1
|
Sun Z, Li J, Liu H, Fan Z. Insulin-Like Growth Factor-Binding Protein 5 Promotes the Cell Proliferation and Osteogenic Potential of Dental Pulp Stem Cells Dependent on Its Nuclear Localisation Sequence. J Oral Rehabil 2024; 51:2664-2674. [PMID: 39313926 DOI: 10.1111/joor.13863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVES Dental pulp stem cells (DPSCs) have been extensively used for tissue regeneration owing to their notable capabilities. Insulin-like growth factor-binding protein 5 (IGFBP5) regulates osteogenic differentiation of mesenchymal stem cells (MSCs); however, the underlying regulatory mechanisms require further investigation. MATERIALS AND METHODS Carboxyfluorescein succinimidyl ester, an alkaline phosphatase (ALP) activity assay and Alizarin Red staining were used to reveal the role of IGFBP5 in DPSCs. Protein expression levels were determined using western blotting. Immunofluorescence was used to observe cell sub-localisation. Subcutaneous transplantation in nude mice was used to observe the osteogenesis of DPSCs in vivo. RESULTS IGFBP5 enhanced the proliferation and osteogenic differentiation of DPSCs. Deletion of the nuclear localisation sequence (NLS) of IGFBP5 prevented its nuclear import and abolished all its promoting effects on DPSCs; ivermectin stimulation attenuated the enhancement of ALP activity by IGBFP5. Bone-like tissue formation promoted by IGFBP5 in vivo vanishes when the NLS is deleted. Inhibition of IGFBP5 nuclear import attenuated the IGFBP5-induced phosphorylation of JNK (p-JNK) and phosphorylated ERK (p-ERK) in DPSCs. CONCLUSION Our findings suggest that cell proliferation and osteogenic differentiation effects exerted by IGFBP5 on DPSCs are closely associated with their entry into the nucleus, thereby providing a novel potential target for tissue regeneration.
Collapse
Affiliation(s)
- Ziyan Sun
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Preventive Medicine, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Huina Liu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhipeng Fan
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Asgartooran B, Bahadori A, Khamverdi Z, Ayubi E, Farmany A. Effect of different boron contents within boron-doped hydroxyapatite-chitosan nano-composite on the microhardness of demineralized enamel. BMC Oral Health 2024; 24:1419. [PMID: 39574107 PMCID: PMC11580543 DOI: 10.1186/s12903-024-05194-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024] Open
Abstract
OBJECTIVE Dental caries is a prevalent chronic disease globally, arising from an imbalance between tooth remineralization and demineralization. Early detection of lesions is crucial, with non-invasive methods preferred in the initial stages of caries for enamel remineralization and demineralization prevention. This study aimed to assess the impact of varying contents of boron-doped hydroxyapatite-chitosan nano-composite on the microhardness of demineralized enamel. MATERIALS AND METHODS Boron-doped hydroxyapatite-chitosan nano-composites at concentrations of 0%, 5%, 10%, and 15% were prepared using the sol-gel method. Thirty intact human premolars were collected to evaluate tooth remineralization. The teeth were divided into buccal and lingual halves, subjected to pH cycling for 14 days to demineralize the enamel surface. Subsequently, the specimens were randomly divided into four groups (n = 15) based on the application of remineralizing materials: Group 1 - Hydroxyapatite-chitosan nano-composite (HApC); Group 2 - Boron-doped hydroxyapatite-chitosan nano-composite (B@HApC) with 5% boron concentration; Group 3 - Boron-doped hydroxyapatite-chitosan nano-composite (B@HApC) with 10% boron concentration; and Group 4 - Boron-doped hydroxyapatite-chitosan nano-composite (B@HApC) with 15% boron concentration. Microhardness was measured at three points using a Vickers hardness test (300 g force for 10 s). RESULTS The study revealed a significant difference between the nano-composite groups containing 5%, 10%, and 15% boron compared to the control group (p < 0.001). The highest microhardness was observed in the 15% boron group (49.42), with the control group showing the lowest microhardness (20.15). Pairwise comparisons of microhardness indicated a significant difference between Group 4 and the control group (p < 0.05). However, no significant differences were found between Groups 2 and 3, Groups 2 and 4, and Groups 3 and 4 (p > 0.05). Scanning electron microscope (SEM) results supported the microhardness findings, showing a mineral layer covering the porous structure resulting from enamel surface demineralization in the intervention groups, highlighting the effectiveness of boron in enhancing the microhardness of demineralized enamel. CONCLUSION The study demonstrated that a content of 15% boron in B@HApC and higher led to increased microhardness of demineralized enamel. Additionally, content of 5% and 10% boron in the nano-composite also improved microhardness, although not to a statistically significant extent.
Collapse
Affiliation(s)
- Bahare Asgartooran
- Department of Restorative Dentistry, Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amirhossein Bahadori
- Department of Restorative Dentistry, Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Zahra Khamverdi
- Department of Restorative Dentistry, Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Erfan Ayubi
- Social Determinants of Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Farmany
- Dental Implant Research Center, Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
3
|
Mohan S, Kesavan C. T-cell factor 7L2 is a novel regulator of osteoblast functions that acts in part by modulation of hypoxia signaling. Am J Physiol Endocrinol Metab 2022; 322:E528-E539. [PMID: 35466691 PMCID: PMC9169825 DOI: 10.1152/ajpendo.00035.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022]
Abstract
T-cell-like factor (TCF)7l2, a key effector of canonical Wnt signaling, is highly expressed in bone but nothing is known about its role in regulating osteoblast function. To test this, we generated mice with conditional disruption of Tcf7l2 gene in osteoblast lineages using Tcf7l2 floxed and Col1α2-Cre mice. Skeletal parameters were evaluated using heterozygous conditional knockdown (HCKD) mice since homozygous conditional knockout died during pregnancy or immediately after birth. At 5 wk of age, trabecular bone mass of long bones was reduced by 35% as measured by microcomputed tomography (μCT). Histology data showed a 42% reduction in femur trabecular bone mass caused by reduced bone formation. Knockdown of Tcf7l2 expression in osteoblasts decreased proliferation and differentiation by 20%-40%. Expression levels of genes (Hif1α, Vegf, and β-catenin) targeted by TCF7L2 were decreased by 50% in Tcf7l2-deficient osteoblasts and bones of HCKD mice. We found that the Hif1α gene promoter contained multiple putative TCF7L2 motifs and stabilization of HIF1α protein levels rescued expression of TCF7L2 target genes and alkaline phosphatase (ALP) activity in Tcf7l2-deficient osteoblasts. Furthermore, Tcf7l2 overexpression increased proliferation in the presence of canonical Wnt3a that was not affected by β-catenin inhibitor providing evidence for a noncanonical signaling in mediating TCF7L2 effects. Tcf7l2 expression was increased in response to mechanical strain (MS) in vitro and in vivo, and disruption of Tcf7l2 expression in osteoblasts reduced MS-induced ALP activity by 35%. We conclude that Tcf7l2, a mechanoresponsive gene, is an important regulator of osteoblast function acting, in part, via hypoxia signaling.NEW & NOTEWORTHY TCF7L2 is expressed by bone but it was not known whether TCF7L2 expression influenced bone development. By using a mouse model with conditional disruption of Tcf7l2 in osteoblast lineage cells, we have demonstrated for the first time, that TCF7L2 plays an important role in regulating osteoblasts via a noncanonical pathway.
Collapse
Affiliation(s)
- Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, California
- Department of Orthopedics, School of Medicine, Loma Linda University, Loma Linda, California
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California
- Department of Medicine, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
4
|
Dittmer J. Biological effects and regulation of IGFBP5 in breast cancer. Front Endocrinol (Lausanne) 2022; 13:983793. [PMID: 36093095 PMCID: PMC9453429 DOI: 10.3389/fendo.2022.983793] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The insulin-like growth factor receptor (IGF1R) pathway plays an important role in cancer progression. In breast cancer, the IGF1R pathway is linked to estrogen-dependent signaling. Regulation of IGF1R activity is complex and involves the actions of its ligands IGF1 and IGF2 and those of IGF-binding proteins (IGFBPs). Six IGFBPs are known that share the ability to form complexes with the IGFs, by which they control the bioavailability of these ligands. Besides, each of the IGFBPs have specific features. In this review, the focus lies on the biological effects and regulation of IGFBP5 in breast cancer. In breast cancer, estrogen is a critical regulator of IGFBP5 transcription. It exerts its effect through an intergenic enhancer loop that is part of the chromosomal breast cancer susceptibility region 2q35. The biological effects of IGFBP5 depend upon the cellular context. By inhibiting or promoting IGF1R signaling, IGFBP5 can either act as a tumor suppressor or promoter. Additionally, IGFBP5 possesses IGF-independent activities, which contribute to the complexity by which IGFBP5 interferes with cancer cell behavior.
Collapse
|
5
|
Singh AK, Kewalramani N, Mani V, Sharma A, Kumari P, Pal RP. Effects of boric acid supplementation on bone health in crossbred calves under tropical condition. J Trace Elem Med Biol 2021; 63:126647. [PMID: 33010650 DOI: 10.1016/j.jtemb.2020.126647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/13/2020] [Accepted: 09/11/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Boron (B) is thought to play key role in proper bone growth and development as well as have some role in regulation of minerals such as calcium (Ca), phosphorus (P) and magnesium (Mg) which act synergistically with vitamin D. OBJECTIVE Present study was planned in two phases to assess the effect of optimum and supranutritional levels of (B) in the form of boric acid (BA) supplementation on bone health of growing cross bred calves. METHOD During Phase-1, twenty four male crossbred calves were blocked into four groups (n = 6) on the basis of their body weight (154.83 ± 8.5 kg), age (7-9 months) and were supplemented with 0 (C), 2.6 (T-1), 5.4 (T-2) and 10.7 (T-3) g BA for appropriate B (0.175 adjustment factor to calculate B form BA) consumption i.e. 0, 100, 200 and 400 ppm in each group respectively, for 90 days. During phase 2, twenty-one male crossbred calves were divided into 3 groups (n = 7) on the basis of their body weight (103.76 ± 4.34 kg) and age (5-8 months). All the groups were on similar dietary regimen with additional supplementation of boric acid as 0 g (control); 3.6 g (200 ppm B; T-1) and 10.8 g (600 ppm B; T-2), respectively for a period of 120 d. RESULTS From the first experiment it is reported that plasma levels of bovine alkaline phosphatase (BALP), type I collagen cross-linked N-telopeptide (NTx) and Ca were significantly (P < 0.05) affected in T-2 and T-3 groups as compared to T-1 and control groups. Whereas, plasma osteocalcin (OCN) concentration was found to be higher in T-2 and T-3 groups as compared to control group. However, plasma concentrations (ng/mL) of tartrate resistant acid phosphatase (TRAP) remained unaltered due to dietary treatments. Based on the results, another experiment was conducted to validate the above findings and further to determine the effect of still higher i.e supranutritional levels of BA supplementation on bone health of calves. Results revealed that supplementation of BA in T-2 group had no beneficial effect on bone health as the plasma concentration of BALP, OCN, NTx, 25 (OH) vitamin D and Ca as compared to T-1 group in phase 2. Other possible attributes of bone health i.e. plasma concentration of Mg, P, parathyroid hormone (PTH), and calcitonin were not affected by BA supplementation at any levels. CONCLUSION Overall from present study it can be concluded that supplementation of boric acid 3.6 g/d (equivalent to 200 ppm B) in the diet of growing animals has positive effect on bone health related biomarkers (OCN, NTx and BALP) and supplementation of supranutritional level of BA i.e. 10.8 g (equivalent to 600 ppm B) level had neither additional beneficial nor harmful effect on bone health of calves.
Collapse
Affiliation(s)
| | | | - Veena Mani
- National Dairy Research Institute, Karnal, Haryana, India
| | - Amit Sharma
- National Dairy Research Institute, Karnal, Haryana, India
| | - Punita Kumari
- Animal and Fisheries Resources Department, Government of Bihar, India
| | | |
Collapse
|
6
|
Yasuoka H, Garrett SM, Nguyen XX, Artlett CM, Feghali-Bostwick CA. NADPH oxidase-mediated induction of reactive oxygen species and extracellular matrix deposition by insulin-like growth factor binding protein-5. Am J Physiol Lung Cell Mol Physiol 2019; 316:L644-L655. [PMID: 30810066 DOI: 10.1152/ajplung.00106.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor binding protein-5 (IGFBP-5) induces production of the extracellular matrix (ECM) components collagen and fibronectin both in vitro and in vivo and is overexpressed in patients with fibrosing lung diseases, such as idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc). However, the mechanism by which IGFBP-5 exerts its fibrotic effect is incompletely understood. Recent reports have shown a substantial role of reactive oxygen species (ROS) in fibrosis; thus we hypothesized that IGFBP-5 induces production of ROS to mediate the profibrotic process. In vitro analyses revealed that ROS production was induced by recombinant and adenoviral vector-mediated IGFBP-5 (AdBP5) in a dose- and time-dependent manner, regulated through MEK/ERK and JNK signaling, and primarily mediated by NADPH oxidase (Nox). Silencing IGFBP-5 in SSc and IPF fibroblasts reduced ROS production. The antioxidants diphenyleneiodonium and N-acetylcysteine blocked IGFBP-5-stimulated ECM production in normal, SSc, and IPF human primary lung fibroblasts. In murine fibroblasts lacking critical components of the Nox machinery, AdBP5-stimulated ROS production and fibronectin expression were reduced compared with wild-type fibroblasts. IGFBP-5 stimulated transcriptional expression of Nox3 in human fibroblasts while selective knockdown of Nox3 reduced ROS production by IGFBP-5. Thus IGFBP-5 mediates fibrosis through production of ROS in a Nox-dependent manner.
Collapse
Affiliation(s)
- Hidekata Yasuoka
- Department of Internal Medicine, Division of Rheumatology, Fujita Health University School of Medicine , Aichi , Japan
| | - Sara M Garrett
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina , Charleston, South Carolina
| | - Xinh-Xinh Nguyen
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina , Charleston, South Carolina
| | - Carol M Artlett
- Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Carol A Feghali-Bostwick
- Department of Medicine, Division of Rheumatology and Immunology, Medical University of South Carolina , Charleston, South Carolina
| |
Collapse
|
7
|
Araújo T, Khayat A, Quintana L, Calcagno D, Mourão R, Modesto A, Paiva J, Lima A, Moreira F, Oliveira E, Souza M, Othman M, Liehr T, Abdelhay E, Gomes R, Santos S, Assumpção P. Piwi like RNA-mediated gene silencing 1 gene as a possible major player in gastric cancer. World J Gastroenterol 2018; 24:5338-5350. [PMID: 30598579 PMCID: PMC6305533 DOI: 10.3748/wjg.v24.i47.5338] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/07/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To establish a permanent piwi like RNA-mediated gene silencing 1 (PIWIL1) gene knockout in AGP01 gastric cancer cell line using CRISPR-Cas9 system and analyze phenotypic modifications as well as gene expression alterations.
METHODS CRISPR-Cas9 system used was purchased from Dharmacon GE Life Sciences (Lafayette, CO, United States) and permanent knockout was performed according to manufacturer’s recommendations. Wound-healing assay was performed to investigate the effect of PIWIL1 knockout on migration capability of cells and Boyden chamber invasion assay was performed to investigate the effect on invasion capability. For the gene expression analysis, a one-color microarray-based gene expression analysis kit (Agilent Technologies, Santa Clara, CA, United States) was used according to the protocol provided by the manufacturer.
RESULTS PIWIL1 gene knockout caused a significant decrease in AGP01 migration capacity as well as a significant decrease in cell invasiveness. Moreover, functional analysis based on grouping of all differentially expressed mRNAs identified a total of 35 genes (5 up-regulated and 30 down-regulated) encoding proteins involved in cellular invasion and migration. According to current literature, 9 of these 35 genes (DOCK2, ZNF503, PDE4D, ABL1, ABL2, LPAR1, SMAD2, WASF3 and DACH1) are possibly related to the mechanisms used by PIWIL1 to promote carcinogenic effects related to migration and invasion, since their functions are consistent with the changes observed (being up- or down-regulated after knockout).
CONCLUSION Taken together, these data reinforce the idea that PIWIL1 plays a crucial role in the signaling pathway of gastric cancer, regulating several genes involved in migration and invasion processes; therefore, its use as a therapeutic target may generate promising results in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Taíssa Araújo
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - André Khayat
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Luciana Quintana
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Danielle Calcagno
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Ronald Mourão
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Antônio Modesto
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Juliana Paiva
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Adhara Lima
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Fabiano Moreira
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Edivaldo Oliveira
- Laboratório de Cultura de Tecidos e Citogenética, Instituto Evandro Chagas, Belém 66087-082, Brazil
| | - Michel Souza
- Laboratório de Cultura de Tecidos e Citogenética, Instituto Evandro Chagas, Belém 66087-082, Brazil
| | - Moneeb Othman
- Institute of Human Genetics, Universitätsklinikum Jena, Jena 07747, Germany
| | - Thomas Liehr
- Institute of Human Genetics, Universitätsklinikum Jena, Jena 07747, Germany
| | - Eliana Abdelhay
- Laboratório de Célula Tronco, Centro de Transplante de Medula Óssea, Instituto Nacional de Câncer José Alencar Gomes da Silva, Rio de Janeiro 20230-130, Brazil
| | - Renata Gomes
- Laboratório de Célula Tronco, Centro de Transplante de Medula Óssea, Instituto Nacional de Câncer José Alencar Gomes da Silva, Rio de Janeiro 20230-130, Brazil
| | - Sidney Santos
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| | - Paulo Assumpção
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-000, Brazil
| |
Collapse
|
8
|
Beattie J, Al-Khafaji H, Noer PR, Alkharobi HE, Alhodhodi A, Meade J, El-Gendy R, Oxvig C. Insulin- like Growth Factor-Binding Protein Action in Bone Tissue: A Key Role for Pregnancy- Associated Plasma Protein-A. Front Endocrinol (Lausanne) 2018; 9:31. [PMID: 29503631 PMCID: PMC5820303 DOI: 10.3389/fendo.2018.00031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/23/2018] [Indexed: 11/13/2022] Open
Abstract
The insulin-like growth factor (IGF) axis is required for the differentiation, development, and maintenance of bone tissue. Accordingly, dysregulation of this axis is associated with various skeletal pathologies including growth abnormalities and compromised bone structure. It is becoming increasingly apparent that the action of the IGF axis must be viewed holistically taking into account not just the actions of the growth factors and receptors, but also the influence of soluble high affinity IGF binding proteins (IGFBPs).There is a recognition that IGFBPs exert IGF-dependent and IGF-independent effects in bone and other tissues and that an understanding of the mechanisms of action of IGFBPs and their regulation in the pericellular environment impact critically on tissue physiology. In this respect, a group of IGFBP proteinases (which may be considered as ancillary members of the IGF axis) play a crucial role in regulating IGFBP function. In this model, cleavage of IGFBPs by specific proteinases into fragments with lower affinity for growth factor(s) regulates the partition of IGFs between IGFBPs and cell surface IGF receptors. In this review, we examine the importance of IGFBP function in bone tissue with special emphasis on the role of pregnancy associated plasma protein-A (PAPP-A). We examine the function of PAPP-A primarily as an IGFBP-4 proteinase and present evidence that PAPP-A induced cleavage of IGFBP-4 is potentially a key regulatory step in bone metabolism. We also highlight some recent findings with regard to IGFBP-2 and IGFBP-5 (also PAPP-A substrates) function in bone tissue and briefly discuss the actions of the other three IGFBPs (-1, -3, and -6) in this tissue. Although our main focus will be in bone we will allude to IGFBP activity in other cells and tissues where appropriate.
Collapse
Affiliation(s)
- James Beattie
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
- *Correspondence: James Beattie,
| | - Hasanain Al-Khafaji
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
| | - Pernille R. Noer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Hanaa Esa Alkharobi
- Department of Oral Biology, Dental College, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Aishah Alhodhodi
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
| | - Josephine Meade
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
| | - Reem El-Gendy
- Division of Oral Biology, Leeds School of Dentistry, Level 7 Wellcome Trust Brenner Building, University of Leeds, St James University Hospital, Leeds, United Kingdom
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Wang Y, Jia Z, Diao S, Lin X, Lian X, Wang L, Dong R, Liu D, Fan Z. IGFBP5 enhances osteogenic differentiation potential of periodontal ligament stem cells and Wharton's jelly umbilical cord stem cells, via the JNK and MEK/Erk signalling pathways. Cell Prolif 2016; 49:618-627. [DOI: 19.doi: 10.1111/cpr.12284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025] Open
Abstract
AbstractObjectivesMesenchymal stem cell (MSC)‐mediated tissue regeneration represents a promising strategy for repair of tissue defects, but its molecular mechanisms remain unclear, restricting the use of MSCs. Our previous study indicated that insulin‐like growth factor‐binding protein 5 (IGFBP5) exerted a valuable effect on osteogenic differentiation of MSCs, but its molecular mechanisms underlying directed differentiation remained unclear. In this study, we have investigated the molecular role of IGFBP5 in regulating this osteogenic differentiation potential.Materials and methodsPeriodontal ligament stem cells (PDLSCs) were isolated from periodontal ligament tissue. Wharton's jelly of umbilical cord stem cells (WJCMSCs) was obtained commercially. Lentiviral IGFBP5 shRNA was used to silence IGFBP5. Retroviruses expressing wild‐type IGFBP5 were used to overexpress IGFBP5 in the WJCMSCs. Recombinant human IGFBP5 protein (rhIGFBP5) was used to treat PDLSCs for 24 h. Western blot analysis was used to detect the MAPK signalling pathway, and alkaline phosphatase (ALP) activity, Alizarin Red staining and quantitative calcium analysis were used to study osteogenic differentiation potentials.ResultsOverexpression of IGFBP5 or rhIGFBP5 increased expression levels of phosphorylated c‐Jun N‐terminal kinase (p‐JNK), phosphorylated mitogen‐activated protein kinase 1 and 2 (p‐MEK1/2) and phosphorylated extracellular regulated protein kinases (p‐Erk1/2) in both WJCMSCs and PDLSCs. Consistently, silenced IGFBP5 was found to effectively inhibit expression of p‐JNK, p‐Erk1/2 and p‐MEK1/2 in PDLSCs and WJCMSCs. Furthermore, inhibition of JNK by its inhibitor, SP600125, or MEK/Erk signalling by its inhibitor, PD98059, dramatically blocked IGFBP5‐enhanced ALP activity and in vitro mineralization in both PDLSCs and WJCMSCs.ConclusionsOur results demonstrated that IGFBP5 promoted osteogenic differentiation potentials of PDLSCs and WJCMSCs via the JNK and MEK/Erk signalling pathways.
Collapse
Affiliation(s)
- Yuejun Wang
- Department of Endodontics Tianjin Medical University School of Stomatology Tianjin China
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| | - Zhi Jia
- Department of Endodontics Tianjin Medical University School of Stomatology Tianjin China
| | - Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| | - Xiao Lin
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| | - Xiaomeng Lian
- Department of Stomatology Beijing Shijitan hospital Capital Medical University Beijing China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| | - Dayong Liu
- Department of Endodontics Tianjin Medical University School of Stomatology Tianjin China
- Molecular Laboratory for Gene Therapy and Tooth Regeneration Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction Capital Medical University School of Stomatology Beijing China
| |
Collapse
|
10
|
Wang Y, Jia Z, Diao S, Lin X, Lian X, Wang L, Dong R, Liu D, Fan Z. IGFBP5 enhances osteogenic differentiation potential of periodontal ligament stem cells and Wharton's jelly umbilical cord stem cells, via the JNK and MEK/Erk signalling pathways. Cell Prolif 2016; 49:618-27. [PMID: 27484838 DOI: 10.1111/cpr.12284] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 07/11/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Mesenchymal stem cell (MSC)-mediated tissue regeneration represents a promising strategy for repair of tissue defects, but its molecular mechanisms remain unclear, restricting the use of MSCs. Our previous study indicated that insulin-like growth factor-binding protein 5 (IGFBP5) exerted a valuable effect on osteogenic differentiation of MSCs, but its molecular mechanisms underlying directed differentiation remained unclear. In this study, we have investigated the molecular role of IGFBP5 in regulating this osteogenic differentiation potential. MATERIALS AND METHODS Periodontal ligament stem cells (PDLSCs) were isolated from periodontal ligament tissue. Wharton's jelly of umbilical cord stem cells (WJCMSCs) was obtained commercially. Lentiviral IGFBP5 shRNA was used to silence IGFBP5. Retroviruses expressing wild-type IGFBP5 were used to overexpress IGFBP5 in the WJCMSCs. Recombinant human IGFBP5 protein (rhIGFBP5) was used to treat PDLSCs for 24 h. Western blot analysis was used to detect the MAPK signalling pathway, and alkaline phosphatase (ALP) activity, Alizarin Red staining and quantitative calcium analysis were used to study osteogenic differentiation potentials. RESULTS Overexpression of IGFBP5 or rhIGFBP5 increased expression levels of phosphorylated c-Jun N-terminal kinase (p-JNK), phosphorylated mitogen-activated protein kinase 1 and 2 (p-MEK1/2) and phosphorylated extracellular regulated protein kinases (p-Erk1/2) in both WJCMSCs and PDLSCs. Consistently, silenced IGFBP5 was found to effectively inhibit expression of p-JNK, p-Erk1/2 and p-MEK1/2 in PDLSCs and WJCMSCs. Furthermore, inhibition of JNK by its inhibitor, SP600125, or MEK/Erk signalling by its inhibitor, PD98059, dramatically blocked IGFBP5-enhanced ALP activity and in vitro mineralization in both PDLSCs and WJCMSCs. CONCLUSIONS Our results demonstrated that IGFBP5 promoted osteogenic differentiation potentials of PDLSCs and WJCMSCs via the JNK and MEK/Erk signalling pathways.
Collapse
Affiliation(s)
- Yuejun Wang
- Department of Endodontics, Tianjin Medical University School of Stomatology, Tianjin, China.,Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Zhi Jia
- Department of Endodontics, Tianjin Medical University School of Stomatology, Tianjin, China
| | - Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Xiao Lin
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Xiaomeng Lian
- Department of Stomatology, Beijing Shijitan hospital, Capital Medical University, Beijing, China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.
| | - Dayong Liu
- Department of Endodontics, Tianjin Medical University School of Stomatology, Tianjin, China. .,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.
| |
Collapse
|
11
|
Su Y, Nishimoto T, Feghali-Bostwick C. IGFBP-5 Promotes Fibrosis Independently of Its Translocation to the Nucleus and Its Interaction with Nucleolin and IGF. PLoS One 2015; 10:e0130546. [PMID: 26103640 PMCID: PMC4478026 DOI: 10.1371/journal.pone.0130546] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/21/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Insulin-like growth factor binding protein (IGFBP)-5 levels are increased in systemic sclerosis (SSc) skin and lung. We previously reported that IGFBP-5 is a pro-fibrotic factor that induces extracellular matrix (ECM) production and deposition. Since IGFBP-5 contains a nuclear localization signal (NLS) that facilitates its nuclear translocation, we sought to examine the role of nuclear translocation on the fibrotic activity of IGFBP-5 and identify IGFBP-5 binding partners relevant for its nuclear compartmentalization. METHODS We generated functional wild type IGFBP-5 and IGFBP-5 with a mutated NLS or a mutated IGF binding site. Abrogation of nuclear translocation in the NLS mutant was confirmed using immunofluorescence and immunoblotting of nuclear and cytoplasmic cellular extracts. Abrogation of IGF binding was confirmed using western ligand blot. The fibrotic activity of wild type and mutant IGFBP-5 was examined in vitro in primary human fibroblasts and ex vivo in human skin. We identified IGFBP-5 binding partners using immunoprecipitation and mass spectrometry. We examined the effect of nucleolin on IGFBP-5 localization and function via sequence-specific silencing in primary human fibroblasts. RESULTS Our results show that IGFBP-5-induced ECM production in vitro in primary human fibroblasts is independent of its nuclear translocation. The NLS-mutant also induced fibrosis ex vivo in human skin, thus confirming and extending the in vitro findings. Similar findings were obtained with the IGF-binding mutant. Nucleolin, a nucleolar protein that can serve as a nuclear receptor, was identified as an IGFBP-5 binding partner. Silencing nucleolin reduced IGFBP-5 translocation to the nucleus but did not block the ability of IGFBP-5 to induce ECM production and a fibrotic phenotype. CONCLUSIONS IGFBP-5 transport to the nucleus requires an intact NLS and nucleolin. However, nuclear translocation is not necessary for IGFBP-5 fibrotic activity; neither is IGF binding. Our data provide further insights into the role of cellular compartmentalization in IGFBP-5-induced fibrosis.
Collapse
Affiliation(s)
- Yunyun Su
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Tetsuya Nishimoto
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Carol Feghali-Bostwick
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|
12
|
Beattie J, Hawsawi Y, Alkharobi H, El-Gendy R. IGFBP-2 and -5: important regulators of normal and neoplastic mammary gland physiology. J Cell Commun Signal 2015; 9:151-8. [PMID: 25645979 DOI: 10.1007/s12079-015-0260-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/12/2015] [Indexed: 01/16/2023] Open
Abstract
The insulin-like growth factor (IGF) axis plays an important role in mammary gland physiology. In addition, dysregulation of this molecular axis may have a causal role in the aetiology and development of breast cancer (BC). This report discusses the IGF axis in normal and neoplastic mammary gland with special reference to IGF binding proteins (IGFBPs) -2 and -5. We describe how these high affinity binders of IGF-1 and IGF-2 may regulate local actions of growth factors in an autocrine and/or paracrine manner and how they also have IGF-independent effects in mammary gland. We discuss clinical studies which investigate both the prognostic value of IGFBP-2 and -5 expression in BC and possible involvement of these genes in the development of resistance to adjuvant endocrine therapies.
Collapse
Affiliation(s)
- James Beattie
- Department of Oral Biology, School of Dentistry, St James University Hospital, Level 7, Wellcome Trust Brenner Building, Leeds, LS9 7TF, UK,
| | | | | | | |
Collapse
|
13
|
Reeves ME, Firek M, Chen ST, Amaar YG. Evidence that RASSF1C stimulation of lung cancer cell proliferation depends on IGFBP-5 and PIWIL1 expression levels. PLoS One 2014; 9:e101679. [PMID: 25007054 PMCID: PMC4090148 DOI: 10.1371/journal.pone.0101679] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 06/11/2014] [Indexed: 12/24/2022] Open
Abstract
RASSF1C is a major isoform of the RASSF1 gene, and is emerging as an oncogene. This is in contradistinction to the RASSF1A isoform, which is an established tumor suppressor. We have previously shown that RASSF1C promotes lung cancer cell proliferation and have identified RASSF1C target genes with growth promoting functions. Here, we further report that RASSF1C promotes lung cancer cell migration and enhances lung cancer cell tumor sphere formation. We also show that RASSF1C over-expression reduces the inhibitory effects of the anti-cancer agent, betulinic acid (BA), on lung cancer cell proliferation. In previous work, we demonstrated that RASSF1C up-regulates piwil1 gene expression, which is a stem cell self-renewal gene that is over-expressed in several human cancers, including lung cancer. Here, we report on the effects of BA on piwil1 gene expression. Cells treated with BA show decreased piwil1 expression. Also, interaction of IGFBP-5 with RASSF1C appears to prevent RASSF1C from up-regulating PIWIL1 protein levels. These findings suggest that IGFBP-5 may be a negative modulator of RASSF1C/ PIWIL1 growth-promoting activities. In addition, we found that inhibition of the ATM-AMPK pathway up-regulates RASSF1C gene expression.
Collapse
Affiliation(s)
- Mark E. Reeves
- Surgical Oncology Laboratory, Loma Linda VA Medical Center, Loma Linda, California, United States of America
- Department of Surgery, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Matthew Firek
- Surgical Oncology Laboratory, Loma Linda VA Medical Center, Loma Linda, California, United States of America
| | - Shin-Tai Chen
- Musculoskeletal Disease Center, Loma Linda VA Medical Center, Loma Linda, California, United States of America
| | - Yousef G. Amaar
- Surgical Oncology Laboratory, Loma Linda VA Medical Center, Loma Linda, California, United States of America
- Department of Surgery, Loma Linda University School of Medicine, Loma Linda, California, United States of America
- * E-mail:
| |
Collapse
|
14
|
Al-Kharobi H, El-Gendy R, Devine DA, Beattie J. The role of the insulin‑like growth factor (IGF) axis in osteogenic and odontogenic differentiation. Cell Mol Life Sci 2014; 71:1469-76. [PMID: 24232361 PMCID: PMC11113200 DOI: 10.1007/s00018-013-1508-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/25/2013] [Indexed: 11/28/2022]
Abstract
The insulin-like growth factor (IGF) axis is a multicomponent molecular network which has important biological functions in the development and maintenance of differentiated tissue function(s). One of the most important functions of the IGF axis is the control of skeletal tissue metabolism by the finely tuned regulation of the process of osteogenesis. To achieve this, the IGF axis controls the activity of several cell types—osteoprogenitor cells, osteoblasts, osteocytes and osteoclasts to achieve the co-ordinated development of appropriate hard tissue structure and associated matrix deposition. In addition, there is an increasing awareness that the IGF axis also plays a role in the process of odontogenesis (tooth formation). In this review, we highlight some of the key findings in both of these areas. A further understanding of the role of the IGF axis in hard tissue biology may contribute to tissue regeneration strategies in cases of skeletal tissue trauma.
Collapse
Affiliation(s)
- H. Al-Kharobi
- Leeds University School of Dentistry, University of Leeds, Clarendon Way, Leeds, LS2 9LU UK
| | - R. El-Gendy
- Leeds University School of Dentistry, University of Leeds, Clarendon Way, Leeds, LS2 9LU UK
| | - D. A. Devine
- Leeds University School of Dentistry, University of Leeds, Clarendon Way, Leeds, LS2 9LU UK
| | - J. Beattie
- Leeds University School of Dentistry, University of Leeds, Clarendon Way, Leeds, LS2 9LU UK
| |
Collapse
|
15
|
Volodko N, Gordon M, Salla M, Ghazaleh HA, Baksh S. RASSF tumor suppressor gene family: Biological functions and regulation. FEBS Lett 2014; 588:2671-84. [DOI: 10.1016/j.febslet.2014.02.041] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/25/2014] [Accepted: 02/25/2014] [Indexed: 01/22/2023]
|
16
|
Song SE, Kim YW, Kim JY, Lee DH, Kim JR, Park SY. IGFBP5 mediates high glucose-induced cardiac fibroblast activation. J Mol Endocrinol 2013; 50:291-303. [PMID: 23417767 DOI: 10.1530/jme-12-0194] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study examined whether IGF-binding protein 5 (IGFBP5) is involved in the high glucose-induced deteriorating effects in cardiac cells. Cardiac fibroblasts and cardiomyocytes were isolated from the hearts of 1- to 3-day-old Sprague Dawley rats. Treatment of fibroblasts with 25 mM glucose increased the number of cells and the mRNA levels of collagen III, matrix metalloproteinase 2 (MMP2), and MMP9. High glucose increased ERK1/2 activity, and the ERK1/2 inhibitor PD98059 suppressed high glucose-mediated fibroblast proliferation and increased collagen III mRNA levels. Whereas high glucose increased both mRNA and protein levels of IGFBP5 in fibroblasts, high glucose did not affect IGFBP5 protein levels in cardiomyocytes. The high glucose-induced increase in IGFBP5 protein levels was inhibited by PD98059 in fibroblasts. While recombinant IGFBP5 increased ERK phosphorylation, cell proliferation, and the mRNA levels of collagen III, MMP2, and MMP9 in fibroblasts, IGFBP5 increased c-Jun N-terminal kinase phosphorylation and induced apoptosis in cardiomyocytes. The knockdown of IGFBP5 inhibited high glucose-induced cell proliferation and collagen III mRNA levels in fibroblasts. Although high glucose increased IGF1 levels, IGF1 did not increase IGFBP5 levels in fibroblasts. The hearts of Otsuka Long-Evans Tokushima Fatty rats and the cardiac fibroblasts of streptozotocin-induced diabetic rats showed increased IGFBP5 expression. These results suggest that IGFBP5 mediates high glucose-induced profibrotic effects in cardiac fibroblasts.
Collapse
Affiliation(s)
- Seung Eun Song
- Department of Physiology, College of Medicine, Yeungnam University, Daegu 705-717, South Korea
| | | | | | | | | | | |
Collapse
|
17
|
Reeves ME, Aragon RJ, Alfakhouri M, Chen ST, Lowen N, Mohan S, Amaar YG. Ras-Association Domain Family 1C Protein Enhances Breast Tumor Growth in Vivo. CANCER GROWTH AND METASTASIS 2012. [DOI: 10.4137/cgm.s9845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The Ras association domain family 1 (RASSF1) gene is a Ras effector that plays an important role in carcinogenesis. We have previously shown that silencing of RASSF1C decreases and over-expression of RASSF1C increases cell proliferation, migration, and attenuates apoptosis of breast cancer cells in vitro. To further confirm our working hypothesis that RASSF1C may play a role as a growth promoter, we have tested the growth of human breast cancer cells stably over-expressing RASSF1A or RASSF1C in nude mice. Our studies show that breast cancer cells over-expressing HA-RASSF1A developed significantly smaller tumors and cells over-expressing HA-RASSF1C developed significantly larger tumors compared to control cells expressing the vector back bone. We have confirmed the expression of HA-RASSF1A and HA-RASSF1C in tumor tissue using RT-PCR, western blotting and immunohistochemical analyses using HA-antibody. Together, our previous in vitro and current in vivo findings further support our hypothesis that RASSF1C, unlike RASSF1A, is not a tumor suppressor and rather it appears to function as tumor growth promoter in breast cancer cells.
Collapse
Affiliation(s)
- Mark E. Reeves
- Surgical Oncology Laboratory
- Department of Surgery, Loma Linda University, Loma Linda, California
| | - Robert J. Aragon
- Surgical Oncology Laboratory
- Department of Surgery, Loma Linda University, Loma Linda, California
| | | | - Shin-Tai Chen
- Musculoskeletal Disease Center, Loma Linda VA Medical Center
| | - Nancy Lowen
- Musculoskeletal Disease Center, Loma Linda VA Medical Center
| | | | - Yousef G. Amaar
- Surgical Oncology Laboratory
- Department of Surgery, Loma Linda University, Loma Linda, California
| |
Collapse
|
18
|
Reeves ME, Baldwin ML, Aragon R, Baldwin S, Chen ST, Li X, Mohan S, Amaar YG. RASSF1C modulates the expression of a stem cell renewal gene, PIWIL1. BMC Res Notes 2012; 5:239. [PMID: 22591718 PMCID: PMC3512503 DOI: 10.1186/1756-0500-5-239] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 04/02/2012] [Indexed: 02/07/2023] Open
Abstract
Background RASSF1A and RASSF1C are two major isoforms encoded by the Ras association domain family 1 (RASSF1) gene through alternative promoter selection and mRNA splicing. RASSF1A is a well established tumor suppressor gene. Unlike RASSF1A, RASSF1C appears to have growth promoting actions in lung cancer. In this article, we report on the identification of novel RASSF1C target genes in non small cell lung cancer (NSCLC). Methods Over-expression and siRNA techniques were used to alter RASSF1C expression in human lung cancer cells, and Affymetrix-microarray study was conducted using NCI-H1299 cells over-expressing RASSF1C to identify RASSF1C target genes. Results The microarray study intriguingly shows that RASSF1C modulates the expression of a number of genes that are involved in cancer development, cell growth and proliferation, cell death, and cell cycle. We have validated the expression of some target genes using qRT-PCR. We demonstrate that RASSF1C over-expression increases, and silencing of RASSF1C decreases, the expression of PIWIL1 gene in NSCLC cells using qRT-PCR, immunostaining, and Western blot analysis. We also show that RASSF1C over-expression induces phosphorylation of ERK1/2 in lung cancer cells, and inhibition of the MEK-ERK1/2 pathway suppresses the expression of PIWIL1 gene expression, suggesting that RASSF1C may exert its activities on some target genes such as PIWIL1 through the activation of the MEK-ERK1/2 pathway. Also, PIWIL1 expression is elevated in lung cancer cell lines compared to normal lung epithelial cells. Conclusions Taken together, our findings provide significant data to propose a model for investigating the role of RASSF1C/PIWIL1 proteins in initiation and progression of lung cancer.
Collapse
Affiliation(s)
- Mark E Reeves
- Surgical Oncology Laboratory, Loma Linda VA Medical Center, CA 92357, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Xing W, Govoni K, Donahue LR, Kesavan C, Wergedal J, Long C, Bassett JD, Gogakos A, Wojcicka A, Williams GR, Mohan S. Genetic evidence that thyroid hormone is indispensable for prepubertal insulin-like growth factor-I expression and bone acquisition in mice. J Bone Miner Res 2012; 27:1067-79. [PMID: 22513648 PMCID: PMC3399953 DOI: 10.1002/jbmr.1551] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Understanding how bone growth is regulated by hormonal and mechanical factors during early growth periods is important for optimizing the attainment of peak bone mass to prevent or postpone the occurrence of fragility fractures later in life. Using genetic mouse models that are deficient in thyroid hormone (TH) (Tshr(-/-) and Duox2(-/-)), growth hormone (GH) (Ghrhr(lit/lit)), or both (Tshr(-/-); Ghrhr(lit/lit)), we demonstrate that there is an important period prior to puberty when the effects of GH are surprisingly small and TH plays a critical role in the regulation of skeletal growth. Daily administration of T3/T4 during days 5 to 14, the time when serum levels of T3 increase rapidly in mice, rescued the skeletal deficit in TH-deficient mice but not in mice lacking both TH and GH. However, treatment of double-mutant mice with both GH and T3/T4 rescued the bone density deficit. Increased body fat in the TH-deficient as well as TH/GH double-mutant mice was rescued by T3/T4 treatment during days 5 to 14. In vitro studies in osteoblasts revealed that T3 in the presence of TH receptor (TR) α1 bound to a TH response element in intron 1 of the IGF-I gene to stimulate transcription. In vivo studies using TRα and TRβ knockout mice revealed evidence for differential regulation of insulin-like growth factor (IGF)-I expression by the two receptors. Furthermore, blockade of IGF-I action partially inhibited the biological effects of TH, thus suggesting that both IGF-I-dependent and IGF-I-independent mechanisms contribute to TH effects on prepubertal bone acquisition.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, Loma Linda VA HealthCare System, Loma Linda, CA 92357
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Kristen Govoni
- Department of Animal Science, University of Connecticut, Storrs, CT 06269
| | - Leah Rae Donahue
- Genetic Resource Science, The Jackson Laboratory, Bar Harbor, ME 04609
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, Loma Linda VA HealthCare System, Loma Linda, CA 92357
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Jon Wergedal
- Musculoskeletal Disease Center, Loma Linda VA HealthCare System, Loma Linda, CA 92357
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| | - Carlin Long
- Cardiology Division, University of Colorado Health Sciences Center, Denver, CO 80204
| | - J.H. Duncan Bassett
- Molecular Endocrinology Group, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
| | - Apostolos Gogakos
- Molecular Endocrinology Group, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
| | - Anna Wojcicka
- Molecular Endocrinology Group, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
| | - Graham R. Williams
- Molecular Endocrinology Group, Department of Medicine, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Loma Linda VA HealthCare System, Loma Linda, CA 92357
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354
| |
Collapse
|
20
|
Sureshbabu A, Okajima H, Yamanaka D, Tonner E, Shastri S, Maycock J, Szymanowska M, Shand J, Takahashi SI, Beattie J, Allan G, Flint D. IGFBP5 induces cell adhesion, increases cell survival and inhibits cell migration in MCF-7 human breast cancer cells. J Cell Sci 2012; 125:1693-705. [PMID: 22328518 DOI: 10.1242/jcs.092882] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024] Open
Abstract
Maintenance of tissue boundaries is crucial for control of metastasis. We describe a new signalling pathway in which epithelial cell disruption can be minimised and thereby restricts epithelial-mesenchymal transgressions. This involves the release of insulin-like growth factor (IGF)-binding protein 5 (IGFBP5) from apoptotic cells, which increases the adhesion of epithelial cells on mesenchymal but not epithelial extracellular matrix (ECM), and involves the direct interaction of IGFBP5 and α2β1 integrins. IGFBP5 also induced cell adhesion to vitronectin in the absence of αVβ3 integrin, the vitronectin receptor, again through an α2β1-integrin-dependent action, suggesting that IGFBP5 can induce spreading on matrices, even in the absence of the integrins normally used in this process. Using IGFBP5 mutants we demonstrate that the effect is IGF-independent but requires the heparin-binding domain in the C-terminus of IGFBP5. A truncated mutant containing only the C-terminal of IGFBP5 also induced adhesion. Adhesion induced by IGFBP5 was dependent on Cdc42 and resulted in activation of integrin-linked kinase (ILK) and Akt. Consistent with these changes, IGFBP5 facilitated prolonged cell survival in nutrient-poor conditions and decreased phosphorylation of the stress-activated kinase p38 MAPK (MAPK14). Whereas IGFBP5 enhanced adhesion, it inhibited cell migration, although this was not evident using the truncated C-terminal mutant, suggesting that effects of IGFBP5 on adhesion and migration involve different mechanisms. We anticipate that these responses to IGFBP5 would reduce the metastatic potential of cells.
Collapse
Affiliation(s)
- Angara Sureshbabu
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Targeted polyubiquitylation of RASSF1C by the Mule and SCFβ-TrCP ligases in response to DNA damage. Biochem J 2012; 441:227-36. [PMID: 21910689 DOI: 10.1042/bj20111500] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
RASSF1A [Ras association (RalGDS/AF-6) domain family member 1A] and RASSF1C are two ubiquitously expressed isoforms of the RASSF1 gene. The promoter of RASSF1A is frequently hypermethylated, resulting in inactivation in various human cancers. RASSF1A is implicated in the regulation of apoptosis, microtubule stability and cell cycle arrest. However, little is known about the regulation and function of RASSF1C. In the present study we show that exogenously expressed RASSF1C is a very unstable protein that is highly polyubiquitylated and degraded via the proteasome. Furthermore, RASSF1C degradation is enhanced when cells are exposed to stress signals, such as UV irradiation. Mule, a HECT (homologous with E6-associated protein C-terminus) family E3 ligase, but not SCFβ-TrCP [where SCF is Skp1 (S-phase kinase-associated protein 1)/cullin/F-box and β-TrCP is β-bransducin repeat-containing protein] or CUL4 (cullin 4)-DDB1 (damage-specific DNA-binding protein 1), is the E3 ligase for RASSF1C under normal conditions, whereas both Mule and SCFβ-TrCP target RASSF1C degradation in response to UV irradiation. GSK3 (glycogen synthase kinase 3) phosphorylates RASSF1C to promote RASSF1C degradation subsequently, which is negatively regulated by the PI3K (phosphoinositide 3-kinase)/Akt pathway. Thus the present study reveals a novel regulation of RASSF1C and the potentially important role of RASSF1C in DNA damage responses.
Collapse
|
22
|
Abstract
Current theories suggest that mitotic checkpoint proteins are essential for proper cellular response to taxanes, a widely-used family of chemotherapeutic compounds. We recently demonstrated that absence or depletion of protein Daxx increases cellular taxol (paclitaxel) resistance—a common trait of patients diagnosed with several malignancies, including breast cancer. Further investigation of Daxx-mediated taxol response revealed that Daxx is important for the proper timing of mitosis progression and cyclin B stability. Daxx interacts with mitotic checkpoint protein Rassf1 and partially co-localizes with this protein during mitosis. Rassf1/Daxx depletion or expression of Daxx binding domain of Rassf1 elevates cyclin B stability and increases taxol resistance in cells and mouse xenograft models. In breast cancer patients, we observed the inverse correlation between Daxx and clinical response to taxane-based chemotherapy. These data suggest that Daxx and Rassf1 define a mitotic stress checkpoint that enables cells to exit mitosis as micronucleated cells (and eventually die) when encountered with specific mitotic stress stimuli, including taxol. Surprisingly, depletion of Daxx or Rassf1 does not change activity of E3 ubiquitin ligase APC/C in in vitro settings, suggesting necessity of mitotic cellular environment for proper activation of this checkpoint. Daxx and Rassf1 may become useful predictive markers for the proper selection of patients for taxane chemotherapy.
Collapse
|
23
|
Xing W, Pourteymoor S, Mohan S. Ascorbic acid regulates osterix expression in osteoblasts by activation of prolyl hydroxylase and ubiquitination-mediated proteosomal degradation pathway. Physiol Genomics 2011; 43:749-57. [PMID: 21467157 DOI: 10.1152/physiolgenomics.00229.2010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mouse genetic studies reveal that ascorbic acid (AA) is essential for osteoblast (OB) differentiation and that osterix (Osx) was a key downstream target of AA action in OBs. To determine the molecular pathways for AA regulation of Osx expression, we evaluated if AA regulates Osx expression by regulating production and/or actions of local growth factors and extracellular matrix (ECM) proteins. Inhibition of actions of IGFs by inhibitory IGFBP-4, BMPs by noggin, and ECM-mediated integrin signaling by RGD did not block AA effects on Osx expression in OBs. Furthermore, blockade of components of MAPK signaling pathway had no effect on AA-induced Osx expression. Because AA is required for prolyl hydroxylase domain (PHD) activity and because PHD-induced prolyl-hydroxylation targets proteins to proteosomal degradation, we next tested if AA effect on Osx expression involves activation of PHD to hydroxylate and induce ubiquitin-proteosome-mediated degradation of transcriptional repressor(s) of Osx gene. Treatment of OBs with dimethyloxallyl glycine and ethyl 3, 4-dihydroxybenzoate, known inhibitors of PHD, completely blocked AA effect on Osx expression and OB differentiation. Knockdown of PHD2 expression by Lentivirus-mediated shRNA abolished AA-induced Osx induction and alkaline phosphatase activity. Furthermore, treatment of OBs with MG115, inhibitor of proteosomal degradation, completely blocked AA effects on Osx expression. Based on these data, we conclude that AA effect on Osx expression is mediated via a novel mechanism that involves PHD2 and proteosomal degradation of a yet to be identified transcriptional repressor that is independent of BMP, IGF-I, or integrin-mediated signaling in mouse OBs.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, Jerry L. Pettis VA Medical Center, California, USA
| | | | | |
Collapse
|
24
|
Reeves ME, Baldwin SW, Baldwin ML, Chen ST, Moretz JM, Aragon RJ, Li X, Strong DD, Mohan S, Amaar YG. Ras-association domain family 1C protein promotes breast cancer cell migration and attenuates apoptosis. BMC Cancer 2010; 10:562. [PMID: 20955597 PMCID: PMC2965177 DOI: 10.1186/1471-2407-10-562] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 10/18/2010] [Indexed: 11/10/2022] Open
Abstract
Background The Ras association domain family 1 (RASSF1) gene is a Ras effector encoding two major mRNA forms, RASSF1A and RASSF1C, derived by alternative promoter selection and alternative mRNA splicing. RASSF1A is a tumor suppressor gene. However, very little is known about the function of RASSF1C both in normal and transformed cells. Methods Gene silencing and over-expression techniques were used to modulate RASSF1C expression in human breast cancer cells. Affymetrix-microarray analysis was performed using T47D cells over-expressing RASSF1C to identify RASSF1C target genes. RT-PCR and western blot techniques were used to validate target gene expression. Cell invasion and apoptosis assays were also performed. Results In this article, we report the effects of altering RASSF1C expression in human breast cancer cells. We found that silencing RASSF1C mRNA in breast cancer cell lines (MDA-MB231 and T47D) caused a small but significant decrease in cell proliferation. Conversely, inducible over-expression of RASSF1C in breast cancer cells (MDA-MB231 and T47D) resulted in a small increase in cell proliferation. We also report on the identification of novel RASSF1C target genes. RASSF1C down-regulates several pro-apoptotic and tumor suppressor genes and up-regulates several growth promoting genes in breast cancer cells. We further show that down-regulation of caspase 3 via overexpression of RASSF1C reduces breast cancer cells' sensitivity to the apoptosis inducing agent, etoposide. Furthermore, we found that RASSF1C over-expression enhances T47D cell invasion/migration in vitro. Conclusion Together, our findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor, but instead may play a role in stimulating metastasis and survival in breast cancer cells.
Collapse
Affiliation(s)
- Mark E Reeves
- Surgical Oncology Laboratory, 11201 Benton Street (151), Loma Linda VA Medical Center, California 92350, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yasuoka H, Hsu E, Ruiz XD, Steinman RA, Choi AMK, Feghali-Bostwick CA. The fibrotic phenotype induced by IGFBP-5 is regulated by MAPK activation and egr-1-dependent and -independent mechanisms. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:605-15. [PMID: 19628764 DOI: 10.2353/ajpath.2009.080991] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have previously shown that insulin-like growth factor (IGF) binding protein- 5 (IGFBP-5) is overexpressed in lung fibrosis and induces the production of extracellular matrix components, such as collagen and fibronectin, both in vitro and in vivo. The exact mechanism by which IGFBP-5 exerts these novel fibrotic effects is unknown. We thus examined the signaling cascades that mediate IGFBP-5-induced fibrosis. We demonstrate for the first time that IGFBP-5 induction of extracellular matrix occurs independently of IGF-I, and results from IGFBP-5 activation of MAPK signaling, which facilitates the translocation of IGFBP-5 to the nucleus. We examined the effects of IGFBP-5 on early growth response (Egr)-1, a transcription factor that is central to growth factor-mediated fibrosis. Egr-1 was up-regulated by IGFBP-5 in a MAPK-dependent manner and bound to nuclear IGFBP-5. In fibroblasts from Egr-1 knockout mice, induction of fibronectin by IGFBP-5 was abolished. Expression of Egr-1 in these cells rescued the extracellular matrix-promoting effects of IGFBP-5. Moreover, IGFBP-5 induced cell migration in an Egr-1-dependent manner. Notably, Egr-1 levels, similar to IGFBP-5, were increased in vivo in lung tissues and in vitro in primary fibroblasts of patients with pulmonary idiopathic fibrosis. Taken together, our findings suggest that IGFBP-5 induces a fibrotic phenotype via the activation of MAPK signaling and the induction of nuclear Egr-1 that interacts with IGFBP-5 and promotes fibrotic gene transcription.
Collapse
Affiliation(s)
- Hidekata Yasuoka
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, NW 628 MUH, 3459 Fifth Avenue, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
26
|
Liu X, Zeng B, Ma J, Wan C. Comparative proteomic analysis of osteosarcoma cell and human primary cultured osteoblastic cell. Cancer Invest 2009; 27:345-52. [PMID: 19212829 DOI: 10.1080/07357900802438577] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
To identify new biomarkers that facilitate the accurate early diagnosis of osteosarcoma and that may possibly include novel therapeutic candidates, we performed a proteomic approach to compare osteosarcoma cells and human primary cultured osteoblastic cells. Image analysis of silver-stained 2-DE gels revealed that the level of 12 protein spots was significantly different between the two groups of samples (p < .004). After mass spectroscopic identification and database searches, we found that in osteosarcoma cells, the level of HSP70, actin capping protein, ATP synthase, Mthsp75, UQCRC1, Ras-related nuclear protein, UCH-L1, and PRDX4 was elevated. However, the level of pyruvate dehydrogenase E1, Prohibitin, and Annexin V was decreased. Subsequent Western blot analyses of UQCRC1, UCH-L1, and PRDX4 in osteosarcoma tissues confirmed the results obtained by the proteomic analyses. These identified proteins may be potential molecular targets for osteosarcomatous diagnostics and therapeutics.
Collapse
Affiliation(s)
- Xudong Liu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai JiaoTong University, Shanghai, China.
| | | | | | | |
Collapse
|
27
|
Yasuoka H, Yamaguchi Y, Feghali-Bostwick CA. The pro-fibrotic factor IGFBP-5 induces lung fibroblast and mononuclear cell migration. Am J Respir Cell Mol Biol 2009; 41:179-88. [PMID: 19131643 DOI: 10.1165/rcmb.2008-0211oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We have previously shown that insulin-like growth factor-binding protein-5 (IGFBP-5) is overexpressed in fibrotic lung tissues and that it induces production of extracellular matrix components such as collagen and fibronectin both in vitro and in vivo. We recently observed mononuclear cell infiltration in lung tissues of mice expressing IGFBP-5. We therefore examined the role of IGFBP-5 on the migration of immune cells. Migration assays demonstrated that IGFBP-5 induced migration of peripheral blood mononuclear cells (PBMCs) in a dose-dependent manner. Preferential migration of monocytes/macrophages, natural killer cells, and T cells was observed. Moreover, the CD4/CD8 ratio of migrating cells was significantly higher in vitro and in vivo in response to IGFBP-5. IGFBP-5 resulted in preferential migration of activated CD4(+) T cells and monocytes. Interestingly, IGFBP-5 also induced migration of primary human lung fibroblasts. Exogenous administration of IGFBP-5 induced activation of mitogen-activated protein kinase (MAPK) signaling cascade but not PI3K in PBMCs. IGFBP-5-induced migration was blocked by the MEK1/2 inhibitor U0126, suggesting that IGFBP-5-induced migration occurs via MAPK activation. Furthermore, monocytes treated with recombinant IGFBP-5 expressed the mesenchymal markers alpha-smooth muscle actin and fibronectin in vitro and in vivo, suggesting that IGFBP-5 can induce the transformation of monocytes into mesenchymal cells. Collectively, our results suggest that IGFBP-5 induces cell migration via MAPK-dependent and IGF-I-independent mechanisms.
Collapse
Affiliation(s)
- Hidekata Yasuoka
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
28
|
Flint DJ, Tonner E, Beattie J, Allan GJ. Role of insulin-like growth factor binding proteins in mammary gland development. J Mammary Gland Biol Neoplasia 2008; 13:443-53. [PMID: 18998203 DOI: 10.1007/s10911-008-9095-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 10/28/2008] [Indexed: 01/08/2023] Open
Abstract
Insulin-like growth factors (IGFs) play an important role in mammary gland development and their effects are, in turn, influenced by a family of 6 IGF-binding proteins (IGFBPs). The IGFBPs are expressed in time- and tissue-specific fashion during the periods of rapid growth and involution of the mammary gland. The precise roles of these proteins in vivo have, however, been difficult to determine. This review examines the indirect evidence (evolution, chromosomal location and roles in lower life-forms) the evidence from in vitro studies and the attempts to examine their roles in vivo, using IGFBP-deficient and over-expression models. Evidence exists for a role of the IGFBPs in inhibition of the survival effects of IGFs as well as in IGF-enhancing effects from in vitro studies. The location of the IGFBPs, often associated with the extracellular matrix, suggests roles as a reservoir of IGFs or as a potential barrier, restricting access of IGFs to distinct cellular compartments. We also discuss the relative importance of IGF-dependent versus IGF-independent effects. IGF-independent effects include nuclear localization, activation of proteases and interaction with a variety of extracellular matrix and cell surface proteins. Finally, we examine the increasing evidence for the IGFBPs to be considered as part of a larger family of extracellular matrix proteins involved in morphogenesis and tissue re-modeling.
Collapse
Affiliation(s)
- D J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0NR, UK.
| | | | | | | |
Collapse
|
29
|
Strohbach C, Kleinman S, Linkhart T, Amaar Y, Chen ST, Mohan S, Strong D. Potential involvement of the interaction between insulin-like growth factor binding protein (IGFBP)-6 and LIM mineralization protein (LMP)-1 in regulating osteoblast differentiation. J Cell Biochem 2008; 104:1890-905. [PMID: 18395833 DOI: 10.1002/jcb.21761] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-like growth factor binding protein (IGFBP)-6 has been reported to inhibit differentiation of myoblasts and osteoblasts. In the current study, we explored the mechanisms underlying IGFBP-6 effects on osteoblast differentiation. During MC3T3-E1 osteoblast differentiation, we found that IGFBP-6 protein was down-regulated. Overexpression of IGFBP-6 in MC3T3-E1 and human bone cells inhibited nodule formation, osteocalcin mRNA expression and ALP activity. Furthermore, accumulation of IGFBP-6 in the culture media was not required for any of these effects suggesting that IGFBP-6 suppressed osteoblast differentiation by an intracellular mechanism. A yeast two-hybrid screen of an osteosarcoma library was conducted to identify intracellular binding partners to account for IGFBP-6 inhibitory effects on osteoblast differentiation. LIM mineralizing protein (LMP-1) was identified as a high affinity IGFBP-6 binding partner. Physical interaction between IGFBP-6 and LMP-1 was confirmed by co-immunoprecipitation. Fluorescent protein fusion constructs for LMP-1 and IGFBP-6 were transiently transfected into osteoblasts to provide evidence of subcellular locations for each protein. Coexpression of LMP-1-GFP and IGFBP-6-RFP resulted in overlapping subcellular localization of LMP-1 and IGFBP-6. To determine if there was a functional association of IGFBP-6 and LMP-1 as well as a physical association, we studied the effect of IGFBP-6, LMP-1 and their combination on type I procollagen promoter activity. LMP-1 increased promoter activity while IGFBP-6 reduced promoter activity, and coexpression of LMP-1 with IGFBP-6 abrogated IGFBP-6 suppression. These studies provide evidence that overexpression of IGFBP-6 suppresses human and murine osteoblast differentiation, that IGFBP-6 and LMP-1 physically interact, and supports the conclusion that this interaction may be functionally relevant.
Collapse
Affiliation(s)
- Cassandra Strohbach
- Jerry L. Pettis Memorial VA Center, Musculoskeletal Disease Center, Loma Linda, CA 92357, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Akkiprik M, Feng Y, Wang H, Chen K, Hu L, Sahin A, Krishnamurthy S, Ozer A, Hao X, Zhang W. Multifunctional roles of insulin-like growth factor binding protein 5 in breast cancer. Breast Cancer Res 2008; 10:212. [PMID: 18710598 PMCID: PMC2575530 DOI: 10.1186/bcr2116] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The insulin-like growth factor axis, which has been shown to protect cells from apoptosis, plays an essential role in normal cell physiology and in cancer development. The family of insulin-like growth factor binding proteins (IGFBPs) has been shown to have a diverse spectrum of functions in cell growth, death, motility, and tissue remodeling. Among the six IGFBP family members, IGFBP-5 has recently been shown to play an important role in the biology of breast cancer, especially in breast cancer metastasis; however, the exact mechanisms of action remain obscure and sometimes paradoxical. An in-depth understanding of IGFBP-5 would shed light on its potential role as a target for breast cancer therapeutics.
Collapse
Affiliation(s)
- Mustafa Akkiprik
- Department of Medical Biology, Marmara University, School of Medicine, 34668 Istanbul, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bobola N, Engist B. IGFBP5 is a potential regulator of craniofacial skeletogenesis. Genesis 2008; 46:52-9. [DOI: 10.1002/dvg.20360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Gorustovich AA, Steimetz T, Nielsen FH, Guglielmotti MB. Histomorphometric Study of Alveolar Bone Healing in Rats Fed a Boron-Deficient Diet. Anat Rec (Hoboken) 2008; 291:441-7. [DOI: 10.1002/ar.20672] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Jurgeit A, Berlato C, Obrist P, Ploner C, Massoner P, Schmölzer J, Haffner MC, Klocker H, Huber LA, Geley S, Doppler W. Insulin-like growth factor-binding protein-5 enters vesicular structures but not the nucleus. Traffic 2007; 8:1815-1828. [PMID: 17892529 DOI: 10.1111/j.1600-0854.2007.00655.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In addition to its extracellular function as a secreted protein, IGF-binding protein (IGFBP)-5 has been postulated to act as a signaling molecule in the nucleus. This study aims to assess the significance of this postulated nuclear localization. By confocal immunofluorescence microscopy, we detected IGFBP-5 in the vesicular compartment of mammary epithelial cells in culture, while no nuclear staining was observed. Immunohistochemistry performed on paraffin sections of the involuting mammary gland revealed IGFBP-5 positive staining of epithelial cells only outside the nucleus. To evaluate the contribution of reuptake of extracellular IGFBP-5, T47D cells were incubated with Alexa Fluor 647-labeled IGFBP-5. The protein was taken up into intracellular vesicles and again was neither detectable in the cytoplasm outside of vesicular structures nor in the nucleus. Quantification of the time and concentration dependence of uptake by immunoblotting revealed that the process was saturable at IGFBP-5 concentrations between 1 and 2 mum and partially reversible with 30% remaining in the cell after a 1-h chase. The observation of nuclear uptake of IGFBP-5 was restricted to artificial conditions such as expression of non-secreted forms of IGFBP-5 or selective permeabilization of the plasma membrane by digitonin.
Collapse
Affiliation(s)
- Andreas Jurgeit
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
- Present address: Institute of Zoology, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Chiara Berlato
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Peter Obrist
- Department of Pathology, St. Vinzenz Hospital Zams, 6511 Zams, Austria
| | - Christian Ploner
- Division of Molecular Pathophysiology, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Petra Massoner
- Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Judith Schmölzer
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Michael C Haffner
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Helmut Klocker
- Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Wolfgang Doppler
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| |
Collapse
|
34
|
Beattie J, Phillips K, Shand JH, Szymanowska M, Flint DJ, Allan GJ. Molecular interactions in the insulin-like growth factor (IGF) axis: a surface plasmon resonance (SPR) based biosensor study. Mol Cell Biochem 2007; 307:221-36. [PMID: 17899320 DOI: 10.1007/s11010-007-9601-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Accepted: 08/24/2007] [Indexed: 12/18/2022]
Abstract
This review describes a comprehensive analysis of a surface plasmon resonance (SPR)-based biosensor study of molecular interactions in the insulin-like growth factor (IGF) molecular axis. In this study, we focus on the interaction between the polypeptide growth factors IGF-I and IGF-II with six soluble IGF binding proteins (IGFBP 1-6), which occur naturally in various biological fluids. We have describe the conditions required for the accurate determination of kinetic rate constants for these interactions and highlight the experimental and theoretical pitfalls, which may be encountered in the early stages of such a study. We focus on IGFBP-5 and describe a site-directed mutagenesis study, which examines the contribution of various residues in the protein to high affinity interaction with IGF-I and -II. We analyse the interaction of IGFBP-5 (and IGFBP-3) with heparin and other biomolecules and describe experiments, which were designed to monitor multi-protein complex formation in this molecular axis.
Collapse
Affiliation(s)
- James Beattie
- Strathclyde Institute of Pharmacy & Biomedical Science, Royal College Building, University of Strathclyde, Glasgow, UK.
| | | | | | | | | | | |
Collapse
|
35
|
van der Weyden L, Adams DJ. The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta Rev Cancer 2007; 1776:58-85. [PMID: 17692468 PMCID: PMC2586335 DOI: 10.1016/j.bbcan.2007.06.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 06/26/2007] [Accepted: 06/26/2007] [Indexed: 12/13/2022]
Abstract
Ras proteins play a direct causal role in human cancer with activating mutations in Ras occurring in approximately 30% of tumours. Ras effectors also contribute to cancer, as mutations occur in Ras effectors, notably B-Raf and PI3-K, and drugs blocking elements of these pathways are in clinical development. In 2000, a new Ras effector was identified, RAS-association domain family 1 (RASSF1), and expression of the RASSF1A isoform of this gene is silenced in tumours by methylation of its promoter. Since methylation is reversible and demethylating agents are currently being used in clinical trials, detection of RASSF1A silencing by promoter hypermethylation has potential clinical uses in cancer diagnosis, prognosis and treatment. RASSF1A belongs to a new family of RAS effectors, of which there are currently 8 members (RASSF1-8). RASSF1-6 each contain a variable N-terminal segment followed by a Ras-association (RA) domain of the Ral-GDS/AF6 type, and a specialised coiled-coil structure known as a SARAH domain extending to the C-terminus. RASSF7-8 contain an N-terminal RA domain and a variable C-terminus. Members of the RASSF family are thought to function as tumour suppressors by regulating the cell cycle and apoptosis. This review will summarise our current knowledge of each member of the RASSF family and in particular what role they play in tumourigenesis, with a special focus on RASSF1A, whose promoter methylation is one of the most frequent alterations found in human tumours.
Collapse
Affiliation(s)
- Louise van der Weyden
- Experimental Cancer Genetics Laboratory, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton Cambridge, UK.
| | | |
Collapse
|
36
|
Xing W, Singgih A, Kapoor A, Alarcon CM, Baylink DJ, Mohan S. Nuclear factor-E2-related factor-1 mediates ascorbic acid induction of osterix expression via interaction with antioxidant-responsive element in bone cells. J Biol Chem 2007; 282:22052-61. [PMID: 17510056 DOI: 10.1074/jbc.m702614200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently found that deletion of the gulonolactone oxidase gene, which is involved in the synthesis of ascorbic acid (AA), was responsible for the fracture phenotype in spontaneous fracture mice. To explore the molecular mechanisms by which AA regulates osteoblast differentiation, we examined the effect of AA on osterix expression via Nrf1 (NF-E2-related factor-1) binding to antioxidant-responsive element (ARE) in bone marrow stromal (BMS) cells. AA treatment caused a 6-fold increase in osterix expression in mutant BMS cells at 24 h, which was unaffected by pretreatment with protein synthesis inhibitor. Sequence analyses of mouse osterix promoter revealed a putative ARE located at -1762 to -1733 upstream of the transcription start site to which Nrf potentially binds. A gel mobility shift assay revealed that nuclear proteins from AA-treated BMS cells bound to radiolabeled ARE much more strongly than nuclear extracts from AA-untreated cells. A chromatin immunoprecipitation assay with Nrf1 antibody confirmed the interaction of Nrf1 with the mouse osterix promoter. A reporter assay demonstrated that the promoter activity of mouse osterix containing an ARE was stimulated 4-fold by a 48-h treatment with AA in spontaneous fracture BMS cells. Treatment of mutant BMS cells with AA resulted in a 3.9-fold increase in the nuclear accumulation of Nrf1. Transfection of mutant BMS cells with Nrf1 small interfering RNA decreased Nrf1 protein by 4.5-fold, blocked AA induction of osterix expression, and impaired BMS cell differentiation. Our data provided the first experimental evidence that AA modulated osterix expression via a novel mechanism involving Nrf1 nuclear translocation and Nrf1 binding to ARE to activate genes critical for cell differentiation.
Collapse
Affiliation(s)
- Weirong Xing
- Musculoskeletal Disease Center, Jerry L Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA 92357, USA
| | | | | | | | | | | |
Collapse
|
37
|
Graham ME, Kilby DM, Firth SM, Robinson PJ, Baxter RC. The in vivo phosphorylation and glycosylation of human insulin-like growth factor-binding protein-5. Mol Cell Proteomics 2007; 6:1392-405. [PMID: 17496250 DOI: 10.1074/mcp.m700027-mcp200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mass spectrometry is often used to determine post-translational modifications by analysis of tryptic digests of proteins. Here we demonstrate that the analysis of tryptic peptides together with analysis of the full-length protein provided optimal characterization of insulin-like growth factor-binding protein-5 (IGFBP-5) phosphorylation and glycosylation. IGFBP-5 binds insulin-like growth factors with high affinity and has important roles in cell survival, differentiation, and apoptosis. Until now, the primary structure of IGFBP-5 has been incompletely defined. We analyzed human IGFBP-5 from T47D cells by mass spectrometry to determine all of the in vivo post-translational modifications. In full-length IGFBP-5, 31% of the protein was unmodified, 37% was monophosphorylated, and 4% was diphosphorylated with no other modification. The remaining 27% was glycosylated, more than half of which was also monophosphorylated. The major phosphorylation site was Ser(96) in the central domain, and a minor phosphorylation site was Ser(248) near the C terminus. Neither site was phosphorylated in vitro by casein kinase 2, ruling it out as the in vivo kinase. An in vivo phosphorylation site was also found in IGFBP-2 at an analogous position, Ser(106). IGFBP-5 was heterogeneously O-glycosylated mainly by sialylated core 1 type glycans. The most abundant structure contained N-acetylhexosamine, hexose, and two N-acetylneuraminic acid carbohydrates. A small amount of sialylated core 2 type glycan was also present. Phosphorylation and O-glycosylation both affected IGFBP-5 binding to heparin but not insulin-like growth factor binding or ternary complex formation with the acid-labile subunit. The results reveal the first description of the in vivo phosphorylation of IGFBP-5 and its glycan composition.
Collapse
Affiliation(s)
- Mark E Graham
- Cell Signalling Unit, Children's Medical Research Institute, Locked Bag 23, Wentworthville, New South Wales 2145, Australia
| | | | | | | | | |
Collapse
|
38
|
Estrabaud E, Lassot I, Blot G, Le Rouzic E, Tanchou V, Quemeneur E, Daviet L, Margottin-Goguet F, Benarous R. RASSF1C, an Isoform of the Tumor Suppressor RASSF1A, Promotes the Accumulation of β-Catenin by Interacting with βTrCP. Cancer Res 2007; 67:1054-61. [PMID: 17283138 DOI: 10.1158/0008-5472.can-06-2530] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Ras-association domain family 1 (RASSF1) gene has seven different isoforms; isoform A is a tumor-suppressor gene (RASSF1A). The promoter of RASSF1A is inactivated in many cancers, whereas the expression of another major isoform, RASSF1C, is not affected. Here, we show that RASSF1C, but not RASSF1A, interacts with betaTrCP. Binding of RASSF1C to betaTrCP involves serine 18 and serine 19 of the SS(18)GYXS(19) motif present in RASSF1C but not in RASSF1A. This motif is reminiscent of the canonical phosphorylation motif recognized by betaTrCP; however, surprisingly, the association between RASSF1C and betaTrCP does not occur via the betaTrCP substrate binding domain, the WD40 repeats. Overexpression of RASSF1C, but not of RASSF1A, resulted in accumulation and transcriptional activation of the beta-catenin oncogene, due to inhibition of its betaTrCP-mediated degradation. Silencing of RASSF1A by small interfering RNA was sufficient for beta-catenin to accumulate, whereas silencing of both RASSF1A and RASSF1C had no effect. Thus, RASSF1A and RASSF1C have opposite effects on beta-catenin degradation. Our results suggest that RASSF1C expression in the absence of RASSF1A could play a role in tumorigenesis.
Collapse
Affiliation(s)
- Emilie Estrabaud
- Institut Cochin, Département Maladies Infectieuses; Institut National de la Sante et de la Recherche Medicale U567, 27 rue du Faubourg Saint-Jacques, Bâtiment G Roussy, 75014 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Amaar YG, Minera MG, Hatran LK, Strong DD, Mohan S, Reeves ME. Ras association domain family 1C protein stimulates human lung cancer cell proliferation. Am J Physiol Lung Cell Mol Physiol 2006; 291:L1185-90. [PMID: 16891396 DOI: 10.1152/ajplung.00072.2006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recently, the Ras association domain family 1 gene (RASSF1) has been identified as a Ras effector encoding two major mRNA forms, RASSF1A and RASSF1C, derived by alternative promoter selection and alternative mRNA splicing. RASSF1A is a tumor suppressor gene. However, the function of RASSF1C, both in normal and cancer cells, is still unknown. To learn more about the function of RASSF1C in human cancer cells, we tested the effect of silencing RASSF1C mRNA with small interfering RNA on lung cancer cells (NCI H1299) that express RASSF1C but not RASSF1A. Small interfering RNA specific for RASSF1C reduced RASSF1C mRNA levels compared with controls. This reduction in RASSF1C expression caused a significant decrease in lung cancer cell proliferation. Furthermore, overexpression of RASSF1C increased cell proliferation in lung cancer cells. Finally, we found that RASSF1C, unlike RASSF1A, does not upregulate N-cadherin 2 and transglutaminase 2 protein expression in NCI H1299 lung cancer cells. This suggests that RASSF1C and RASSF1A have different effector targets. Together, our findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor but rather stimulates lung cancer cell proliferation.
Collapse
Affiliation(s)
- Yousef G Amaar
- Surgical Oncology Laboratory, Loma Linda University, Loma Linda, California, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Gorustovich AA, López JMP, Guglielmotti MB, Cabrini RL. Biological performance of boron-modified bioactive glass particles implanted in rat tibia bone marrow. Biomed Mater 2006; 1:100-5. [DOI: 10.1088/1748-6041/1/3/002] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
41
|
Beattie J, Allan GJ, Lochrie JD, Flint DJ. Insulin-like growth factor-binding protein-5 (IGFBP-5): a critical member of the IGF axis. Biochem J 2006; 395:1-19. [PMID: 16526944 PMCID: PMC1409685 DOI: 10.1042/bj20060086] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/30/2006] [Indexed: 11/17/2022]
Abstract
The six members of the insulin-like growth factor-binding protein family (IGFBP-1-6) are important components of the IGF (insulin-like growth factor) axis. In this capacity, they serve to regulate the activity of both IGF-I and -II polypeptide growth factors. The IGFBPs are able to enhance or inhibit the activity of IGFs in a cell- and tissue-specific manner. One of these proteins, IGFBP-5, also has an important role in controlling cell survival, differentiation and apoptosis. In this review, we report on the structural and functional features of the protein which are important for these effects. We also examine the regulation of IGFBP-5 expression and comment on its potential role in tumour biology, with special reference to work with breast cancer cells.
Collapse
Key Words
- extracellular matrix (ecm)
- glycosaminoglycan
- insulin-like growth factor-i (igf-i)
- insulin-like growth factor-binding protein 5 (igfbp-5)
- mammary gland
- proteolysis
- adam, adisintegrin and metalloprotease
- ap-2, activator protein 2
- cat, chloramphenicol acetyltransferase
- cbp-4, c-terminus of insulin-like growth factor-binding protein 4 (residues 151–232)
- c/ebp, ccaat/enhancer-binding protein
- ecm, extracellular matrix
- er, oestrogen receptor
- erk1/2, extracellular-signal-regulated protein kinase 1/2
- fhl-2, four-and-a-half lim domain 2
- gag, glycosaminoglycan
- gh, growth hormone
- igf, insulin-like growth factor
- igfbp, igf-binding protein
- igf-ir, igf-i receptor
- igf-iir, igf-ii receptor
- ir, insulin receptor
- irs, ir substrate
- mapk, mitogen-activated protein kinase
- nbp-4, n-terminus of igfbp-4 (residues 3–82)
- oe2, oestradiol
- op-1, osteogenic protein-1
- opn, osteopontin
- pai-1, plasminogen activator inhibitor-1
- papp, pregnancy-associated plasma protease
- pge2, prostaglandin e2
- psmc, porcine smooth-muscle cell
- ra, retinoic acid
- rassf1c, isoform c of the ras association family 1 protein group
- rt, reverse transcription
- spr, surface plasmon resonance
- tpa, tissue plasminogen activator
- tsp-1, thrombospondin-1
- vn, vitronectin
Collapse
Affiliation(s)
- James Beattie
- Hannah Research Institute, Ayr KA6 5HL, Scotland, UK.
| | | | | | | |
Collapse
|
42
|
Amaar YG, Tapia B, Chen ST, Baylink DJ, Mohan S. Identification and characterization of novel IGFBP5 interacting protein: evidence IGFBP5-IP is a potential regulator of osteoblast cell proliferation. Am J Physiol Cell Physiol 2005; 290:C900-6. [PMID: 16269403 PMCID: PMC2902994 DOI: 10.1152/ajpcell.00563.2004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin-like growth factor binding protein-5 (IGFBP5) is a multifunctional protein, which acts not only as a traditional binding protein, but also functions as a growth factor independent of IGFs to stimulate bone formation. It has been predicted that the intrinsic growth factor action of IGFBP5 involves binding of IGFBP5 to a putative receptor to induce downstream signaling pathways and/or nuclear translocation of IGFBP5 to influence transcription of genes involved in osteoblast cell proliferation/differentiation. Our study indentified proteins that bound to IGFBP5 using IGFBP5 as bait in a yeast two-hybrid screen of the U2 human osteosarcoma cell cDNA library. One of the clones that interacted strongly with the bait under high-stringency conditions corresponded to a novel IGFBP5 interacting protein (IGFBP5-IP) encoded by a gene that resides in mouse chromosome 10. The interaction between IGFBP5-IP and IGFBP5 is confirmed by in vitro coimmunoprecipitation studies that used pFlag and IGFBP5 polyclonal antibody, and cell lysates overexpressing both IGFBP5-IP and IGFBP5. Northern blot and RT-PCR analysis showed that the IGFBP-IP is expressed in both untransformed normal human osteoblasts and in osteosarcoma cell lines, which are known to produce IGFBP5. To determine the roles of IGFBP5-IP, we evaluated the effect of blocking the expression of IGFBP5-IP on osteoblast proliferation. We found that using a IGFBP5-IP-specific small interfering-hairpin plasmid resulted in a decrease in both basal and IGFBP5-induced osteoblast cell proliferation. On the basis of these findings, we predict that IGFBP5-IP may act as intracellular mediator of growth promoting actions of IGFBP5 and perhaps other osteoregulatory agents in bone cells.
Collapse
Affiliation(s)
- Yousef G Amaar
- Musculoskeletal Disease Center, PO Box 151, Jerry L. Pettis Veterans Affairs Medical Center, 11201 Benton St., Loma Linda, CA 92357, USA
| | | | | | | | | |
Collapse
|