse
Affiliation(s)
- Derek J Theisen
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Stephen T Ferris
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Nicole Kretzer
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
- Howard Hughes Medical Institute, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri.
| |
Collapse
|
12
|
Tension enhances cell proliferation and collagen synthesis by upregulating expressions of integrin αvβ3 in human keloid-derived mesenchymal stem cells. Life Sci 2018; 219:272-282. [PMID: 30597173 DOI: 10.1016/j.lfs.2018.12.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/19/2018] [Accepted: 12/25/2018] [Indexed: 12/21/2022]
Abstract
AIMS Keloids are a dermal fibrotic disease whose etiology remains totally unknown and for which there is no successful treatment. Mechanical tension, in addition, is closely associated with the germination and development of keloids. In this study, we investigated the influence of human keloid-derived mesenchymal stem cells (KD-MSCs) on cell proliferation, collagen synthesis, and expressions of integrin αvβ3 under tension. MAIN METHODS KD-MSCs and human normal skin-derived mesenchymal stem cells (NS-MSCs) were isolated and cultured in stem cell medium with a gradual increase in the serum concentration. Cell proliferation and collagen synthesis were detected by Cell Counting Kit-8 (CCK-8) assay and hydroxyproline content analysis under tension respectively. We investigated the messenger RNA expressions of nine integrin subunits, including integrin units α2, α3, α5, αv, α8, α10, α11, β1, and β3, in KD-MSCs stimulated with tension. Identification of differentially expressed genes was performed by Western blot analysis and immunocytochemistry staining. KEY FINDINGS We obtained high-purity KD-MSCs and NS-MSCs using the culture method of decreasing serum concentration gradient gradually. Furthermore, we found that tension enhances cell proliferation and collagen synthesis and promotes expressions of integrin αvβ3 in KD-MSCs. In addition, blocking experiments showed that increased integrin αvβ3 expression affects cell proliferation and collagen synthesis of KD-MSCs under tension. SIGNIFICANCE Our results suggest that integrin αvβ3 receptor may be sensitive molecules of mechanical tension and could contribute to the occurrence and development of keloids. It could lead to novel targets for therapeutic intervention, treatment, and prevention of recurrence for keloid disorders.
Collapse
|
13
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
14
|
Cavelier S, Dastjerdi AK, McKee MD, Barthelat F. Bone toughness at the molecular scale: A model for fracture toughness using crosslinked osteopontin on synthetic and biogenic mineral substrates. Bone 2018; 110:304-311. [PMID: 29486368 DOI: 10.1016/j.bone.2018.02.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 12/20/2022]
Abstract
The most prominent structural components in bone are collagen and mineral. However, bone additionally contains a substantial amount of noncollagenous proteins (most notably of the SIBLING protein family), some of which may act as cohesive/adhesive "binders" for the composite hybrid collagen/mineral scaffolding, whether in the bulk phase of bone, or at its interfaces. One such noncollagenous protein - osteopontin (OPN) - appears to be critical to the deformability and fracture toughness of bone. In the present study, we used a reconstructed synthetic mineral-OPN-mineral interface, and a biogenic (natural tooth dentin) mineral/collagen-OPN-mineral/collagen interface, to measure the fracture toughness of OPN on mineralized substrates. We used this system to test the hypothesis that OPN crosslinking by the enzyme tissue transglutaminase 2 (TG2) that is found in bone enhances interfacial adhesion to increase the fracture toughness of bone. For this, we prepared double-cantilever beam substrates of synthetic pure hydroxyapatite mineral, and of narwhal dentin, and directly apposed them to one another under different intervening OPN/crosslinking conditions, and fracture toughness was tested using a miniaturized loading stage. The work-of-fracture of the OPN interface was measured for different OPN formulations (monomer vs. polymer), crosslinking states, and substrate composition. Noncrosslinked OPN provided negligible adhesion on pure hydroxyapatite, whereas OPN crosslinking (by the chemical crosslinker glutaraldehyde, and TG2 enzyme) provided strong interfacial adhesion for both hydroxyapatite and dentin using monomeric and polymeric OPN. Pre-coating of the substrate beams with monomeric OPN further improved the adhesive performance of the samples, likely by allowing effective binding of this nascent OPN form to mineral/matrix components, with this pre-attachment providing a protein layer for additional crosslinking between the substrates.
Collapse
Affiliation(s)
- S Cavelier
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada
| | - A K Dastjerdi
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada
| | - M D McKee
- Faculty of Dentistry, Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.
| | - F Barthelat
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
15
|
Ogawa T, Li Y, Lua I, Hartner A, Asahina K. Isolation of a unique hepatic stellate cell population expressing integrin α8 from embryonic mouse livers. Dev Dyn 2018; 247:867-881. [PMID: 29665133 DOI: 10.1002/dvdy.24634] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/16/2018] [Accepted: 04/10/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hepatic stellate cells (HSCs) play an important role in liver fibrogenesis. However, little is known about their phenotype and role in liver development. The aim of this study is to identify specific markers for embryonic HSCs. RESULTS Using antibodies against ALCAM and PDPN, we separated mesothelial cells (MCs) and HSCs from developing livers and identified integrin α8 (ITGA8) as a marker for embryonic desmin+ HSCs that are preferentially localized near the developing liver surface and α-smooth muscle actin+ perivascular mesenchymal cells around the vein. A cell lineage-tracing study revealed that upon differentiation, MC-derived HSCs or perivascular mesenchymal cells express ITGA8 during liver development. Using anti-ITGA8 antibodies, we succeeded in isolating MC-derived HSCs and perivascular mesenchymal cells from embryonic livers. In direct co-culture, ITGA8+ mesenchymal cells promoted the expression of hepatocyte and cholangiocyte markers in hepatoblasts. In the normal adult liver, expression of ITGA8 was restricted to portal fibroblasts in the portal triad. Upon liver injury, myofibroblasts increased the expression of ITGA8. CONCLUSIONS ITGA8 is a specific cell surface marker of MC-derived HSCs and perivascular mesenchymal cells in the developing liver. Our data suggest that ITGA8+ mesenchymal cells maintain the phenotype of hepatoblast in liver development. Developmental Dynamics 247:867-881, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tomohiro Ogawa
- Southern California Research Center for ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California.,Center for the Advancement of Higher Education, Faculty of Engineering, Kindai University, Hiroshima, Japan
| | - Yuchang Li
- Southern California Research Center for ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Ingrid Lua
- Southern California Research Center for ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Kinji Asahina
- Southern California Research Center for ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
16
|
Zimmerman SE, Hiremath C, Tsunezumi J, Yang Z, Finney B, Marciano DK. Nephronectin Regulates Mesangial Cell Adhesion and Behavior in Glomeruli. J Am Soc Nephrol 2018; 29:1128-1140. [PMID: 29335243 DOI: 10.1681/asn.2017070752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/13/2017] [Indexed: 01/03/2023] Open
Abstract
A critical aspect of kidney function occurs at the glomerulus, the capillary network that filters the blood. The glomerular basement membrane (GBM) is a key component of filtration, yet our understanding of GBM interactions with mesangial cells, specialized pericytes that provide structural stability to glomeruli, is limited. We investigated the role of nephronectin (Npnt), a GBM component and known ligand of α8β1 integrin. Immunolocalization and in situ hybridization studies in kidneys of adult mice revealed that nephronectin is produced by podocytes and deposited into the GBM. Conditional deletion of Npnt from nephron progenitors caused a pronounced increase in mesangial cell number and mesangial sclerosis. Nephronectin colocalized with α8β1 integrin to novel, specialized adhesion structures that occurred at sites of mesangial cell protrusion at the base of the capillary loops. Absence of nephronectin disrupted these adhesion structures, leading to mislocalization of α8β1. Podocyte-specific deletion of Npnt also led to mesangial sclerosis in mice. These results demonstrate a novel role for nephronectin and α8β1 integrin in a newly described adhesion complex and begin to uncover the molecular interactions between the GBM and mesangial cells, which govern mesangial cell behavior and may have a role in pathologic states.
Collapse
Affiliation(s)
- Susan E Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chitkale Hiremath
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jun Tsunezumi
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhufeng Yang
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bronwyn Finney
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Denise K Marciano
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
17
|
Mehta BB, Sharma S, Vasishta RK, Sen RK, Sharma A, Luthra-Guptasarma M. Blocking osteopontin-fibronectin interactions reduce extracellular fibronectin deployment and arthritic immunopathology. Int Immunopharmacol 2018; 55:297-305. [PMID: 29306173 DOI: 10.1016/j.intimp.2017.12.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 02/07/2023]
Abstract
Elevated levels of a thrombin-cleaved fragment of osteopontin (OPNT) are seen in synovial fluid (SF) and tissues of rheumatoid arthritis (RA) patients. OPNT binds to integrins on cell surfaces, inducing adhesion, migration and survival of inflammatory cells in the synovial joints, where OPNT binds to fibronectin to link fibroblast-like synoviocytes (FLS) with B cells, stimulating the latter to produce inflammatory cytokines. Our aim was to block OPNT-fibronectin interactions and examine whether this reduces inflammation. A human antibody (phage displayed) library was used to select scFv antibodies cognate to OPNT, and a particular scFv antibody (scFv 31) was evaluated. Adhesion, migration and fibronectin polymerization of FLS cells derived from RA patients were monitored, in cultures incorporating scFv 31. Also, scFv 31 was used in mice with CAIA (collagen antibody-induced arthritis), subjected to clinical and histological assessment, analysis of fibronectin and cartilage damage and induction of pro-inflammatory cytokines. The scFv antibody, scFv 31, appeared to cause significantly reduced migration of synovial fibroblasts, altered cell morphology, changes in actin stress fiber arrangement, and marked reduction in fibronectin. In CAIA mice, scFv 31 appeared to prevent arthritic changes through inhibition of synovial hypertrophy and loss of articular cartilage, decrease in fibronectin polymerization and expression of pro-inflammatory cytokines implicated in arthritis. Osteopontin-fibronectin interaction(s) appear to play a role in the expression of key inflammatory molecules by B cells infiltrating the synovial joint. The scFv antibody, scFv 31, provides a potential therapeutic lead for inhibition of some processes implicated in rheumatoid arthritis.
Collapse
Affiliation(s)
- Brij Bhushan Mehta
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Saniya Sharma
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Rakesh K Vasishta
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ramesh K Sen
- Department of Orthopaedics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Aman Sharma
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Manni Luthra-Guptasarma
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| |
Collapse
|
18
|
Early and late gene expression profiles of the ovine mucosa in response to Haemonchus contortus infection employing Illumina RNA-seq technology. Parasitol Int 2017; 66:681-692. [PMID: 28552633 DOI: 10.1016/j.parint.2017.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/03/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023]
Abstract
We conducted herein transcriptome sequencing of the ovine abomasal tissues using the Illumina HiSeq 4000 platform to segregate early and late H. contortus-infected sheep (7 and 50days post-infected groups, respectively) from the control naive ones. A total of 548, 357 and 7 were substantially induced genes in 7days post-infection versus uninfected-control group, 50days post-infection versus 7days post-infection (7dpi), and 50days post-infection (50dpi) versus uninfected-control group, respectively. However, a total of 301, 355 and 11 were significantly repressed genes between 7dpi versus uninfected-control group, 50dpi versus 7dpi, and 50dpi versus uninfected-control group, correspondingly. This indicates that H. contortus infection induced a more potent activation of abomasal gene expression in the early stage of infection as compared to the late stage. Seven pathways were annotated by Kyoto Encyclopedia of Genes, and Genomes pathway analysis accounted for the significant percentage in early H. contortus infection. This study shows for the first time that both galectin-11 and matricellular protein osteopontin are up-regulated in abomasal tissue after chronic H. contortus infection, while galectin-4 is specifically down-regulated in the early infection. Additionally, our results showed that the induction or repression of these molecules is likely to determine the infection progression.
Collapse
|
19
|
Das V, Kalyan G, Hazra S, Pal M. Understanding the role of structural integrity and differential expression of integrin profiling to identify potential therapeutic targets in breast cancer. J Cell Physiol 2017; 233:168-185. [DOI: 10.1002/jcp.25821] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/23/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Vishal Das
- Biological Sciences and Technology DivisionCSIR‐North East Institute of Science and TechnologyJorhat, AssamIndia
| | - Gazal Kalyan
- Department of BiotechnologyIndian Institute of Technology Roorkee (IITR)RoorkeeUttarakhandIndia
| | - Saugata Hazra
- Department of BiotechnologyIndian Institute of Technology Roorkee (IITR)RoorkeeUttarakhandIndia
- Centre for NanotechnologyIndian Institute of Technology RoorkeeRoorkeeUttarakhandIndia
| | - Mintu Pal
- Biological Sciences and Technology DivisionCSIR‐North East Institute of Science and TechnologyJorhat, AssamIndia
| |
Collapse
|
20
|
Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Mol Cancer 2017; 16:31. [PMID: 28148268 PMCID: PMC5286812 DOI: 10.1186/s12943-017-0597-8] [Citation(s) in RCA: 405] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/19/2017] [Indexed: 02/08/2023] Open
Abstract
Tumour progression is dependent on the interaction between tumour cells and cells of the surrounding microenvironment. The tumour is a dynamic milieu consisting of various cell types such as endothelial cells, fibroblasts, cells of the immune system and mesenchymal stem cells (MSCs). MSCs are multipotent stromal cells that are known to reside in various areas such as the bone marrow, fat and dental pulp. MSCs have been found to migrate towards inflammatory sites and studies have shown that they also migrate towards and incorporate into the tumour. The key question is how they interact there. MSCs may interact with tumour cells through paracrine signalling. On the other hand, MSCs have the capacity to differentiate to various cell types such as osteocytes, chondrocytes and adipocytes and it is possible that MSCs differentiate at the site of the tumour. More recently it has been shown that cross-talk between tumour cells and MSCs has been shown to increase metastatic potential and promote epithelial-to-mesenchymal transition. This review will focus on the role of MSCs in tumour development at various stages of progression from growth of the primary tumour to the establishment of distant metastasis.
Collapse
Affiliation(s)
- Sarah M Ridge
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, Costello Road, Galway, Ireland.,Prostate Cancer Institute, School of Medicine, Costello Road, Galway, Ireland
| | - Francis J Sullivan
- Prostate Cancer Institute, School of Medicine, Costello Road, Galway, Ireland
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, Costello Road, Galway, Ireland. .,Prostate Cancer Institute, School of Medicine, Costello Road, Galway, Ireland.
| |
Collapse
|
21
|
Herdl S, Huebner H, Volkert G, Marek I, Menendez-Castro C, Noegel SC, Ruebner M, Rascher W, Hartner A, Fahlbusch FB. Integrin α8 Is Abundant in Human, Rat, and Mouse Trophoblasts. Reprod Sci 2017; 24:1426-1437. [PMID: 28136130 DOI: 10.1177/1933719116689597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Integrins exert regulatory functions in placentogenesis. Null mutation of certain integrin α subunits leads to placental defects with subsequent fetal growth restriction or embryonic lethality in mice. So far, the placental role of α8 integrin remains to be determined. METHODS Localization of α8 integrin and its ligands, fibronectin (FN) and osteopontin (OPN), was studied by immunohistochemistry in human, rat, and mouse placenta. The vascularization of the placental labyrinth layer of α8 integrin-deficient mice was determined by CD31 staining. In humans, α8 integrin expression was assessed via real-time polymerase chain reaction in healthy placentas, in the placental pathologies such as intrauterine growth restriction (IUGR), preeclampsia, and HELLP-syndrome (hemolysis, elevated liver enzymes, low platelet count), as well as in primary extravillous trophoblasts (EVT) and villous trophoblasts. RESULTS In humans, α8 integrin was detected in first and third trimester syncytiotrophoblast and EVT. Although OPN showed the same localization, FN was observed in EVT only. No expressional changes in α8 integrin were detected in the placental pathologies studied. Rodent placenta showed α8 integrin expression in giant cells and in the labyrinth layer. The localization of OPN and FN, however, showed species-specific differences. Knockout of α8 integrin in mice did not cause IUGR, despite some reduction in labyrinth layer vascularization. CONCLUSION α8 Integrin is expressed in functional placental compartments among its ligands, OPN and/or FN, across species. Although this may point to a regulatory role in trophoblast function, our data from α8 integrin-deficient mice indicated only mild placental pathology. Thus, the lack of placental α8 integrin seems to be largely compensated for.
Collapse
Affiliation(s)
- Sebastian Herdl
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hanna Huebner
- 2 Department of Gynaecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gudrun Volkert
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ines Marek
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Carlos Menendez-Castro
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie C Noegel
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Ruebner
- 2 Department of Gynaecology and Obstetrics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Rascher
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Hartner
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian B Fahlbusch
- 1 Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
22
|
Wen Y, Jeong S, Xia Q, Kong X. Role of Osteopontin in Liver Diseases. Int J Biol Sci 2016; 12:1121-8. [PMID: 27570486 PMCID: PMC4997056 DOI: 10.7150/ijbs.16445] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/08/2016] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN), a multifunctional protein, is involved in numerous pathological conditions including inflammation, immunity, angiogenesis, fibrogenesis and carcinogenesis in various tissues. Extensive studies have elucidated the critical role of OPN in cell signaling such as regulation of cell proliferation, migration, inflammation, fibrosis and tumor progression. In the liver, OPN interacts with integrins, CD44, vimentin and MyD88 signaling, thereby induces infiltration, migration, invasion and metastasis of cells. OPN is highlighted as a chemoattractant for macrophages and neutrophils during injury in inflammatory liver diseases. OPN activates hepatic stellate cells (HSCs) to exert an enhancer in fibrogenesis. The role of OPN in hepatocellular carcinoma (HCC) has also generated significant interests, especially with regards to its role as a diagnostic and prognostic factor. Interestingly, OPN acts an opposing role in liver repair under different pathological conditions. This review summarizes the current understanding of OPN in liver diseases. Further understanding of the pathophysiological role of OPN in cellular interactions and molecular mechanisms associated with hepatic inflammation, fibrosis and cancer may contribute to the development of novel strategies for clinical diagnosis, monitoring and therapy of liver diseases.
Collapse
Affiliation(s)
- Yankai Wen
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China;; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Seogsong Jeong
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoni Kong
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Thrombin Cleavage of Osteopontin Modulates Its Activities in Human Cells In Vitro and Mouse Experimental Autoimmune Encephalomyelitis In Vivo. J Immunol Res 2016; 2016:9345495. [PMID: 27478856 PMCID: PMC4961817 DOI: 10.1155/2016/9345495] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 12/14/2022] Open
Abstract
Osteopontin is a proinflammatory cytokine and plays a pathogenetic role in multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), by recruiting autoreactive T cells into the central nervous system. Osteopontin functions are modulated by thrombin cleavage generating N- and C-terminal fragment, whose individual roles are only partly known. Published data are difficult to compare since they have been obtained with heterogeneous approaches. Interestingly, thrombin cleavage of osteopontin unmasks a cryptic domain of interaction with α4β1 integrin that is the main adhesion molecule involved in lymphocyte transmigration to the brain and is the target for natalizumab, the most potent drug preventing relapses. We produced recombinant osteopontin and its N- and C-terminal fragments in an eukaryotic system in order to allow their posttranslational modifications. We investigated, in vitro, their effect on human cells and in vivo in EAE. We found that the osteopontin cleavage plays a key role in the function of this cytokine and that the two fragments exert distinct effects both in vitro and in vivo. These findings suggest that drugs targeting each fragment may be used to fine-tune the pathological effects of osteopontin in several diseases.
Collapse
|
24
|
Khalifeh-Soltani A, Ha A, Podolsky MJ, McCarthy DA, McKleroy W, Azary S, Sakuma S, Tharp KM, Wu N, Yokosaki Y, Hart D, Stahl A, Atabai K. α8β1 integrin regulates nutrient absorption through an Mfge8-PTEN dependent mechanism. eLife 2016; 5. [PMID: 27092791 PMCID: PMC4868538 DOI: 10.7554/elife.13063] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/18/2016] [Indexed: 12/25/2022] Open
Abstract
Coordinated gastrointestinal smooth muscle contraction is critical for proper nutrient absorption and is altered in a number of medical disorders. In this work, we demonstrate a critical role for the RGD-binding integrin α8β1 in promoting nutrient absorption through regulation of gastrointestinal motility. Smooth muscle-specific deletion and antibody blockade of α8 in mice result in enhanced gastric antral smooth muscle contraction, more rapid gastric emptying, and more rapid transit of food through the small intestine leading to malabsorption of dietary fats and carbohydrates as well as protection from weight gain in a diet-induced model of obesity. Mechanistically, ligation of α8β1 by the milk protein Mfge8 reduces antral smooth muscle contractile force by preventing RhoA activation through a PTEN-dependent mechanism. Collectively, our results identify a role for α8β1 in regulating gastrointestinal motility and identify α8 as a potential target for disorders characterized by hypo- or hyper-motility. DOI:http://dx.doi.org/10.7554/eLife.13063.001 Animals absorb nutrients from the food they eat in a complicated process that involves multiple steps. In the mouth, teeth break down the food into smaller chunks. Then the food passes through the stomach and small intestine, where enzymes break it down into individual molecules that are small enough to be absorbed by cells that line the small intestine. These cells then package the molecules and release them into the bloodstream so that they can be distributed to the rest of the body. Muscles in the wall of the small intestine control how quickly food travels through this part of the gut. If food moves too quickly, the cells that line the intestine have less time to absorb the food molecules and may fail to absorb enough nutrients. If the food moves too slowly, an individual may experience nausea or vomiting, or the contents of their stomach may spill into their lungs. In 2014, researchers reported that a protein in breast milk called Mfge8 helps to boost the number of fat molecules absorbed from food. Now, Khalifeh-Soltani et al. – including some of the same researchers involved in the earlier work – show that Mfge8 also slows the rate at which food travels through the small intestine in mice. Mfge8 binds to another protein called integrin α8β1 to control how often the intestine muscles contract. Genetically engineered mice that lacked integrin α8β1 developed diarrhea and food passed through their intestines more quickly than in normal mice. Furthermore, these mice did not gain as much weight as normal mice when they were fed a high-fat diet. Khalifeh-Soltani et al.’s findings show that Mfge8 has a dual role in controlling the absorption of food molecules in the small intestine. The next challenge is to find out whether drugs that alter the activity of integrin α8β1 could be used to help treat patients with diseases in which food moves too quickly, or too slowly, through the gut. DOI:http://dx.doi.org/10.7554/eLife.13063.002
Collapse
Affiliation(s)
- Amin Khalifeh-Soltani
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Arnold Ha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Donald A McCarthy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - William McKleroy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Saeedeh Azary
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Stephen Sakuma
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Kevin M Tharp
- Metabolic Biology, University of California, Berkeley, Berkeley, United States.,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, United States
| | - Nanyan Wu
- Lung Biology Center, University of California, San Francisco, San Francisco, United States
| | - Yasuyuki Yokosaki
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Health Administration Center, Hiroshima University, Hiroshima, Japan
| | - Daniel Hart
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Andreas Stahl
- Metabolic Biology, University of California, Berkeley, Berkeley, United States.,Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, United States
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States.,Department of Medicine, University of California, San Francisco, San Francisco, United States.,Lung Biology Center, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
25
|
Jürets A, Le Bras M, Staffler G, Stein G, Leitner L, Neuhofer A, Tardelli M, Turkof E, Zeyda M, Stulnig TM. Inhibition of Cellular Adhesion by Immunological Targeting of Osteopontin Neoepitopes Generated through Matrix Metalloproteinase and Thrombin Cleavage. PLoS One 2016; 11:e0148333. [PMID: 26840958 PMCID: PMC4740464 DOI: 10.1371/journal.pone.0148333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/15/2016] [Indexed: 01/10/2023] Open
Abstract
Osteopontin (OPN), a secreted protein involved in inflammatory processes and cancer, induces cell adhesion, migration, and activation of inflammatory pathways in various cell types. Cells bind OPN via integrins at a canonical RGD region in the full length form as well as to a contiguous cryptic site that some have shown is unmasked upon thrombin or matrix metalloproteinase cleavage. Thus, the adhesive capacity of osteopontin is enhanced by proteolytic cleavage that may occur in inflammatory conditions such as obesity, atherosclerosis, rheumatoid arthritis, tumor growth and metastasis. Our aim was to inhibit cellular adhesion to recombinant truncated proteins that correspond to the N-terminal cleavage products of thrombin- or matrix metalloproteinase-cleaved OPN in vitro. We specifically targeted the cryptic integrin binding site with monoclonal antibodies and antisera induced by peptide immunization of mice. HEK 293 cells adhered markedly stronger to truncated OPN proteins than to full length OPN. Without affecting cell binding to the full length form, the raised monoclonal antibodies specifically impeded cellular adhesion to the OPN fragments. Moreover, we show that the peptides used for immunization were able to induce antisera, which impeded adhesion either to all OPN forms, including the full-length form, or selectively to the corresponding truncated recombinant proteins. In conclusion, we developed immunological tools to selectively target functional properties of protease-cleaved OPN forms, which could find applications in treatment and prevention of various inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Alexander Jürets
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | | | - Gesine Stein
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lukas Leitner
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Angelika Neuhofer
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matteo Tardelli
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Edvin Turkof
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Maximilian Zeyda
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas M. Stulnig
- Christian Doppler Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
26
|
Jacobo-Estrada T, Cardenas-Gonzalez M, Santoyo-Sánchez M, Parada-Cruz B, Uria-Galicia E, Arreola-Mendoza L, Barbier O. Evaluation of kidney injury biomarkers in rat amniotic fluid after gestational exposure to cadmium. J Appl Toxicol 2016; 36:1183-93. [PMID: 26815315 DOI: 10.1002/jat.3286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/24/2022]
Abstract
Cadmium is a well-characterized nephrotoxic agent that is also capable of accumulating and diffusing across the placenta; however, only a few studies have addressed its effects over fetal kidneys and none of them has used a panel of sensitive and specific biomarkers for the detection of kidney injury. The goal of this study was to determine cadmium renal effects in rat fetuses by the quantification of early kidney injury biomarkers. Pregnant Wistar rats were exposed by inhalation to an isotonic saline solution or to CdCl2 solution (DDel =1.48 mg Cd kg(-1) day(-1) ) during gestational days (GD) 8-20. On GD 21, dams were euthanized and samples obtained. Kidney injury biomarkers were quantified in amniotic fluid samples and fetal kidneys were microscopically evaluated to search for histological alterations. Our results showed that cadmium exposure significantly raised albumin, osteopontin, vascular endothelial growth factor and tissue inhibitor of metalloproteinases-1 levels in amniotic fluid, whereas it decreased creatinine. Clusterin, calbindin and IFN-inducible protein 10 did not show any change. Accordingly, histological findings showed tubular damage and precipitations in the renal pelvis. In conclusion, gestational exposure to cadmium induces structural alterations in fetal renal tissue that can be detected by some kidney injury biomarkers in amniotic fluid samples. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tania Jacobo-Estrada
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Mariana Cardenas-Gonzalez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Mitzi Santoyo-Sánchez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Benjamín Parada-Cruz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| | - Esther Uria-Galicia
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomas, CP 11340, México, D.F., México
| | - Laura Arreola-Mendoza
- Departamento de Biociencias e Ingeniería, Centro Interdisciplinario de Investigaciones y Estudios sobre Medio Ambiente y Desarrollo, Instituto Politécnico Nacional, 30 de Junio de 1520 s/n, Col. Barrio la Laguna Ticomán, CP 07340, México, D.F., México
| | - Olivier Barbier
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, México, D.F., México
| |
Collapse
|
27
|
Roy D, Das K, Mondal S, Bhowmick D, Dey S, Majumder GC, Mukherjee B, Bhattacharyya D. Epididymal protein ASF is a D-galactose-specific lectin with apoptotic effect on human breast cancer cell line MCF7. Int J Biol Macromol 2015; 84:208-20. [PMID: 26706839 DOI: 10.1016/j.ijbiomac.2015.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 11/27/2022]
Abstract
Isolated caprine epididymal plasma glycoprotein "anti sticking factor" (ASF) interacts with caudal sperm surface in a D-galactose dependent manner. ASF acts as a Ca(2+) dependent soluble lectin principally activated in acidic pH. As a D-galactose specific lectin, it has a specific affinity for fibronectin as well as fibronectin receptor, i.e. integrins α5β3 and α5β1. By virtue of this particular property, it hampers the in vitro adhesion of the adherent breast cancer cell MCF7 with fibronectin. The effective anti-adhesive concentration of ASF promotes p53 dependent apoptosis in MCF7, which was established by Hoechst 33342 staining, DNA fragmentation assay, FITC tagged Annexin-V flowcytometry and western blot analysis. We suggest that ASF inhibits fibronectin-integrin interactions by binding with them and induces adhesion dependent apoptosis on adherent MCF7.
Collapse
Affiliation(s)
- Debarun Roy
- Division of Cryobiology, Centre for Rural and Cryogenic Technologies, Jadavpur University, Kolkata, 700032 West Bengal, India
| | - Kaushik Das
- Cell Biology and Physiology Division, Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
| | - Subhasish Mondal
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 West Bengal, India
| | - Debajit Bhowmick
- CU-BD Center of Excellence for Nanobiotechnology, Centre for Research in Nanoscience and Nanotechnology, Calcutta University, JD-2, Sector-III, Kolkata, 700098 West Bengal, India
| | - Souvik Dey
- Division of Cryobiology, Centre for Rural and Cryogenic Technologies, Jadavpur University, Kolkata, 700032 West Bengal, India
| | - Gopal C Majumder
- Division of Cryobiology, Centre for Rural and Cryogenic Technologies, Jadavpur University, Kolkata, 700032 West Bengal, India
| | - Biswajit Mukherjee
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 West Bengal, India
| | - Debdas Bhattacharyya
- Division of Cryobiology, Centre for Rural and Cryogenic Technologies, Jadavpur University, Kolkata, 700032 West Bengal, India.
| |
Collapse
|
28
|
Liu Y, Gu X, Lin Q, Tian T, Shao L, Yuan C, Zhang B, Fan K. Prognostic significance of osteopontin in patients with non-small cell lung cancer: results from a meta-analysis. Int J Clin Exp Med 2015; 8:12765-12773. [PMID: 26550190 PMCID: PMC4612875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/03/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUNDS Non-small cell lung cancer (NSCLC) is one of the most common malignancies with a high mortality level. Recently, a variety of studies explored the role of osteopontin (OPN) expression in the prognosis of NSCLC, but the results were controversial. METHODS We performed a meta-analysis of eligible studies to evaluate the prognostic significance of OPN expression in NSCLC patients. In order to assess the association between OPN and OS and DFS/PFS, hazard ratio (HR) with 95% confidence interval (CI) was calculated. RESULTS A total of ten studies comprising 1420 patients were included in the meta-analysis. The summary results indicated that high OPN expression was a poor predictor for OS (HR = 2.19, 95% CI: 1.6-2.98), and DFS/PFS (HR = 2, 95% CI: 1.66-2.41). Subgroup analysis revealed that high OPN expression was a negative prognostic marker for OS and DFS/PFS regardless of ethnicity background, treatment and OPN detection method. CONCLUSION Our results showed that increased OPN expression significantly correlated with poor OS and DPS/PFS in NSCLC patients.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cancer Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Xiaobin Gu
- Department of Cancer Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Qunying Lin
- Department of Respiratory Medicine, Hospital of Putian UniversityPutian, Fujian, China
| | - Tian Tian
- Department of Cancer Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Lijuan Shao
- School of Medicine, Nankai UniversityTianjin, China
| | - Chao Yuan
- Department of Cancer Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
| | - Bo Zhang
- Department of Cancer Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
- Department of International Joint Cancer Institute, The Second Military Medical UniversityShanghai, China
| | - Kexing Fan
- Department of Cancer Center, Chinese PLA General Hospital and Chinese PLA Medical SchoolBeijing, China
- Department of International Joint Cancer Institute, The Second Military Medical UniversityShanghai, China
| |
Collapse
|
29
|
Marek I, Volkert G, Hilgers KF, Bieritz B, Rascher W, Reinhardt DP, Hartner A. Fibrillin-1 and alpha8 integrin are co-expressed in the glomerulus and interact to convey adhesion of mesangial cells. Cell Adh Migr 2015; 8:389-95. [PMID: 25482639 DOI: 10.4161/cam.28988] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Fibrillin-1 is a microfibrillar extracellular matrix protein that was described to be a ligand for α8 integrin. α8 integrin is a matrix receptor specifically expressed in mesangial and smooth muscle cells of the kidney. In previous studies we detected glomerular expression of fibrillin-1. Moreover, fibrillin-1 promoted adhesion, migration, and proliferation of mesangial cells. We hypothesized that fibrillin-1 and α8 integrin might interact in the glomerulus, and thus, regulate mesangial cell properties. Our studies showed that fibrillin-1 and α8 integrin colocalize in the glomerular mesangium. Induction of experimental glomerulonephritis led to an increase of both fibrillin-1 and α8 integrin expression. In vitro studies revealed that mesangial cells deficient for α8 integrin adhere weaker to fibrillin-1 and migrate more easily on fibrillin-1 than wild-type mesangial cells. Baseline proliferation on fibrillin-1 is higher in α8 integrin-deficient mesangial cells, but the induction of proliferation is not different in α8 integrin-deficient and wild-type mesangial cells. We conclude that fibrillin-1 and α8 integrin interact, and thus, regulate mesangial cell adhesion and migration. The concomitant induction of both fibrillin-1 and α8 integrin in a self-limited model of glomerular injury points to a protective role of the interaction of fibrillin-1 with α8 integrin in the glomerulus resulting in reduced damage of the glomerular tuft as a consequence of firm adhesion of mesangial cells.
Collapse
Affiliation(s)
- Ines Marek
- a Department for Pediatrics and Adolescent Medicine ; University Hospital of Erlangen ; Erlangen , Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Menendez-Castro C, Cordasic N, Neureiter D, Amann K, Marek I, Volkert G, Stintzing S, Jahn A, Rascher W, Hilgers KF, Hartner A. Under-expression of α8 integrin aggravates experimental atherosclerosis. J Pathol 2015; 236:5-16. [PMID: 25511181 DOI: 10.1002/path.4501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/28/2014] [Accepted: 12/09/2014] [Indexed: 11/08/2022]
Abstract
Integrins play an important role in vascular biology. The α8 integrin chain attenuates smooth muscle cell migration but its functional role in the development of atherosclerosis is unclear. Therefore, we studied the contribution of α8 integrin to atherosclerosis and vascular remodelling. We hypothesized that α8 integrin expression is reduced in atherosclerotic lesions, and that its under-expression leads to a more severe course of atherosclerosis. α8 Integrin was detected by immunohistochemistry and qPCR and α8 integrin-deficient mice were used to induce two models of atherosclerotic lesions. First, ligation of the carotid artery led to medial thickening and neointima formation, which was quantified in carotid cross-sections. Second, after crossing into ApoE-deficient mice, the formation of advanced vascular lesions with atherosclerotic plaques was quantified in aortic en face preparations stained with Sudan IV. Parameters of renal physiology and histopathology were assessed: α8 integrin was detected in the media of human and murine vascular tissue and was down-regulated in arteries with advanced atherosclerotic lesions. In α8 integrin-deficient mice (α8(-/-) ) as well as α8(+/-) and α8(+/+) littermates, carotid artery ligation increased media:lumen ratios in all genotypes, with higher values in ligated α8(-/-) and α8(+/-) compared to ligated α8(+/+) animals. Carotid artery ligation increased smooth muscle cell number in the media of α8(+/+) mice and, more prominently, of α8(-/-) or α8(+/-) mice. On an ApoE(-/-) background, α8(+/-) and α8(-/-) mice developed more atherosclerotic plaques than α8(+/+) mice. α8 Integrin expression was reduced in α8(+/-) animals. Renal damage with increased serum creatinine and glomerulosclerosis was detected in α8(-/-) mice only. Thus, under-expression of α8 integrin aggravates vascular lesions, while a complete loss of α8 integrin results in reduced renal mass and additional renal disease in the presence of generalized atherosclerosis. Our data support the hypothesis that integrin α8β1 has a protective role in arterial remodelling and atherosclerosis.
Collapse
Affiliation(s)
- Carlos Menendez-Castro
- Department of Paediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Johnson GA, Burghardt RC, Bazer FW. Osteopontin: a leading candidate adhesion molecule for implantation in pigs and sheep. J Anim Sci Biotechnol 2014; 5:56. [PMID: 25671104 PMCID: PMC4322467 DOI: 10.1186/2049-1891-5-56] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/25/2014] [Indexed: 11/10/2022] Open
Abstract
Osteopontin (OPN; also known as Secreted Phosphoprotein 1, SPP1) is a secreted extra-cellular matrix (ECM) protein that binds to a variety of cell surface integrins to stimulate cell-cell and cell-ECM adhesion and communication. It is generally accepted that OPN interacts with apically expressed integrin receptors on the uterine luminal epithelium (LE) and conceptus trophectoderm to attach the conceptus to the uterus for implantation. Research conducted with pigs and sheep has significantly advanced understanding of the role(s) of OPN during implantation through exploitation of the prolonged peri-implantation period of pregnancy when elongating conceptuses are free within the uterine lumen requiring extensive paracrine signaling between conceptus and endometrium. This is followed by a protracted and incremental attachment cascade of trophectoderm to uterine LE during implantation, and development of a true epitheliochorial or synepitheliochorial placenta exhibited by pigs and sheep, respectively. In pigs, implanting conceptuses secrete estrogens which induce the synthesis and secretion of OPN in adjacent uterine LE. OPN then binds to αvβ6 integrin receptors on trophectoderm, and the αvβ3 integrin receptors on uterine LE to bridge conceptus attachment to uterine LE for implantation. In sheep, implanting conceptuses secrete interferon tau that prolongs the lifespan of CL. Progesterone released by CL then induces OPN synthesis and secretion from the endometrial GE into the uterine lumen where OPN binds integrins expressed on trophectoderm (αvβ3) and uterine LE (identity of specific integrins unknown) to adhere the conceptus to the uterus for implantation. OPN binding to the αvβ3 integrin receptor on ovine trophectoderm cells induces in vitro focal adhesion assembly, a prerequisite for adhesion and migration of trophectoderm, through activation of: 1) P70S6K via crosstalk between FRAP1/MTOR and MAPK pathways; 2) MTOR, PI3K, MAPK3/MAPK1 (Erk1/2) and MAPK14 (p38) signaling to stimulate trohectoderm cell migration; and 3) focal adhesion assembly and myosin II motor activity to induce migration of trophectoderm cells. Further large in vivo focal adhesions assemble at the uterine-placental interface of both pigs and sheep and identify the involvement of sizable mechanical forces at this interface during discrete periods of trophoblast migration, attachment and placentation in both species.
Collapse
Affiliation(s)
- Greg A Johnson
- />Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Robert C Burghardt
- />Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Fuller W Bazer
- />Department of Animal Science, Texas A&M University, College Station, TX 77843 USA
| |
Collapse
|
32
|
Kiyozumi D, Sato-Nishiuchi R, Sekiguchi K. In Situ Detection of Integrin Ligands. CURRENT PROTOCOLS IN CELL BIOLOGY 2014; 65:9.7.1-17. [PMID: 26061156 DOI: 10.1002/0471143030.cb0907s65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Integrins are cell surface receptors for cell adhesion. Integrin-mediated cell adhesion regulates various cellular processes, including cell survival, migration, proliferation, and differentiation. In vivo, ligands for integrins are immobilized within extracellular matrices, insoluble sheet-like or fibrous supramolecular complexes that associate with or surround cells. To better understand the molecular basis of integrin-mediated regulation of cellular behavior in vivo, it is of critical importance to collect information regarding the activities as well as spatial distributions of integrin ligands in situ. This unit describes a protocol for detecting the spatial distribution of the complement of integrin ligands in situ by overlaying soluble recombinant integrins.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ryoko Sato-Nishiuchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Kiyotoshi Sekiguchi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
33
|
Kiyozumi D, Sato-Nishiuchi R, Sekiguchi K. In situ detection of integrin ligands. CURRENT PROTOCOLS IN CELL BIOLOGY 2014; 65:10.19.1-17. [PMID: 25447073 DOI: 10.1002/0471143030.cb1019s65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Integrins are cell surface receptors for cell adhesion. Integrin-mediated cell adhesion regulates various cellular processes, including cell survival, migration, proliferation, and differentiation. In vivo, ligands for integrins are immobilized within extracellular matrices, insoluble sheet-like or fibrous supramolecular complexes that associate with or surround cells. To better understand the molecular basis of integrin-mediated regulation of cellular behavior in vivo, it is of critical importance to collect information regarding the activities as well as spatial distributions of integrin ligands in situ. This unit describes a protocol for detecting the spatial distribution of the complement of integrin ligands in situ by overlaying soluble recombinant integrins.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Laboratory of Extracellular Matrix Biochemistry, Institute for Protein Research, Osaka University, Osaka, Japan
| | | | | |
Collapse
|
34
|
Zhang F, Luo W, Li Y, Gao S, Lei G. Role of osteopontin in rheumatoid arthritis. Rheumatol Int 2014; 35:589-95. [PMID: 25163663 DOI: 10.1007/s00296-014-3122-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 08/22/2014] [Indexed: 01/01/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by joint swelling, joint tenderness, and destruction of synovial joints, leading to severe disability and premature mortality. RA is a multifactorial disease with genetic, environmental, and stochastic components related to its susceptibility. It has been demonstrated that the expression of osteopontin (OPN) is upregulated in the RA patients. Numerous studies have indicated that the full-length OPN or even OPN fragments, such as thrombin-cleaved OPN and its receptors, play the key roles in RA pathogenesis. Therapeutic application of siRNA to target OPN or neutralizing antibodies related to OPN epitopes in RA animal models are in progress, and some results are encouraging. However, there is a long way to go along with the clinical trials. This review focuses on the recent development in research associated with the OPN role in the pathogenesis of RA and provides insights concerning the OPN targeting as therapeutic approaches for patients with RA.
Collapse
Affiliation(s)
- Fangjie Zhang
- Department of Orthopaedics, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | | | | | | | | |
Collapse
|
35
|
Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol 2014; 37:1-14. [PMID: 25064829 PMCID: PMC4379989 DOI: 10.1016/j.matbio.2014.07.005] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
The concept of a matricellular protein was first proposed by Paul Bornstein in the mid-1990s to account for the non-lethal phenotypes of mice with inactivated genes encoding thrombospondin-1, tenascin-C, or SPARC. It was also recognized that these extracellular matrix proteins were primarily counter or de-adhesive. This review reappraises the matricellular concept after nearly two decades of continuous investigation. The expanded matricellular family as well as the diverse and often unexpected functions, cellular location, and interacting partners/receptors of matricellular proteins are considered. Development of therapeutic strategies that target matricellular proteins are discussed in the context of pathology and regenerative medicine.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States.
| | | |
Collapse
|
36
|
Kaleta B. Role of osteopontin in systemic lupus erythematosus. Arch Immunol Ther Exp (Warsz) 2014; 62:475-82. [PMID: 24917428 PMCID: PMC4244532 DOI: 10.1007/s00005-014-0294-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/07/2014] [Indexed: 12/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multisystemic disease, caused by a variety of factors, which lead to immunological abnormalities. Osteopontin (OPN) is a pleiotropic protein, important in bone remodeling and immune system signaling. OPN, produced by various cells, including immune cells, plays a key role in regulating T-helper 1/T-helper 2 balance, stimulating B lymphocytes to produce antibodies, regulating macrophages, neutrophils and inducing dendritic cells. OPN expression is influenced by genetic polymorphisms of its promoter, hormones and cytokines. Over expression of OPN has been associated with the pathogenesis of immune-mediated diseases. OPN has been implicated in the development of murine model of lupus and in humans with SLE. In this review, I will present current state of research on the role of OPN and OPN gene polymorphisms in pathogenesis and clinical course of SLE. A better understanding of the role of OPN in SLE will contribute to more precise diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Beata Kaleta
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland,
| |
Collapse
|
37
|
Volkert G, Jahn A, Dinkel C, Fahlbusch F, Zürn C, Hilgers KF, Rascher W, Hartner A, Marek I. Contribution of the α8 Integrin Chain to the Expression of Extracellular Matrix Components. ACTA ACUST UNITED AC 2014; 21:89-98. [DOI: 10.3109/15419061.2013.876012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
38
|
Bazer FW, Johnson GA. Pig blastocyst–uterine interactions. Differentiation 2014; 87:52-65. [DOI: 10.1016/j.diff.2013.11.005] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 11/27/2022]
|
39
|
Lund SA, Wilson CL, Raines EW, Tang J, Giachelli CM, Scatena M. Osteopontin mediates macrophage chemotaxis via α4 and α9 integrins and survival via the α4 integrin. J Cell Biochem 2013. [PMID: 23192608 DOI: 10.1002/jcb.24462] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Osteopontin (OPN) is highly expressed by macrophages and plays a key role in the pathology of several chronic inflammatory diseases including atherosclerosis and the foreign body reaction. However, the molecular mechanism behind OPN regulation of macrophage functions is not well understood. OPN is a secreted molecule and interacts with several integrins via two domains: the RGD sequence binding to α(v) -containing integrins, and the SLAYGLR sequence binding to α(4) β(1), α(4) β(7), and α(9) β(1) integrins. Here we determined the role of OPN in macrophage survival, chemotaxis, and activation state. For survival studies, OPN treated-bone marrow derived macrophages (BMDMs) were challenged with growth factor withdrawal and neutralizing integrin antibodies. We found that survival in BMDMs is mediated primarily through the α(4) integrin. In chemotaxis studies, we observed that migration to OPN was blocked by neutralizing α(4) and α(9) integrin antibodies. Further, OPN did not affect macrophage activation as measured by IL-12 production. Finally, the relative contributions of the RGD and the SLAYGLR functional domains of OPN to leukocyte recruitment were evaluated in an in vivo model. We generated chimeric mice expressing mutated forms of OPN in myeloid-derived leukocytes, and found that the SLAYGLR functional domain of OPN, but not the RGD, mediates macrophage accumulation in response to thioglycollate-elicited peritonitis. Collectively, these data indicate that α(4) and α(9) integrins interacting with OPN via the SLAYGLR domain play a key role in macrophage biology by regulating migration, survival, and accumulation.
Collapse
Affiliation(s)
- Susan Amanda Lund
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | | | |
Collapse
|
40
|
Inagaki FF, Tanaka M, Inagaki NF, Yagai T, Sato Y, Sekiguchi K, Oyaizu N, Kokudo N, Miyajima A. Nephronectin is upregulated in acute and chronic hepatitis and aggravates liver injury by recruiting CD4 positive cells. Biochem Biophys Res Commun 2013. [DOI: 10.1016/j.bbrc.2012.11.076] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
41
|
Hartner A, Menendez-Castro C, Cordasic N, Marek I, Volkert G, Klanke B, Rascher W, Hilgers KF. Tubulointerstitial de novo expression of the α8 integrin chain in a rodent model of renal fibrosis--a potential target for anti-fibrotic therapy? PLoS One 2012; 7:e48362. [PMID: 23144868 PMCID: PMC3493553 DOI: 10.1371/journal.pone.0048362] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 09/24/2012] [Indexed: 11/24/2022] Open
Abstract
In the normal kidney, the α8 integrin chain is expressed only on mesangial cells and vascular smooth muscle cells. α8 integrin ligates several matrix molecules including fibronectin, osteopontin and fibrillin-1. Recently, we detected de novo expression of α8 integrin on epithelial cells in renal cysts. We hypothesized that the α8 integrin chain is induced in tubular epithelia undergoing dedifferentiation and contributes to the fibrotic response in the tubulointerstitium (TI) after unilateral ureteral obstruction (UUO). After induction of UUO in rats by ligation of the right ureter, increased expression of the α8 integrin chain and its ligands was observed. In the TI, α8 integrin was localized to cytokeratin-positive epithelial cells and to interstitial fibroblasts; and colocalized with its ligands. In mice underexpressing α8 integrin UUO led to collagen deposition and fibroblast activation comparable to wild types. Mice lacking α8 integrin showed even more TI damage, fibroblast activation and collagen deposition after UUO compared to wild type mice. We conclude that the expression of the α8 integrin chain and its ligands is strongly induced in the TI after UUO, but underexpression of α8 integrin does not attenuate TI fibrosis. Mice lacking the α8 integrin chain are even more susceptible to TI damage than wild type mice. Thus, interactions of α8 integrin with its ligands do not seem to contribute to the development or progression of TI fibrosis in UUO. Targeting α8 integrin might not be a useful approach for anti-fibrotic therapy.
Collapse
Affiliation(s)
- Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Rubert M, Monjo M, Lyngstadaas SP, Ramis JM. Effect of alginate hydrogel containing polyproline-rich peptides on osteoblast differentiation. Biomed Mater 2012; 7:055003. [PMID: 22782012 DOI: 10.1088/1748-6041/7/5/055003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Polyproline-rich synthetic peptides have previously been shown to induce bone formation and mineralization in vitro and to decrease bone resorption in vivo. Alginate hydrogel formulations containing these synthetic peptides (P2, P5, P6) or Emdogain® (EMD) were tested for surface coating of bone implants. In an aqueous environment, the alginate hydrogels disclosed a highly compact structure suitable for cell adhesion and proliferation. Lack of cytotoxicity of the alginate-gel coating containing peptides was tested in MC3T3-E1 cell cultures. In the present study, relative mRNA expression levels of integrin alpha 8 were induced by P5 compared to untreated alginate gel, and osteopontin mRNA levels were increased after 21 days of culture by treatment with synthetic peptides or EMD compared to control. Further, in agreement with previous results when the synthetic peptides were administered in the culture media, osteocalcin mRNA was significantly upregulated after long-term treatment with the formulated synthetic peptides compared to untreated and EMD alginate gel. These results indicate that the alginate gel is a suitable carrier for the delivery of synthetic peptides, and that the formulation is promising as biodegradable and biocompatible coating for bone implants.
Collapse
Affiliation(s)
- M Rubert
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences, University of Balearic Islands, Palma de Mallorca, Spain
| | | | | | | |
Collapse
|
43
|
Bazer FW, Song G, Kim J, Erikson DW, Johnson GA, Burghardt RC, Gao H, Carey Satterfield M, Spencer TE, Wu G. Mechanistic mammalian target of rapamycin (MTOR) cell signaling: effects of select nutrients and secreted phosphoprotein 1 on development of mammalian conceptuses. Mol Cell Endocrinol 2012; 354:22-33. [PMID: 21907263 DOI: 10.1016/j.mce.2011.08.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/16/2011] [Accepted: 08/17/2011] [Indexed: 01/30/2023]
Abstract
Morphological differentiation of uterine glands in mammals is a postnatal event vulnerable to adverse effects of endocrine disruptors. Exposure of ewe lambs to a progestin from birth to postnatal day 56 prevents development of uterine glands and, as adults, the ewes are unable to exhibit estrous cycles or maintain pregnancy. Uterine epithelia secrete proteins and transport nutrients into the uterine lumen necessary for conceptus development, pregnancy recognition signaling and implantation, including arginine and secreted phosphoprotein 1 (SPP1). Arginine can be metabolized to nitric oxide and to polyamines or act directly to activate MTOR cell signaling to stimulate proliferation, migration, and mRNA translation in trophectoderm cells. SPP1 binds αvβ3 and α5β1 integrins and induces focal adhesion assembly, adhesion and migration of conceptus trophectoderm cells during implantation. Thus, arginine and SPP1 mediate growth, migration, cytoskeletal remodeling and adhesion of trophectoderm essential for pregnancy recognition signaling and implantation.
Collapse
Affiliation(s)
- Fuller W Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science, Texas A&M University, College Station, TX, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sánchez-Cortés J, Mrksich M. Using self-assembled monolayers to understand α8β1-mediated cell adhesion to RGD and FEI motifs in nephronectin. ACS Chem Biol 2011; 6:1078-86. [PMID: 21790180 PMCID: PMC3200005 DOI: 10.1021/cb200186j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nephronectin is an extracellular matrix protein that interacts with the α8β1 integrin receptor and plays a role in tissue and organ development, though the motifs that mediate adhesion to the receptor remain unclear. This paper describes the use of self-assembled monolayers to study the adhesion of α8β1-presenting cells to the RGD and DLFEIFEIER ligands in nephronectin and found that both ligands can independently mediate cell adhesion through nonoverlapping binding sites on the integrin. Peptide truncation experiments showed FEI to be the minimal binding sequence within the DLFEIFEIER sequence, and adhesion experiments with peptides that include both the RGD and FEI sequences demonstrate that the two peptides bind synergistically to the receptor. Finally, a peptide array was used to establish a strict requirement for the glutamate residue of FEI and tolerance of other aromatic and hydrophobic residues in the first and third positions, respectively. This work provides an enhanced understanding of the binding of nephronectin with α8β1 and identifies a peptide ligand that can be used for targeting the α8β1 integrin.
Collapse
Affiliation(s)
- Juan Sánchez-Cortés
- Department of Chemistry and Howard Hughes Medical Institute, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Milan Mrksich
- Department of Chemistry and Howard Hughes Medical Institute, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| |
Collapse
|
45
|
Zargham R. Tensegrin in context: Dual role of α8 integrin in the migration of different cell types. Cell Adh Migr 2011; 4:485-90. [PMID: 20543583 DOI: 10.4161/cam.4.4.12403] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
α8β1 integrin is highly expressed in cells with contractile function, such as mesangial cells of the kidneys and vascular smooth muscle cells (VSMCs). Although it promotes migration of neural crest cells and breast cancer cells, recent studies suggest that α8 integrin has a negative regulatory role in VSMC migration. In this review, the question of why α8β1 integrin plays a dual role in cell migration is raised and discussed. It seems that cells require optimum contractility and balanced tensile forces for migration. α8β1 integrin promotes migration of cells that are initially in a less than optimal contractile state (e.g. neural cells) and reduces the migration of cells known as contractile cells. α8β1 integrin can be called “Tensegrin” as it fits perfectly into the tensegrity model (tensional integrity) and seems to play a prominent role in the integration of the tensile forces.
Collapse
Affiliation(s)
- Ramin Zargham
- McGill University, Experimental Medicine Department, Montreal, QC, Canada.
| |
Collapse
|
46
|
Luo X, Ruhland MK, Pazolli E, Lind AC, Stewart SA. Osteopontin stimulates preneoplastic cellular proliferation through activation of the MAPK pathway. Mol Cancer Res 2011; 9:1018-29. [PMID: 21673011 DOI: 10.1158/1541-7786.mcr-10-0472] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alterations in the microenvironment collaborate with cell autonomous mutations during the transformation process. Indeed, cancer-associated fibroblasts and senescent fibroblasts stimulate tumorigenesis in xenograft models. Because senescent fibroblasts accumulate with age, these findings suggest that they contribute to age-related increases in tumorigenesis. Previously we showed that senescence-associated stromal-derived osteopontin contributes to preneoplastic cell growth in vitro and in xenografts, suggesting that it impacts neoplastic progression. Analysis of fibroblasts within premalignant and malignant skin lesions ranging from solar/actinic keratosis to squamous cell carcinoma revealed they express osteopontin. Given the stromal expression of osteopontin, we investigated how osteopontin impacts preneoplastic cell growth. We show that osteopontin promotes preneoplastic keratinocyte cellular proliferation and cell survival through the CD44 cell receptor and activation of the MAPK pathway. These data suggest that stromal-derived osteopontin impacts tumorigenesis by stimulating preneoplastic cell proliferation thus allowing expansion of initiated cells in early lesions.
Collapse
Affiliation(s)
- Xianmin Luo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
The secreted phosphorylated protein osteopontin (OPN) is expressed in a variety of tissues and bodily fluids, and is associated with pathologies including tissue injury, infection, autoimmune disease and cancer. Macrophages are ubiquitous, heterogeneous cells that mediate aspects of cell and tissue damage in all these pathologies. Here, the role of OPN in macrophage function is reviewed. OPN is expressed in macrophage cells in multiple pathologies, and the regulation of its expression in these cells has been described in vitro. The protein has been implicated in multiple functions of macrophages, including cytokine expression, expression of inducible nitric oxide synthase, phagocytosis and migration. Indeed, the role of OPN in cells of the macrophage lineage might underlie its physiological role in many pathologies. However, there are numerous instances where the published literature is inconsistent, especially in terms of OPN function in vitro. Although the heterogeneity of OPN and its receptors, or of macrophages themselves, might underlie some of these inconsistencies, it is important to understand the role of OPN in macrophage biology in order to exploit its function therapeutically.
Collapse
|
48
|
Fujiwara H, Ferreira M, Donati G, Marciano DK, Linton JM, Sato Y, Hartner A, Sekiguchi K, Reichardt LF, Watt FM. The basement membrane of hair follicle stem cells is a muscle cell niche. Cell 2011; 144:577-89. [PMID: 21335239 DOI: 10.1016/j.cell.2011.01.014] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 10/24/2010] [Accepted: 01/10/2011] [Indexed: 12/17/2022]
Abstract
The hair follicle bulge in the epidermis associates with the arrector pili muscle (APM) that is responsible for piloerection ("goosebumps"). We show that stem cells in the bulge deposit nephronectin into the underlying basement membrane, thus regulating the adhesion of mesenchymal cells expressing the nephronectin receptor, α8β1 integrin, to the bulge. Nephronectin induces α8 integrin-positive mesenchymal cells to upregulate smooth muscle markers. In nephronectin knockout mice, fewer arrector pili muscles form in the skin, and they attach to the follicle above the bulge, where there is compensatory upregulation of the nephronectin family member EGFL6. Deletion of α8 integrin also abolishes selective APM anchorage to the bulge. Nephronectin is a Wnt target; epidermal β-catenin activation upregulates epidermal nephronectin and dermal α8 integrin expression. Thus, bulge stem cells, via nephronectin expression, create a smooth muscle cell niche and act as tendon cells for the APM. Our results reveal a functional role for basement membrane heterogeneity in tissue patterning. PAPERCLIP:
Collapse
Affiliation(s)
- Hironobu Fujiwara
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Marek I, Volkert G, Jahn A, Fahlbusch F, Zürn C, Ozcan Z, Goppelt-Struebe M, Hilgers KF, Rascher W, Hartner A. Lack of α8 integrin leads to morphological changes in renal mesangial cells, but not in vascular smooth muscle cells. BMC Cell Biol 2010; 11:102. [PMID: 21194485 PMCID: PMC3022721 DOI: 10.1186/1471-2121-11-102] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 12/31/2010] [Indexed: 11/21/2022] Open
Abstract
Background Extracellular matrix receptors of the integrin family are known to regulate cell adhesion, shape and functions. The α8 integrin chain is expressed in glomerular mesangial cells and in vascular smooth muscle cells. Mice deficient for α8 integrin have structural alterations in glomeruli but not in renal arteries. For this reason we hypothesized that mesangial cells and vascular smooth muscle cells differ in their respective capacity to compensate for the lack of α8 integrin. Results Wild type and α8 integrin-deficient mesangial cells varied markedly in cell morphology and expression or localization of cytoskeletal molecules. In α8 integrin-deficient mesangial cells α-smooth muscle actin and CTGF were downregulated. In contrast, there were no comparable differences between α8 integrin-deficient and wild type vascular smooth muscle cells. Expression patterns of integrins were altered in α8 integrin-deficient mesangial cells compared to wild type mesangial cells, displaying a prominent overexpression of α2 and α6 integrins, while expression patterns of the these integrins were not different between wild type and α8 integrin-deficient vascular smooth muscle cells, respectively. Cell proliferation was augmented in α8 integrin-deficient mesangial cells, but not in vascular smooth muscle cells, compared to wild type cells. Conclusions Our findings suggest that α8 integrin deficiency has differential effects in mesangial cells and vascular smooth muscle cells. While the phenotype of vascular smooth muscle cells lacking α8 integrin is not altered, mesangial cells lacking α8 integrin differ considerably from wild type mesangial cells which might be a consequence of compensatory changes in the expression patterns of other integrins. This could result in glomerular changes in α8 integrin-deficient mice, while the vasculature is not affected in these mice.
Collapse
Affiliation(s)
- Ines Marek
- Hospital for Children and Adolescents, Universität Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hartner A, Cordasic N, Menendez-Castro C, Volkert G, Yabu JM, Kupraszewicz-Hutzler M, Rascher W, Hilgers KF. Lack of {alpha}8-integrin aggravates podocyte injury in experimental diabetic nephropathy. Am J Physiol Renal Physiol 2010; 299:F1151-7. [PMID: 20826576 DOI: 10.1152/ajprenal.00058.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Development of diabetic nephropathy is accompanied by changes in integrin-mediated cell-matrix interactions. The α8-integrin chain is specifically expressed in mesangial cells of the glomerulus. During experimental hypertension, α8-integrin plays a protective role in the glomerulus. We hypothesized that α8-integrin is involved in maintaining the integrity of the glomerulus in diabetic nephropathy. Experimental streptozotocin (STZ) diabetes led to an increased expression and glomerular deposition of α8-integrin. To test the functional role of α8-integrin, STZ diabetes was induced in mice with a homozygous (α8-/-) or heterozygous (α8+/-) deletion of the α8-integrin gene and in wild-type litters (α8+/+). Blood glucose and mean arterial blood pressure were not different in α8-/- and α8+/+ mice after 6 wk of diabetes. However, diabetic α8-/- mice developed significantly higher albuminuria and more glomerulosclerosis than diabetic α8+/+ mice. Moreover, in diabetic α8-/- mice, the number of glomerular cells staining positive for the podocyte markers WT-1 and vimentin were reduced more prominently than in diabetic α8+/+. The filtration barrier protein nephrin was downregulated in diabetic glomeruli with the strongest reduction observed in α8-/- mice. Taken together, α8-/- mice developed more severe glomerular lesions and podocyte damage after onset of STZ diabetes than α8+/+ mice, indicating that α8-integrin is protective for the structure and function of the glomerulus and maintains podocyte integrity during the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Andrea Hartner
- Dept. of Pediatrics, Loschgestrasse 15, D-91054 Erlangen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|