1
|
Wang Y, Zhang H, Zhang X, Mu P, Zhao L, Qi R, Zhang Y, Zhu X, Dong Y. The role of IGFBP-3 in tumor development and progression: enlightenment for diagnosis and treatment. Med Oncol 2024; 41:141. [PMID: 38714554 DOI: 10.1007/s12032-024-02373-x] [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: 02/08/2024] [Accepted: 03/25/2024] [Indexed: 05/10/2024]
Abstract
IGFBP-3 is aberrantly expressed in many tumor types, and its serum and tumor tissue levels provide auxiliary information for assessing the degree of tumor malignancy and patient prognosis, making it a potential therapeutic target for human malignancies and conferring it remarkable clinical value for determining patient prognosis. In this review, we provide a comprehensive overview of the aberrant expression, diverse biological effects, and clinical implications of IGFBP-3 in tumors and its role as a potential prognostic marker and therapeutic target for tumors. In addition, we summarize the signaling pathways through which IGFBP-3 exerts its effects. IGFBP-3 comprises an N-terminal, an intermediate region, and a C-terminal structural domain, each exerting different biological effects in several tumor cell types in an IGF-dependent/non-independent manner. IGFBP-3 shares an intricate relationship with the tumor microenvironment, thereby affecting tumor growth. Overall, IGFBP-3 is an essential regulatory factor that mediates tumor occurrence and progression. Gaining deeper insights into the fundamental characteristics of IGFBP-3 and its role in various tumor types will provide new perspectives and allow for the development of novel strategies for cancer diagnosis, treatment, and prognostic evaluation.
Collapse
Affiliation(s)
- Yudi Wang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - He Zhang
- Department of Immunology, Qiqihar Medical University, Qiqihar, China
| | - Xuehua Zhang
- Department of Precision Biomedical Laboratory, Liaocheng People's Hospital, Liaocheng, China
| | - Peizheng Mu
- School of Computer and Control Engineering, Yantai University, Yantai, China
| | - Leilei Zhao
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Ruomei Qi
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Yurui Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, China.
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, China.
| |
Collapse
|
2
|
Pan H, Huan C, Hou Y, Yan P, Yang F, Jiang L, Gao S. Porcine IGFBP3 promotes porcine circovirus type 2 replication via PERK/eIF2α mediated DNA damage. Vet Microbiol 2023; 287:109897. [PMID: 37922860 DOI: 10.1016/j.vetmic.2023.109897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
The infection of porcine circovirus type 2 (PCV2) triggers activation of the protein kinase RNA-like endoplasmic reticulum kinase (PERK) pathway and leads to DNA damage. Insulin-like growth factor-binding protein 3 (IGFBP3) may interact with the endoplasmic reticulum (ER). It remains unclear whether IGFBP3 regulates DNA damage via ER stress to mediate PCV2 replication. In this study, we observed an upregulation of porcine IGFBP3 expression during PCV2 infection, and overexpression of IGFBP3 enhanced the expression of PCV2 Cap protein, PCV2 DNA copy number, and viral titers in PK-15 B6 cells and 3D4/21 cells. Additionally, overexpression of IGFBP3 induced an increase in the DNA damage marker γH2AX by activating the PERK/eIF2α pathway without concomitant activation of ATF4, IRE1α, and ATF6α/GRP78 pathways in PK-15 B6 cells and 3D4/21 cells. Knockdown of IGFBP3 had a reverse effect on PCV2 replication in PK-15 B6 cells and 3D4/21 cells. Furthermore, treatment with etoposide enhanced PCV2 replication while KU57788 decreased it. GSK2606414 and salubrinal limited both DNA damage and viral replication. Therefore, our findings suggest that porcine IGFBP3 promotes PCV2 replication through the PERK/eIF2α pathway-mediated induction of DNA damage in PK-15 B6 cells and 3D4/21 cells. Our study provides a basis for exploring novel antiviral strategies via the extensive understanding of the relationships between host cellular proteins and viral replication.
Collapse
Affiliation(s)
- Haochun Pan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China
| | - Changchao Huan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China
| | - Yutong Hou
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China
| | - Ping Yan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China
| | - Fan Yang
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China
| | - Luyao Jiang
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China
| | - Song Gao
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, China.
| |
Collapse
|
3
|
Wei L, Liping Z, Suya K. Expression of insulin-like growth factor binding protein-3 in HELLP syndrome. BMC Pregnancy Childbirth 2023; 23:778. [PMID: 37950229 PMCID: PMC10637003 DOI: 10.1186/s12884-023-06074-7] [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: 05/18/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE To investigate the expression of insulin-like growth factor binding protein-3(IGFBP-3) in HELLP syndrome and its possible role in the pathogenesis of this disease. METHODS 1) 87 subjects were enrolled, including 29 patients with HELLP syndrome, 29 patients with pre-eclampsia (PE), and 29 healthy gravidae as control. The levels of IGFBP-3, IGF-1, TGF-β1, and VEGF in maternal and umbilical blood of them were detected using ELISA. Correlation analysis was used to observe the correlation between IGFBP-3 and IGF-1/TGF-β1/VEGF in maternal and umbilical blood, as well as that between maternal serum IGFBP-3 and clinical diagnostic indicators of HELLP syndrome. 2) Human hepatic sinusoid endothelial cells (HLSEC) and human umbilical vein endothelial cells (HUVEC) were cultured with different concentrations of IGFBP-3. After 72 h of culture, cell apoptosis and the normal living cells rate were detected and compared. RESULTS 1) In both maternal and umbilical blood of HELLP group, levels of IGFBP-3 and TGF-β1 were higher than control and PE group, IGF-1was lower than control group, VEGF was lower than control and PE group. IGFBP-3 in maternal blood was correlated with IGF-1/TGF-β1/ VEGF, while IGFBP-3 in umbilical blood was linked to IGF-1/TGF-β1. In maternal blood, there was a negative correlation between PLT and IGFBP-3, and a positive correlation between ALT/AST/LDH and IGFBP-3. 2) After cultured with IGFBP-3, the total apoptosis rate of either HLSEC or HUVEC was considerably elevated, while the normal living rate was decreased. CONCLUSION The expression of IGFBP-3 is elevated in HELLP syndrome, which may subsequently promote cell apoptosis by affecting the expression and function of IGF-1, VEGF, and TGFβ1 in the IGF/PI3K/Akt, TGF-β1/Smad3, and VEGF/eNOS/NO pathways. IGFBP-3 aggravates inflammatory reactions of the vascular endothelium and liver under hypoxia, affects the normal function of cells, and plays a role in the pathogenesis of diseases.
Collapse
Affiliation(s)
- Li Wei
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, No. 26, Daoqian Street, Suzhou, Jiangsu, China
| | - Zhou Liping
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, No. 26, Daoqian Street, Suzhou, Jiangsu, China
| | - Kang Suya
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, No. 26, Daoqian Street, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Baxter RC. Signaling Pathways of the Insulin-like Growth Factor Binding Proteins. Endocr Rev 2023; 44:753-778. [PMID: 36974712 PMCID: PMC10502586 DOI: 10.1210/endrev/bnad008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/25/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
The 6 high-affinity insulin-like growth factor binding proteins (IGFBPs) are multifunctional proteins that modulate cell signaling through multiple pathways. Their canonical function at the cellular level is to impede access of insulin-like growth factor (IGF)-1 and IGF-2 to their principal receptor IGF1R, but IGFBPs can also inhibit, or sometimes enhance, IGF1R signaling either through their own post-translational modifications, such as phosphorylation or limited proteolysis, or by their interactions with other regulatory proteins. Beyond the regulation of IGF1R activity, IGFBPs have been shown to modulate cell survival, migration, metabolism, and other functions through mechanisms that do not appear to involve the IGF-IGF1R system. This is achieved by interacting directly or functionally with integrins, transforming growth factor β family receptors, and other cell-surface proteins as well as intracellular ligands that are intermediates in a wide range of pathways. Within the nucleus, IGFBPs can regulate the diverse range of functions of class II nuclear hormone receptors and have roles in both cell senescence and DNA damage repair by the nonhomologous end-joining pathway, thus potentially modifying the efficacy of certain cancer therapeutics. They also modulate some immune functions and may have a role in autoimmune conditions such as rheumatoid arthritis. IGFBPs have been proposed as attractive therapeutic targets, but their ubiquity in the circulation and at the cellular level raises many challenges. By understanding the diversity of regulatory pathways with which IGFBPs interact, there may still be therapeutic opportunities based on modulation of IGFBP-dependent signaling.
Collapse
Affiliation(s)
- Robert C Baxter
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital,St Leonards, NSW 2065, Australia
| |
Collapse
|
5
|
Zhang X, Wang G, Gong Y, Zhao L, Song P, Zhang H, Zhang Y, Ju H, Wang X, Wang B, Ren H, Zhu X, Dong Y. IGFBP3 induced by the TGF-β/EGFRvIII transactivation contributes to the malignant phenotype of glioblastoma. iScience 2023; 26:106639. [PMID: 37192967 PMCID: PMC10182331 DOI: 10.1016/j.isci.2023.106639] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/09/2023] [Accepted: 04/05/2023] [Indexed: 05/18/2023] Open
Abstract
Dual or multi-targets therapy targeting epidermal growth factor receptor variant III (EGFRvIII) and other molecular may relax the constraint for glioblastoma (GBM), putting forward the urgent requirement of finding candidate molecules. Here, the insulin-like growth factor binding protein-3 (IGFBP3) was considered a candidate, whereas the mechanisms of IGFBP3 production remain unclear. We treated GBM cells with exogenous transforming growth factor β (TGF-β) to simulate the microenvironment. We found that TGF-β and EGFRvIII transactivation induced the activation of transcription factor c-Jun, which specifically bound to the promoter region of IGFBP3 through Smad2/3 and ERK1/2 pathways and promoted the production and secretion of IGFBP3. IGFBP3 knockdown inhibited the activation of TGF-β and EGFRvIII signals and the malignant behaviors triggered by them in vitro and in vivo. Collectively, our results indicated a positive feedback loop of p-EGFRvIII/IGFBP3 under administration of TGF-β, blocking IGFBP3 may be an additional target in EGFRvIII-expressing GBM-selective therapeutic strategy.
Collapse
Affiliation(s)
- Xuehua Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Guoyan Wang
- Clinical Laboratory of Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264199, China
| | - Yujiao Gong
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Leilei Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Ping Song
- Department of Ophthalmology, Jiarun Hospital of Harbin, Harbin, Heilongjiang 150000, China
| | - He Zhang
- Department of Immunology, Qiqihar Medical University, Qiqihar, Heilongjiang 161000, China
| | - Yurui Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Huanyu Ju
- Department of Immunology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xiaoyu Wang
- Department of Neurology, Hongda Hospital, Jinxiang, Shandong 272200, China
| | - Bin Wang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Huan Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518000, China
- Corresponding author
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong 264005, China
- Corresponding author
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, Shandong 264003, China
- Corresponding author
| |
Collapse
|
6
|
Bersin TV, Mapes HM, Journey ML, Beckman BR, Lema SC. Insulin-like growth factor-1 (Igf1) signaling responses to food consumption after fasting in the Pacific rockfish Sebastes carnatus. Comp Biochem Physiol A Mol Integr Physiol 2023; 282:111444. [PMID: 37201654 DOI: 10.1016/j.cbpa.2023.111444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 05/20/2023]
Abstract
Fish adjust rates of somatic growth in the face of changing food consumption. As in other vertebrates, growth in fish is regulated by the growth hormone (Gh)/insulin-like growth factor-1 (Igf1) endocrine axis, and changes in food intake impact growth via alterations to Gh/Igf1 signaling. Understanding the time course by which the Gh/Igf1 axis responds to food consumption is crucial to predict how rapidly changes in food abundance might lead to altered growth dynamics. Here, we looked at the response times of plasma Igf1 and liver Igf1 signaling-associated gene expression to refeeding after food deprivation in juvenile gopher rockfish (Sebastes carnatus), one of several species of northern Pacific Ocean Sebastes rockfishes targeted by fisheries or utilized for aquaculture. Gopher rockfish were fasted for 30 d, after which a subset was fed to satiation for 2 h, while other rockfish continued to be fasted. Refed fish exhibited higher hepatosomatic index (HSI) values and increased Igf1 after food consumption. Gene transcripts for Gh receptor 1 (ghr1), but not ghr2, increased in the liver after eating. Transcripts encoding igf1 also increased in the liver of refed fish 2-4 d after feeding, only to return to levels similar as continually fasted rockfish by 9 d after feeding. Liver mRNA abundances for Igf binding protein (Igfbp) genes igfbp1a, igfbp1b, and igfbp3a declined within 2 d of feeding. These findings provide evidence that circulating Igf1 in rockfish reflects a fish's feeding experience within the previous few days, and suggest that feeding-induced increases in Igf1 are being mediated in part by altered liver sensitivity to Gh due to upregulated Gh receptor 1 expression.
Collapse
Affiliation(s)
- Theresa V Bersin
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Hayley M Mapes
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Meredith L Journey
- Lynker Technology, 202 Church St SE #536, Leesburg, VA 20175, USA; Under Contract to Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA 98112, USA
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA 98112, USA
| | - Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA.
| |
Collapse
|
7
|
Wang L, Du A, Lu Y, Zhao Y, Qiu M, Su Z, Shu H, Shen H, Sun W, Kong X. Peptidase Inhibitor 16 Attenuates Left Ventricular Injury and Remodeling After Myocardial Infarction by Inhibiting the HDAC1-Wnt3a-β-Catenin Signaling Axis. J Am Heart Assoc 2023; 12:e028866. [PMID: 37158154 DOI: 10.1161/jaha.122.028866] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Background Myocardial infarction (MI) is a cardiovascular disease with high morbidity and mortality. PI16 (peptidase inhibitor 16), as a secreted protein, is highly expressed in heart diseases such as heart failure. However, the functional role of PI16 in MI is unknown. This study aimed to investigate the role of PI16 after MI and its underlying mechanisms. Methods and Results PI16 levels after MI were measured by enzyme-linked immunosorbent assay and immunofluorescence staining, which showed that PI16 was upregulated in the plasma of patients with acute MI and in the infarct zone of murine hearts. PI16 gain- and loss-of-function experiments were used to investigate the potential role of PI16 after MI. In vitro, PI16 overexpression inhibited oxygen-glucose deprivation-induced apoptosis in neonatal rat cardiomyocytes, whereas knockdown of PI16 exacerbated neonatal rat cardiomyocyte apoptosis. In vivo, left anterior descending coronary artery ligation was performed on PI16 transgenic mice, PI16 knockout mice, and their littermates. PI16 transgenic mice showed decreased cardiomyocyte apoptosis at 24 hours after MI and improved left ventricular remodeling at 28 days after MI. Conversely, PI16 knockout mice showed aggravated infract size and remodeling. Mechanistically, PI16 downregulated Wnt3a (wingless-type MMTV integration site family, member 3a)/β-catenin pathways, and the antiapoptotic role of PI16 was reversed by recombinant Wnt3a in oxygen-glucose deprivation-induced neonatal rat cardiomyocytes. PI16 also inhibited HDAC1 (class I histone deacetylase) expression, and overexpression HDAC1 abolished the inhibition of apoptosis and Wnt signaling of PI16. Conclusions In summary, PI16 protects against cardiomyocyte apoptosis and left ventricular remodeling after MI through the HDAC1-Wnt3a-β-catenin axis.
Collapse
Affiliation(s)
- Luyang Wang
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Anning Du
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Yan Lu
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Yunxi Zhao
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Ming Qiu
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
- School of Medicine Southeast University Nanjing Jiangsu China
| | - Zhenyang Su
- School of Medicine Southeast University Nanjing Jiangsu China
| | - Huanyu Shu
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Hui Shen
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Wei Sun
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
| | - Xiangqing Kong
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University Nanjing Jiangsu China
- Cardiovascular Research Center The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University Suzhou China
| |
Collapse
|
8
|
Insulin‑like growth factor axis: A potential nanotherapy target for resistant cervical cancer tumors (Review). Oncol Lett 2023; 25:128. [PMID: 36844628 PMCID: PMC9950333 DOI: 10.3892/ol.2023.13714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 09/07/2022] [Indexed: 02/12/2023] Open
Abstract
Cervical cancer is among the most frequently occurring neoplasms worldwide, and it particularly affects individuals in developing countries. Factors such as the low quality of screening tests, the high incidence of locally advanced cancer stages and the intrinsic resistance of certain tumors are the main causes of failure in the treatment of this neoplasm. Due to advances in the understanding of carcinogenic mechanisms and bioengineering research, advanced biological nanomaterials have been manufactured. The insulin-like growth factor (IGF) system comprises multiple growth factor receptors, including IGF receptor 1. These receptors are activated by binding to their respective growth factor ligands, IGF-1 and IGF-2, and insulin, and play an important role in the development, maintenance, progression, survival and treatment resistance of cervical cancer. In the present review, the role of the IGF system in cervical cancer and three nanotechnological applications that use elements of this system are described, namely Trap decoys, magnetic iron oxide nanoparticles and protein nanotubes. Their use in the treatment of resistant cervical cancer tumors is also discussed.
Collapse
|
9
|
Dya GA, Klychnikov OI, Adasheva DA, Vladychenskaya EA, Katrukha AG, Serebryanaya DV. IGF-Binding Proteins and Their Proteolysis as a Mechanism of Regulated IGF Release in the Nervous Tissue. BIOCHEMISTRY (MOSCOW) 2023; 88:S105-S122. [PMID: 37069117 DOI: 10.1134/s0006297923140079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Insulin-like growth factors 1 and 2 (IGF-1 and IGF-2) play a key role in the maintenance of the nervous tissue viability. IGF-1 and IGF-2 exhibit the neuroprotective effects by stimulating migration and proliferation of nervous cells, activating cellular metabolism, inducing regeneration of damaged cells, and regulating various stages of prenatal and postnatal development of the nervous system. The availability of IGFs for the cells is controlled via their interaction with the IGF-binding proteins (IGFBPs) that inhibit their activity. On the contrary, the cleavage of IGFBPs by specific proteases leads to the IGF release and activation of its cellular effects. The viability of neurons in the nervous tissue is controlled by a complex system of trophic factors secreted by auxiliary glial cells. The main source of IGF for the neurons are astrocytes. IGFs can accumulate as an extracellular free ligand near the neuronal membranes as a result of proteolytic degradation of IGFBPs by proteases secreted by astrocytes. This mechanism promotes interaction of IGFs with their genuine receptors and triggers intracellular signaling cascades. Therefore, the release of IGF by proteolytic cleavage of IGFBPs is an important mechanism of neuronal protection. This review summarizes the published data on the role of IGFs and IGFBPs as the key players in the neuroprotective regulation with a special focus on the specific proteolysis of IGFBPs as a mechanism for the regulation of IGF bioavailability and viability of neurons.
Collapse
Affiliation(s)
- German A Dya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Oleg I Klychnikov
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elizaveta A Vladychenskaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexey G Katrukha
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
10
|
Green CJ, Span M, Rayhanna MH, Perera M, Day ML. Insulin-like Growth Factor Binding Protein 3 Increases Mouse Preimplantation Embryo Cleavage Rate by Activation of IGF1R and EGFR Independent of IGF1 Signalling. Cells 2022; 11:cells11233762. [PMID: 36497022 PMCID: PMC9736160 DOI: 10.3390/cells11233762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
The viability of embryos cultured in vitro is poor compared to those that develop in vivo. The lack of maternally derived growth factors in vitro may contribute to this problem. Insulin-like growth factor binding protein 3 (IGFBP3) is one such growth factor that has been identified in the maternal reproductive system. This study examined the role of autocrine and exogenous IGFBP3 in mouse preimplantation embryos. Embryos expressed IGFBP3 across all stages of preimplantation development, and addition of exogenous IGFBP3 to embryo culture media increased the rate of development to the 2-, 4-, 5-, and 8-cell stages. Addition of inhibitors of the IGF1 and EGF receptors prevented this IGFBP3-mediated improvement in developmental rate, but the effect was not cumulative, indicating that both receptors are transactivated downstream of IGFBP3 as part of the same signalling pathway. Acute exposure to IGFBP3 increased phosphorylation of Akt and rps6 in 4-8 cell embryos, suggesting activation of the PI3-kinase/Akt pathway downstream of the IGF1 and EGFR receptors to promote cell proliferation and survival. In conclusion, addition of IGFBP3 to embryo culture media increases early cleavage rates independent of IGF1 signalling and therefore, IGFBP3 addition to IVF culture media should be considered.
Collapse
|
11
|
Zhu M, Han H, Hu L, Cao Y, Fan Z. Insulin-binding protein-5 down-regulates the balance of Th17/Treg. Front Immunol 2022; 13:1019248. [PMID: 36389828 PMCID: PMC9664073 DOI: 10.3389/fimmu.2022.1019248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
The inflammatory response plays critical important role in tissue hemostasis. Our previous study showed insulin-binding protein-5 (IGFBP5) could enhance the regeneration of tissue defect under inflammation condition, but the function of IGFBP5 in controlling inflammation and regulating immune responses remains unclear. In present study, we studied the regulatory effect of IGFBP5 on T cell immune response in vitro, and the maintenance of Th17/Treg balance in vivo by using dextran sulfate sodium salt (DSS)-induced colitis in mice. The results showed that IGFBP5 inhibited the differentiation of CD4+ T cells into Th17 subset while promoted its differentiation into Treg subsets. Further results of animal experiments demonstrated that recombinant IGFBP5 reversed the imbalance of Th17/Treg and alleviated the severity of DSS-induced colitis. The percentage of Th17 cells decreased and the percentage of Treg cells increased in the inflamed colon tissue and mesenteric lymph nodes of mice with colitis after IGFBP5 treatment. Besides, pro-inflammatory cytokines such as TNF-α, IL-1β and IFN-γ in serum were suppressed after the treatment of IGFBP5. Moreover, the function of IGFBP5 in regulating Th17/Treg balance could be inhibited by the inhibitors of ERK or JNK pathway. In conclusion, all these data showed that IGFBP5 could regulate Th17/Treg balance via ERK or JNK pathways. The findings of our study provide a theoretical basis for the application of IGFBP5 in inflammatory diseases.
Collapse
Affiliation(s)
- Mengyuan Zhu
- 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
| | - Han Han
- Department of General Dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Lei Hu
- 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,*Correspondence: Lei Hu, ; Yu Cao, ; Zhipeng Fan,
| | - Yu Cao
- Department of General Dentistry, Capital Medical University School of Stomatology, Beijing, China,*Correspondence: Lei Hu, ; Yu Cao, ; Zhipeng Fan,
| | - 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,Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China,*Correspondence: Lei Hu, ; Yu Cao, ; Zhipeng Fan,
| |
Collapse
|
12
|
Mascarenhas DD. Immodulin peptides influence musculoskeletal homeostasis by linking extracellular cues to macrophage and myoblast nuclear receptors. Eur J Transl Myol 2022; 32:10695. [PMID: 36121116 PMCID: PMC9830397 DOI: 10.4081/ejtm.2022.10695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/08/2022] [Indexed: 01/13/2023] Open
Abstract
Immodulins are synthetic peptides derived from the C-terminal domains of insulin-like growth factor binding proteins (IGFBPs). Immodulins from the 3/5/6 (but not 1/2/4) IGFBP evolutionary clade transduce extracellular matrix (ECM) signals to RXR, NR4A1 and PPAR-alpha nuclear receptors (NRs) to stimulate novel macrophage lineages. The rationale of this study was to reconcile physical associations of immodulins with ECM and NRs, effects of siRNAs and chemical inhibitors in vivo, and immodulin-driven pro-differentiation effects in cell culture. When added to THP1D cells, immodulins stimulate CD169+ Clec9a+ and Clec12a+ macrophage lineages via a EP300/RXRγ/Nur77 transcriptional mechanism. This phenomenon is accompanied by the secretion of CCL22, IL-10 and TGFbeta and the ability to stimulate FoxP3+ T-cells in co-culture. ECM ligands of 3/5/6 immodulins include iron, zinc, glycosaminoglycans, transferrin and phosphatidylinositol-4,5,-biphosphate (PIP2), which can influence their pro-differentiation effects. Remarkably, immodulins also stimulate myogenesis in C2C12 myoblasts, thereby revealing a novel link between immune and musculoskeletal homeostasis. Distinct NR agonists stimulate these companion differentiation processes. Using solution NMR to guide design, immodulins with a tripeptide extension near the iron-binding pocket demonstrated higher iron-binding and improved pro-differentiation activities. Transferrin-bound immodulin shows binding preference for both high-molecular-weight hyaluronan (HMWHA) and HMWHA:CD44 complexes at endosomal pH, and interacts with PIP2 at normal physiological pH, offering intriguing mechanistic insights.
Collapse
Affiliation(s)
- Desmond D. Mascarenhas
- Ph.D. Mayflower Organization for Research and Education, 428 Oakmead Parkway, Sunnyvale, CA 94085, USA. Phone: 001 408-523-6279 Bibliography online: https://www.ncbi.nlm.nih.gov/myncbi/desmond.mascarenhas.1/bibliography/public/ ORCID iD: 0000-0002-0710-9960
| |
Collapse
|
13
|
Cohick WS. The role of the IGF system in mammary physiology of ruminants. Domest Anim Endocrinol 2022; 79:106709. [PMID: 35078102 DOI: 10.1016/j.domaniend.2021.106709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022]
Abstract
The IGF system plays a central role in all stages of mammary development, lactation and involution. IGFs exert their effects on the mammary gland through both endocrine and paracrine/autocrine mechanisms and the importance of circulating versus local IGF action remains an open question, especially in ruminants. At the whole organ level, a critical role for IGFs in ductal morphogenesis and lobuloalveolar development has been established, while at the cellular level the ability of IGFs to stimulate cell proliferation and control cell survival contributes to the number of milk-secreting cells in the gland. Much of this work has been conducted in rodents which provide an affordable research model and allow for genetic manipulation of specific components of the IGF system. Research into the role of the IGF system in dairy cows has generally supported information obtained with rodents though large gaps in our knowledge remain and species differences are not well defined. Examples include whether exogenous somatotropin exerts its effects on the mammary gland through local IGF-1 synthesis which is accepted dogma in rodents, what the role of IGF-1 versus IGF-2 is in the mammary gland, and how the IGFBPs regulate IGF bioactivity. This last area is particularly under-investigated in ruminants both at the whole animal and the cellular and molecular levels. Given that the IGF system may underlie many management practices that could contribute to enhancing productive efficiency of lactation, more research into the basic biology of this important system is warranted.
Collapse
Affiliation(s)
- Wendie S Cohick
- Rutgers, The State University of New Jersey, Department of Animal Science, New Brunswick, NJ 08901, USA.
| |
Collapse
|
14
|
Türkez H, Arslan ME, Sönmez E, Tatar A, Geyikoğlu F, Açikyildiz M, Mardinoğlu A. Safety Assessments of Nickel Boride Nanoparticles on the Human Pulmonary Alveolar Cells by Using Cell Viability and Gene Expression Analyses. Biol Trace Elem Res 2021; 199:2602-2611. [PMID: 32909113 DOI: 10.1007/s12011-020-02374-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022]
Abstract
Nickel boride is generally used in the steel industry as a melting accelerator due to its feature of creating a protective and stable attribute at high temperatures. It is also used to improve the hardenability of the steel with boron addition in the production. Thus, safety studies and biocompatibility analysis of nickel boride should be performed comprehensively to understand the limitations of use in various areas. In the present study, nickel boride nanoparticles (Ni2B NPs) were synthesized by a single-step method and molecule characterizations were performed via the use of X-ray diffraction analysis (XRD), transmission electron microscopy (TEM), scanning electron microscopy (SEM), and energy dispersive X-ray (EDX) analyses. Cytotoxicity properties of Ni2B NPs were identified on human pulmonary alveolar epithelial cells (HPAEpiC) by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), neutral red (NR), and lactate dehydrogenase (LDH) assays. Illumina human ht-12 v4.0 whole-genome microarray analysis was conducted to investigate NiB2 NPs effects on gene expression regulations of HPAEpiC cells. The database for annotation, visualization, and integrated discovery (DAVID) analysis was performed to reveal the relationship between Ni2B NP application and cellular pathway alterations. According to cytotoxicity analysis, the IC50 value for Ni2B NP application was found as 81.99 mg/L concentration. Microarray analysis of Ni2B NP application was shown for the first time that 693 gene expression changes (FC ≥ 2) occurred significantly over 40.000 gene probes and Ni2B NPs were observed to affect microtubule regulation, centrosome organization, and phosphoprotein synthesis.
Collapse
Affiliation(s)
- Hasan Türkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey.
| | - Erdal Sönmez
- Advanced Materials Research Laboratory, Department of Nanoscience & Nanoengineering, Graduate School of Natural and Applied Sciences, Ataturk University, Erzurum, Turkey
| | - Abdulgani Tatar
- Department of Medical Genetics, Medical Faculty, Atatürk University, Erzurum, Turkey
| | - Fatime Geyikoğlu
- Department of Biology, Faculty of Arts and Sciences, Atatürk University, Erzurum, Turkey
| | - Metin Açikyildiz
- Department of Chemistry, Faculty of Science and Art, Kilis 7 Aralık University, Kilis, Turkey
| | - Adil Mardinoğlu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| |
Collapse
|
15
|
Mattoso Miskulin Cardoso AP, Tavares Pereira M, Dos Santos Silva R, Medeiros de Carvalho Sousa LM, Giometti IC, Kowalewski MP, de Carvalho Papa P. Global transcriptome analysis implicates cholesterol availability in the regulation of canine cyclic luteal function. Gen Comp Endocrinol 2021; 307:113759. [PMID: 33771531 DOI: 10.1016/j.ygcen.2021.113759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/06/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022]
Abstract
Considering the key role of the corpus luteum in the regulation of the canine diestrus, the present study aimed to investigate changes in the luteal transcriptome of pseudopregnant dogs (n = 18) from days (D) 10, 20, 30, 40, 50 and 60 post-ovulation. After RNAsequencing was performed, data was analyzed by resorting to several informatic tools. A total of 3300 genes were differently expressed among all samples (FDR < 0.01). By comparing different time points, enriched biological processes as response to estradiol and lipids (D20 vs D10) and intracellular cholesterol transport (D40 vs D60) were observed. Moreover, LXR/RXR (liver X receptor- retinoid X receptor) signaling appeared as an overrepresented pathway in all comparisons. Thus, the expression of 19 genes involved in intracellular cholesterol availability was further evaluated; most were affected by time (P < 0.05). Adding to the deep transcriptomic analysis, presented data implies the importance of cholesterol regulation in luteal physiology of pseudopregnant dogs.
Collapse
Affiliation(s)
| | - Miguel Tavares Pereira
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Renata Dos Santos Silva
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Ines Cristina Giometti
- Faculty of Veterinary Medicine, University of Western São Paulo, Presidente Prudente, Brazil
| | | | - Paula de Carvalho Papa
- School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil; Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
LeRoith D, Holly JMP, Forbes BE. Insulin-like growth factors: Ligands, binding proteins, and receptors. Mol Metab 2021; 52:101245. [PMID: 33962049 PMCID: PMC8513159 DOI: 10.1016/j.molmet.2021.101245] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/09/2021] [Accepted: 04/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The insulin-like growth factor family of ligands (IGF-I, IGF-II, and insulin), receptors (IGF-IR, M6P/IGF-IIR, and insulin receptor [IR]), and IGF-binding proteins (IGFBP-1-6) play critical roles in normal human physiology and disease states. SCOPE OF REVIEW Insulin and insulin receptors are the focus of other chapters in this series and will therefore not be discussed further. Here we review the basic components of the IGF system, their role in normal physiology and in critical pathology's. While this review concentrates on the role of IGFs in human physiology, animal models have been essential in providing understanding of the IGF system, and its regulation, and are briefly described. MAJOR CONCLUSIONS IGF-I has effects via the circulation and locally within tissues to regulate cellular growth, differentiation, and survival, thereby controlling overall body growth. IGF-II levels are highest prenatally when it has important effects on growth. In adults, IGF-II plays important tissue-specific roles, including the maintenance of stem cell populations. Although the IGF-IR is closely related to the IR it has distinct physiological roles both on the cell surface and in the nucleus. The M6P/IGF-IIR, in contrast, is distinct and acts as a scavenger by mediating internalization and degradation of IGF-II. The IGFBPs bind IGF-I and IGF-II in the circulation to prolong their half-lives and modulate tissue access, thereby controlling IGF function. IGFBPs also have IGF ligand-independent cell effects.
Collapse
Affiliation(s)
- Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeff M P Holly
- Translational Health Sciences, Bristol Medical School, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Briony E Forbes
- Discipline of Medical Biochemistry, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, Australia
| |
Collapse
|
17
|
Draghici CC, Miulescu RG, Petca RC, Petca A, Dumitrașcu MC, Șandru F. Teratogenic effect of isotretinoin in both fertile females and males (Review). Exp Ther Med 2021; 21:534. [PMID: 33815607 DOI: 10.3892/etm.2021.9966] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Isotretinoin is an oral derivate of vitamin A that has been used since 1982 for the treatment of multiple dermatologic conditions such as severe acne, rosacea, scarring alopecia, ichthyosis or non-melanoma skin cancer prophylaxis. The recommended dose is 0.5-1 mg/kg/day for a period of 4-6 months in sebaceous gland pathologies. There are many adverse effects caused by isotretinoin but by far the most important is the teratogenicity induced by this drug which is estimated to have a 20-35% risk to infants that are exposed to isotretinoin in utero and includes numerous congenital defects such as craniofacial defects, cardiovascular and neurological malformations or thymic disorders. Isotretinoin induces apoptosis and cell cycle arrest in human sebocytes, emphasizing these as processes associated with its teratogenic effect. The aim of this review is to analyze the latest literature data regarding the teratogenic effect of isotretinoin for both fertile females and males and its biological effects underlying the occurrence of congenital malformations under the influence of isotretinoin.
Collapse
Affiliation(s)
- Carmen-Cristina Draghici
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Physioplogy, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Raluca-Gabriela Miulescu
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Farmacology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Răzvan-Cosmin Petca
- Department of Urology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Urology, 'Prof. Dr. Theodor Burghele' Clinical Hospital, 061344 Bucharest, Romania
| | - Aida Petca
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Obstetrics and Gynecology, 'Elias' Emergency Hospital, 011461 Bucharest, Romania
| | - Mihai Cristian Dumitrașcu
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania.,Department of Obstetrics and Gynecology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Florica Șandru
- Department of Dermatology, 'Elias' Emergency University Hospital, 011461 Bucharest, Romania.,Department of Dermatology, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
18
|
Beletskiy A, Chesnokova E, Bal N. Insulin-Like Growth Factor 2 As a Possible Neuroprotective Agent and Memory Enhancer-Its Comparative Expression, Processing and Signaling in Mammalian CNS. Int J Mol Sci 2021; 22:ijms22041849. [PMID: 33673334 PMCID: PMC7918606 DOI: 10.3390/ijms22041849] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
A number of studies performed on rodents suggest that insulin-like growth factor 2 (IGF-2) or its analogs may possibly be used for treating some conditions like Alzheimer’s disease, Huntington’s disease, autistic spectrum disorders or aging-related cognitive impairment. Still, for translational research a comparative knowledge about the function of IGF-2 and related molecules in model organisms (rats and mice) and humans is necessary. There is a number of important differences in IGF-2 signaling between species. In the present review we emphasize species-specific patterns of IGF-2 expression in rodents, humans and some other mammals, using, among other sources, publicly available transcriptomic data. We provide a detailed description of Igf2 mRNA expression regulation and pre-pro-IGF-2 protein processing in different species. We also summarize the function of IGF-binding proteins. We describe three different receptors able to bind IGF-2 and discuss the role of IGF-2 signaling in learning and memory, as well as in neuroprotection. We hope that comprehensive understanding of similarities and differences in IGF-2 signaling between model organisms and humans will be useful for development of more effective medicines targeting IGF-2 receptors.
Collapse
|
19
|
Zielinska HA, Daly CS, Alghamdi A, Bahl A, Sohail M, White P, Dean SR, Holly JMP, Perks CM. Interaction between GRP78 and IGFBP-3 Affects Tumourigenesis and Prognosis in Breast Cancer Patients. Cancers (Basel) 2020; 12:E3821. [PMID: 33352865 PMCID: PMC7767108 DOI: 10.3390/cancers12123821] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 01/09/2023] Open
Abstract
Insulin-like growth factor binding protein 3 (IGFBP-3) plays a key role in breast cancer progression and was recently shown to bind to the chaperone protein glucose-regulated protein 78 (GRP78); however, the clinical significance of this association remains poorly investigated. Here we report a direct correlation between the expression of GRP78 and IGFBP-3 in breast cancer cell lines and tumour sections. Kaplan-Meier survival plots revealed that patients with low GRP78 expression that are positive for IGFBP-3 had poorer survival rates than those with low IGFBP-3 levels, and we observed a similar trend in the publicly available METABRIC gene expression database. With breast cancer cells, in vitro IGFBP-3 enhanced induced apoptosis, however when GRP78 expression was silenced the actions of IGFBP-3 were switched from increasing to inhibiting ceramide (C2)-induced cell death and promoted cell invasion. Using immunofluorescence and cell surface biotinylation, we showed that knock-down of GRP78 negated the entry of IGFBP-3 into the cells. Together, our clinical and experimental results suggest that loss of GRP78 reduces IGFBP-3 entry into cells switching its actions to promote tumorigenesis and predicts a poor prognosis in breast cancer patients.
Collapse
Affiliation(s)
- Hanna A. Zielinska
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
| | - Carl S. Daly
- Faculty of Health Sciences, University of the West England, Bristol BS16 1QY, UK; (C.S.D.); (P.W.); (S.R.D.)
| | - Ahmad Alghamdi
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
- Faculty of Applied medical Sciences, Taif University, Taif, Saudi Arabia
| | - Amit Bahl
- Bristol Haematology and Oncology Centre, Department of Clinical Oncology, University Hospitals Bristol, Bristol BS2 8ED, UK;
| | - Muhammed Sohail
- Faculty of Life Sciences, School of Cellular and Molecular Medicine, Bristol University, Bristol BS8 1TD, UK;
| | - Paul White
- Faculty of Health Sciences, University of the West England, Bristol BS16 1QY, UK; (C.S.D.); (P.W.); (S.R.D.)
| | - Sarah R. Dean
- Faculty of Health Sciences, University of the West England, Bristol BS16 1QY, UK; (C.S.D.); (P.W.); (S.R.D.)
| | - Jeff M. P. Holly
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
| | - Claire M. Perks
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
| |
Collapse
|
20
|
Molecular evidence of IGFBP-3 dependent and independent VD3 action and its nonlinear response on IGFBP-3 induction in prostate cancer cells. BMC Cancer 2020; 20:802. [PMID: 32831047 PMCID: PMC7446217 DOI: 10.1186/s12885-020-07310-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/17/2020] [Indexed: 12/02/2022] Open
Abstract
Background Clinical trials have been conducted to clarify the beneficial effects of VD3 (1α,25-dihydroxy vitamin D3, also known as calcitriol) treatment in prostate cancer. However, the molecular mechanisms underlying these effects are not fully understood. Recent studies on IGFBP-3 have indicated its intracellular functions in cell growth and apoptosis. The aim of this study was to confirm the benefits of low-dose VD3 treatment and clarify the molecular mechanisms underlying these beneficial effects in prostate cancer cells. Methods The molecular effects of simultaneous treatment of LNCaP cells and their genetically modified cell lines with low concentration of docetaxel and VD3 were biologically and biochemically analyzed. To further determine the effects of VD3 treatment on IGFBP-3 induction system, cells were temporarily treated with VD3 in combination with a transcriptional inhibitor or protein synthesis inhibitor. Bcl-2 protein and its mRNA behavior were also observed in Igfbp-3 expression-modified LNCaP cells to determine the involvement of IGFBP-3 in the suppression of Bcl-2 by VD3 treatment. Results Changes in IGFBP-3 expression levels in LNCaP cells indicated that it mediated the inhibition of cell growth induced by VD3 treatment. IGFBP-3 was also found to be a mediator of the enhanced cytotoxicity of prostate cancer cells to VD3 in combination with the anti-cancer drug. We further identified the distinct property of the IGFBP-3 induction system, wherein temporal VD3 stimulation-induced prolonged IGFBP-3 expression and VD3 treatment-induced increase in IGFBP-3 expression were optimized based on the protein concentration rather than the mRNA concentration. Meanwhile, Bcl-2 expression was down-regulated by VD3 treatment in an IGFBP-3-independent manner. Conclusion These findings indicate the molecular mechanisms of IGFBP-3 induction stimulated by VD3 and IGFBP-3 independent Bcl-2 suppression by VD3 treatment in prostate cancer cells. The results could prompt a re-evaluation of VD3 usage in therapy for patients with prostate cancer.
Collapse
|
21
|
Hao J, Yang H, Cao Y, Zhang C, Fan Z. IGFBP5 enhances the dentinogenesis potential of dental pulp stem cells via JNK and ErK signalling pathways. J Oral Rehabil 2020; 47:1557-1565. [DOI: 10.1111/joor.13047] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/13/2020] [Accepted: 06/23/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Junling Hao
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing China
- Department of Endodontics Capital Medical University School of Stomatology Beijing China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing China
| | - Chen Zhang
- Department of Endodontics 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 ReconstructionCapital Medical University School of Stomatology Beijing China
| |
Collapse
|
22
|
Shih HJ, Chen CL, Torng PL. IGFBP3 inhibits angiogenesis through intracellular regulation of THBS1 expression. Am J Cancer Res 2020; 10:1728-1744. [PMID: 32642286 PMCID: PMC7339270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/19/2020] [Indexed: 06/11/2023] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP3) has been postulated to be a mediator of growth suppression signaling. It was shown to function as a suppressor of invasion in epithelial ovarian cancer (EOC). In this study, we identified an angiogenesis inhibitor, thrombospondin-1 (THBS1), which correlated with IGFBP3 expression in EOC cells. After restoring IGFBP3 expression in an EOC cell line using an inducible plasmid, the transfectants showed an increase in IGFBP3 associated with a parallel increase in THBS1. IGFBP3 decreased cell capillary tube formation in HUVECs, which was reversed after anti-THBS1 treatment. IGFBP3 also decreased blood vessel development in chick embryo chorioallantoic membrane (CAM) assay, which was reversed after THBS1 silencing using THBS1 siRNA. Heterotransplantation of IGFBP3 transfectants significantly decreased tumor growth and vascular formation. Luciferase promoter assay illustrated that THBS1 promoter was activated in the presence of both intracellular and extracellular IGFBP3. The signal was stronger in intracellular IGFBP3 expression than that in extracellular IGFBP3 neutralization. In conclusion, we have identified a novel association between IGFBP3 expression and THBS1 elevation, which consequently results in a decrease in angiogenesis. IGFBP3 could activate THBS1 through promoter regulation mainly via an intracellular signaling pathway. Such angiogenesis-regulating ability could be associated with tumor progression and may represent a major function of IGFBP3 as an onco-suppressor in the pathogenesis of ovarian cancer.
Collapse
Affiliation(s)
- Ho-Jun Shih
- Graduate Institute of Clinical Medicine College of Medicine, National Taiwan UniversityTaiwan
| | - Chi-Ling Chen
- Graduate Institute of Clinical Medicine College of Medicine, National Taiwan UniversityTaiwan
- Department of Internal Medicine and Hepatitis Research Center, National Taiwan University College of Medicine and HospitalTaipei, Taiwan
| | - Pao-Ling Torng
- Department of Obstetrics and Gynecology, National Taiwan University HospitalTaipei, Taiwan
- Department of Obstetrics and Gynecology, Hsin-Chu Branch, National Taiwan University HospitalHsin-Chu, Taiwan
| |
Collapse
|
23
|
Yaqoob U, Luo F, Greuter T, Jalan Sakrikar N, Sehrawat TS, Lu J, Hu X, Gao J, Kostallari E, Chen J, Arab JP, Martin-Mateos R, Cao S, Shah VH. GIPC-Regulated IGFBP-3 Promotes HSC Migration In Vitro and Portal Hypertension In Vivo Through a β1-Integrin Pathway. Cell Mol Gastroenterol Hepatol 2020; 10:545-559. [PMID: 32447051 PMCID: PMC7399184 DOI: 10.1016/j.jcmgh.2020.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Transforming growth factor (TGF-β)-induced activation of quiescent hepatic stellate cells (HSCs) and their transformation to myofibroblasts is a key event in liver fibrosis and portal hypertension. GIPC (also referred to as synectin) is a downstream signal activation molecule of TGF-β and other receptors. In this study, we sought to identify novel genes targeted by TGF-β and GIPC and elucidate if and how they may contribute to liver fibrosis. METHODS We performed sequential messenger RNA sequencing analysis on TGF-β-stimulated HSCs and then on TGF-β-stimulated HSCs in the presence and absence of GIPC also referred to as synectin (GIPC) knockdown. Insulin-like growth factor binding protein-3 (IGFBP-3) transport protein emerged as a top activation target of both TGF-β and GIPC. Quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, targeted chromatin immunoprecipitation, and Western blot analysis were done for further confirmation. RESULTS IGFBP-3, an insulin growth factor transport protein, emerged as a top activation target of both TGF-β and GIPC, which was confirmed by quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and Western blot analysis. Targeted chromatin immunoprecipitation showed that GIPC increases the histone 3 lysine 27 (H3K27) acetylation activating mark and concurrently decreases the H3K27 inhibitory trimethylation (H3K27m3) mark, providing an epigenetic correlate to the gene regulation changes. In vivo, global knockout of IGFBP-3 mice resulted in attenuation of HSC activation markers and attenuation of portal pressure in response to chronic liver injury models. Analysis of serum levels from cirrhotic patients also showed an IGFBP-3 increase of more than 2-fold compared with healthy controls. Finally, in vitro mechanism studies showed that IGFBP-3 promotes HSC migration through integrin-dependent phosphorylation of protein kinase B. CONCLUSIONS TGF-β up-regulates IGFBP-3 through GIPC, leading to increased HSC migration in vitro and promotes portal hypertension in vivo. These studies support the role of IGFBP-3 as a potential pathophysiologic target or biomarker in chronic liver disease.
Collapse
Affiliation(s)
- Usman Yaqoob
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Fanghong Luo
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Cancer Research Center, Medical College, Xiamen University, Xiamen, China
| | - Thomas Greuter
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Nidhi Jalan Sakrikar
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Tejasav S Sehrawat
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jianwen Lu
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Xiao Hu
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jinhang Gao
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Enis Kostallari
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jingbiao Chen
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Juan Pablo Arab
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Rosa Martin-Mateos
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology and Hepatology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Sheng Cao
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | - Vijay H Shah
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
24
|
Rao P, Suvas PK, Jerome AD, Steinle JJ, Suvas S. Role of Insulin-Like Growth Factor Binding Protein-3 in the Pathogenesis of Herpes Stromal Keratitis. Invest Ophthalmol Vis Sci 2020; 61:46. [PMID: 32106295 PMCID: PMC7329945 DOI: 10.1167/iovs.61.2.46] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose The goal of this study was to determine the role of insulin-like growth factor-binding protein-3 (IGFBP-3) in the pathogenesis of herpes stromal keratitis (HSK). Methods In an unbiased approach, a membrane-based protein array was carried out to determine the level of expression of pro- and anti-angiogenic molecules in uninfected and HSV-1 infected corneas. Quantitative RT-PCR and ELISA assays were performed to measure the amounts of IGFBP-3 at mRNA and protein levels. Confocal microscopy documented the localization of IGFBP-3 in uninfected and infected corneal tissue. Flow cytometry assay showed the frequency of immune cell types in infected corneas from C57BL/6J (B6) and IGFBP-3 knockout (IGFBP-3-/-) mice. Slit-lamp microscopy was used to quantitate the development of opacity and neovascularization in infected corneas from both groups of mice. Results Quantitation of protein array dot blot showed an increased level of IGFBP-3 protein in HSV-1 infected than uninfected corneas and was confirmed with ELISA and quantitative RT-PCR assays. Cytosolic and nuclear localization of IGFBP-3 were detected in the cells of corneal epithelium, whereas scattered IGFBP-3 staining was evident in the stroma of HSK developing corneas. Increased opacity and hemangiogenesis were noted in the corneas of IGFBP-3-/- than B6 mice during the clinical period of HSK. Furthermore, an increased number of leukocytes comprising of neutrophils and CD4 T cells were found in HSK developing corneas of IGFBP-3-/- than B6 mice. Conclusions Our data showed that lack of IGFBP-3 exacerbates HSK, suggesting the protective effect of IGFBP-3 protein in regulating the severity of HSK.
Collapse
|
25
|
Poreba E, Durzynska J. Nuclear localization and actions of the insulin-like growth factor 1 (IGF-1) system components: Transcriptional regulation and DNA damage response. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 784:108307. [PMID: 32430099 DOI: 10.1016/j.mrrev.2020.108307] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor (IGF) system stimulates growth, proliferation, and regulates differentiation of cells in a tissue-specific manner. It is composed of two insulin-like growth factors (IGF-1 and IGF-2), six insulin-like growth factor-binding proteins (IGFBPs), and two insulin-like growth factor receptors (IGF-1R and IGF-2R). IGF actions take place mostly through the activation of the plasma membrane-bound IGF-Rs by the circulating ligands (IGFs) released from the IGFBPs that stabilize their levels in the serum. This review focuses on the IGF-1 part of the system. The IGF-1 gene, which is expressed mainly in the liver as well as in other tissues, comprises six alternatively spliced exons that code for three protein isoforms (pro-IGF-1A, pro-IGF-1B, and pro-IGF-1C), which are processed to mature IGF-1 and E-peptides. The IGF-1R undergoes autophosphorylation, resulting in a signaling cascade involving numerous cytoplasmic proteins such as AKT and MAPKs, which regulate the expression of target genes. However, a more complex picture of the axis has recently emerged with all its components being translocated to the nuclear compartment. IGF-1R takes part in the regulation of gene expression by forming transcription complexes, modifying the activity of chromatin remodeling proteins, and participating in DNA damage tolerance mechanisms. Four IGFBPs contain a nuclear localization signal (NLS), which targets them to the nucleus, where they regulate gene expression (IGFBP-2, IGFBP-3, IGFBP-5, IGFBP-6) and DNA damage repair (IGFBP-3 and IGFBP-6). Last but not least, the IGF-1B isoform has been reported to be localized in the nuclear compartment. However, no specific molecular actions have been assigned to the nuclear pro-IGF-1B or its derivative EB peptide. Therefore, further studies are needed to shed light on their nuclear activity. These recently uncovered nuclear actions of different components of the IGF-1 axis are relevant in cancer cell biology and are discussed in this review.
Collapse
Affiliation(s)
- Elzbieta Poreba
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| | - Julia Durzynska
- Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
26
|
Sakata J, Hirosue A, Yoshida R, Matsuoka Y, Kawahara K, Arita H, Nakashima H, Yamamoto T, Nagata M, Kawaguchi S, Gohara S, Nagao Y, Yamana K, Toya R, Murakami R, Kuwahara Y, Fukumoto M, Nakayama H. Enhanced Expression of IGFBP-3 Reduces Radiosensitivity and Is Associated with Poor Prognosis in Oral Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12020494. [PMID: 32093285 PMCID: PMC7072421 DOI: 10.3390/cancers12020494] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/09/2020] [Accepted: 02/18/2020] [Indexed: 11/16/2022] Open
Abstract
Insulin-like growth factor (IGF) binding protein-3 (IGFBP-3) modulates various cell functions through IGF-dependent or independent mechanisms. However, its biological roles in the radiosensitivity of oral squamous cell carcinoma (OSCC) remain largely unknown. The purpose of this study was to determine the clinical significance and molecular mechanisms of the association between IGFBP-3 and OSCC radiosensitivity. We performed an immunohistochemical analysis of IGFBP-3 in 52 OSCC specimens from patients treated with preoperative chemoradiotherapy and surgery (phase II study). Associations between IGFBP-3 expression and clinicopathological features were also evaluated. In addition, we examined the effects of IGFBP-3 on post-X-ray irradiation radiosensitivity and DNA damage in vitro. High IGFBP-3 expression was significantly correlated with poor chemoradiotherapy responses and prognosis. With IGFBP-3 knockdown, irradiated OSCC cells exhibited significantly higher radiosensitivity compared with that of control cells. Moreover, IGFBP-3 depletion in OSCC cells reduced phosphorylation of the DNA-dependent protein kinase catalytic subunit (DNA-PKcs), which is required for DNA double-strand break repair during non-homologous end joining. These findings indicate that IGFBP-3 may have a significant role in regulating DNA repair and is be a potential biomarker for predicting clinical response to radiotherapy and prognosis in OSCC.
Collapse
Affiliation(s)
- Junki Sakata
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Akiyuki Hirosue
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
- Correspondence: (A.H.); (H.N.); Tel.: +81-96-373-5288 (A.H. & H.N.)
| | - Ryoji Yoshida
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Yuichiro Matsuoka
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Kenta Kawahara
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Hidetaka Arita
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Hikaru Nakashima
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Tatsuro Yamamoto
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Masashi Nagata
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Sho Kawaguchi
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Shunsuke Gohara
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Yuka Nagao
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Keisuke Yamana
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
| | - Ryo Toya
- Department of Radiation Oncology, Kumamoto University Hospital, Kumamoto 860-8556, Japan;
| | - Ryuji Murakami
- Department of Medical Imaging, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0976, Japan;
| | - Yoshikazu Kuwahara
- Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsushima, Aoba, Sendai, Miyagi 981-8558, Japan;
| | - Manabu Fukumoto
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku, Tokyo 160-8402, Japan;
| | - Hideki Nakayama
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (J.S.); (R.Y.); (Y.M.); (K.K.); (H.A.); (H.N.); (T.Y.); (M.N.); (S.K.); (S.G.); (Y.N.); (K.Y.)
- Correspondence: (A.H.); (H.N.); Tel.: +81-96-373-5288 (A.H. & H.N.)
| |
Collapse
|
27
|
Wang EA, Chen WY, Wong CH. Multiple Growth Factor Targeting by Engineered Insulin-like Growth Factor Binding Protein-3 Augments EGF Receptor Tyrosine Kinase Inhibitor Efficacy. Sci Rep 2020; 10:2735. [PMID: 32066763 PMCID: PMC7026407 DOI: 10.1038/s41598-020-59466-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/23/2020] [Indexed: 01/07/2023] Open
Abstract
Resistance to cancer therapy is a challenge because of innate tumor heterogeneity and constant tumor evolution. Since the pathway of resistance cannot be predicted, combination therapies may address this progression. We discovered that in addition to IGF1 and IGF2, IGFBP-3 binds bFGF, HGF, neuregulin, and PDGF AB with nanomolar affinity. Because growth factors drive resistance, simultaneous inhibition of multiple growth factor pathways may improve the efficacy of precision therapy. Growth factor sequestration by IGFBP-3-Fc enhances the activity of EGFR inhibitors by decreasing cell survival and inhibiting bFGF, HGF, and IGF1 growth factor rescue and also potentiates the activity of other cancer drugs. Inhibition of tumor growth in vivo with adjuvant IGFBP-3-Fc with erlotinib versus erlotinib after treatment cessation supports that the combination reduces cell survival. Inhibition of multiple growth factor pathways may postpone resistance and extend progression-free survival in many cancer indications.
Collapse
Affiliation(s)
- Elizabeth A Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| | - Wan-Yu Chen
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan. .,Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
28
|
Vassilieva I, Kosheverova V, Vitte M, Kamentseva R, Shatrova A, Tsupkina N, Skvortsova E, Borodkina A, Tolkunova E, Nikolsky N, Burova E. Paracrine senescence of human endometrial mesenchymal stem cells: a role for the insulin-like growth factor binding protein 3. Aging (Albany NY) 2020; 12:1987-2004. [PMID: 31951594 PMCID: PMC7053595 DOI: 10.18632/aging.102737] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022]
Abstract
Stress-induced premature cell senescence is well recognized to be accompanied by emerging the senescence-associated secretory phenotype (SASP). Secreted SASP factors can promote the senescence of normal neighboring cells through autocrine/paracrine pathways and regulate the senescence response, as well. Regarding human endometrium-derived mesenchymal stem cells (MESCs), the SASP regulation mechanisms as well as paracrine activity of senescent cells have not been studied yet. Here, we examined the role of insulin-like growth factor binding protein 3 (IGFBP3) in the paracrine senescence induction in young MESCs. The H2O2-induced premature senescence of MESCs led to increased IGFBP3 in conditioned media (CM). The inhibitory analysis of both MAPK and PI3K signaling pathways showed that IGFBP3 releasing from senescent cells is mainly regulated by PI3K/Akt pathway activity. IGFBP3 appears to be an important senescence-mediating factor as its immunodepletion from the senescent CM weakened the pro-senescent effect of CM on young MESCs and promoted their growth. In contrast, young MESCs acquired the senescence phenotype in response to simultaneous addition of recombinant IGFBP3 (rIGFBP3). The mechanism of extracellular IGFBP3 internalization was also revealed. The present study is the first to demonstrate a significant role of extracellular IGFBP3 in paracrine senescence induction of young MESCs.
Collapse
Affiliation(s)
- Irina Vassilieva
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Vera Kosheverova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Mikhail Vitte
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Rimma Kamentseva
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Alla Shatrova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Natalia Tsupkina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Elena Skvortsova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Aleksandra Borodkina
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Elena Tolkunova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Nikolay Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Elena Burova
- Department of Intracellular Signaling and Transport, Institute of Cytology of the Russian Academy of Sciences, St. Petersburg 194064, Russia
| |
Collapse
|
29
|
Stuard WL, Titone R, Robertson DM. The IGF/Insulin-IGFBP Axis in Corneal Development, Wound Healing, and Disease. Front Endocrinol (Lausanne) 2020; 11:24. [PMID: 32194500 PMCID: PMC7062709 DOI: 10.3389/fendo.2020.00024] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022] Open
Abstract
The insulin-like growth factor (IGF) family plays key roles in growth and development. In the cornea, IGF family members have been implicated in proliferation, differentiation, and migration, critical events that maintain a smooth refracting surface that is essential for vision. The IGF family is composed of multiple ligands, receptors, and ligand binding proteins. Expression of IGF type 1 receptor (IGF-1R), IGF type 2 receptor (IGF-2R), and insulin receptor (INSR) in the cornea has been well characterized, including the presence of the IGF-1R and INSR hybrid (Hybrid-R) in the corneal epithelium. Recent data also indicates that each of these receptors display unique intracellular localization. Thus, in addition to canonical ligand binding at the plasma membrane and the initiation of downstream signaling cascades, IGF-1R, INSR, and Hybrid-R also function to regulate mitochondrial stability and nuclear gene expression. IGF-1 and IGF-2, two of three principal ligands, are polypeptide growth factors that function in all cellular layers of the cornea. Unlike IGF-1 and IGF-2, the hormone insulin plays a unique role in the cornea, different from many other tissues in the body. In the corneal epithelium, insulin is not required for glucose uptake, due to constitutive activation of the glucose transporter, GLUT1. However, insulin is needed for the regulation of metabolism, circadian rhythm, autophagy, proliferation, and migration after wounding. There is conflicting evidence regarding expression of the six IGF-binding proteins (IGFBPs), which function primarily to sequester IGF ligands. Within the cornea, IGFBP-2 and IGFBP-3 have identified roles in tissue homeostasis. While IGFBP-3 regulates growth control and intracellular receptor localization in the corneal epithelium, both IGFBP-2 and IGFBP-3 function in corneal fibroblast differentiation and myofibroblast proliferation, key events in stromal wound healing. IGFBP-2 has also been linked to cellular overgrowth in pterygium. There is a clear role for IGF family members in regulating tissue homeostasis in the cornea. This review summarizes what is known regarding the function of IGF and related proteins in corneal development, during wound healing, and in the pathophysiology of disease. Finally, we highlight key areas of research that are in need of future study.
Collapse
|
30
|
Zheng Y, Sowers JY, Houston KD. IGFBP-1 Expression Promotes Tamoxifen Resistance in Breast Cancer Cells via Erk Pathway Activation. Front Endocrinol (Lausanne) 2020; 11:233. [PMID: 32435229 PMCID: PMC7218143 DOI: 10.3389/fendo.2020.00233] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/31/2020] [Indexed: 01/18/2023] Open
Abstract
Insulin-like growth factor (IGF) system plays a significant role in many cellular processes, including proliferation, and survival. In estrogen receptor positive breast cancer, the level of circulating IGF-1 is positively associated with the incidence and at least 50% of cases have elevated IGF-1R signaling. Tamoxifen, a selective estrogen receptor modulator and antagonist for estrogen receptor alpha (ERα) in breast tissue, is a commonly prescribed adjuvant treatment for patients presenting with ERα-positive breast cancer. Unfortunately, tamoxifen resistance is a frequent occurrence in patients receiving treatment and the molecular mechanisms that underlie tamoxifen resistance not adequately defined. It has recently been reported that the inhibition of IGF-1R activation and the proliferation of breast cancer cells upon tamoxifen treatment is mediated by the accumulation of extracellular insulin-like growth factor binding protein 1 (IGFBP-1). Elevated IGFBP-1 expression was observed in tamoxifen-resistant (TamR) MCF-7 and T-47D cells lines suggesting that the tamoxifen-resistant state is associated with IGFBP-1 accumulation. MCF-7 and T-47D breast cancer cells stably transfected with and IGFBP-1 expression vector were generated (MCF7-BP1 and T47D-BP1) to determine the impact of breast cancer cell culture in the presence of increased IGFBP-1 expression. In these cells, the expression of IGF-1R was significantly reduced compared to controls and was similar to our observations in tamoxifen-resistant MCF-7 and T-47D cells. Also similar to TamR breast cancer cells, MCF7-BP1 and T47D-BP1 were resistant to tamoxifen treatment, had elevated epidermal growth factor receptor (EGFR) expression, increased phospho-EGFR (pEGFR), and phospho-Erk (pErk). Furthermore, tamoxifen sensitivity was restored in the MCF7-BP1 and T47D-BP1 upon inhibition of Erk phosphorylation. Lastly, the transient knockdown of IGFBP-1 in MCF7-BP1 and T47D-BP1 inhibited pErk accumulation and increased tamoxifen sensitivity. Taken together, these data support the conclusion that IGFBP-1 is a key component of the development of tamoxifen resistance in breast cancer cells.
Collapse
|
31
|
Hack NL, Cordova KL, Glaser FL, Journey ML, Resner EJ, Hardy KM, Beckman BR, Lema SC. Interactions of long-term food ration variation and short-term fasting on insulin-like growth factor-1 (IGF-1) pathways in copper rockfish (Sebastes caurinus). Gen Comp Endocrinol 2019; 280:168-184. [PMID: 31022390 DOI: 10.1016/j.ygcen.2019.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Accepted: 04/21/2019] [Indexed: 12/26/2022]
Abstract
Variation in food intake affects somatic growth by altering the expression of hormones in the somatotropic endocrine axis including insulin-like growth factor-1 (IGF-1). Here, we examined IGF-1 pathway responses to long- and short-term variation in food availability in copper rockfish (Sebastes caurinus), a nearshore Pacific rockfish important for commercial and recreational fisheries. Juvenile copper rockfish were raised under differing ration amounts (3% or 9% mass feed·g-1 fish wet mass·day-1) for 140 d to simulate 'long-term' feeding variation, after which some fish from both rations were fasted for 12 d to generate 'short-term' conditions of food deprivation. Rockfish on the 9% ration treatment grew more quickly than those on the 3% ration and were larger in mass, length, and body condition (k) after 152 d. Fish on the 9% ration had higher blood glucose than those on the 3% ration, with fasting decreasing blood glucose in both ration treatments, indicating that both long-term and short-term feed treatments altered energy status. Plasma IGF-1 was higher in rockfish from the 9% ration than those in the 3% ration and was also higher in fed fish than fasted fish. Additionally, plasma IGF-1 related positively to individual variation in specific growth rate (SGR). The positive association between IGF-1 and SGR showed discordance in fish that had experienced different levels of food and growth over the long-term but not short-term, suggesting that long-term nutritional experience can influence the relationship between IGF-1 and growth in this species. Rockfish on the 3% ration showed a lower relative abundance of gene transcripts encoding igf1 in the liver, but higher hepatic mRNAs for IGF binding proteins igfbp1a and igfbp1b. Fasting similarly decreased the abundance of igf1 mRNAs in the liver of fish reared under both the 9% and 3% rations, while concurrently increasing mRNAs encoding the IGF binding proteins igfbp1a, -1b, and -3a. Hepatic mRNAs for igfbp2b, -5a, and -5b were lower with long-term ration variation (3% ration) and fasting. Fish that experienced long-term reduced rations also had higher mRNA levels for igfbp3a, -3b, and IGF receptors isoforms A (igf1rA) and B (igf1rB) in skeletal muscle, but lower mRNA levels for igf1. Fasting increased muscle mRNA abundance for igfbp3a, igf1rA, and igf1rB, and decreased levels for igfbp2a and igf1. These data show that a positive relationship between circulating IGF-1 and individual growth rate is maintained in copper rockfish even when that growth variation relates to differences in food consumption across varying time scales, but that long- and short-term variation in food quantity can shift basal concentrations of circulating IGF-1 in this species.
Collapse
Affiliation(s)
- Nicole L Hack
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Kasey L Cordova
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Frances L Glaser
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Meredith L Journey
- Lynker Technology, 202 Church St SE #536, Leesburg, VA 20175, Under Contract to Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA 98112, USA
| | - Emily J Resner
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Kristin M Hardy
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Brian R Beckman
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - Sean C Lema
- Biological Sciences Department, Center for Coastal Marine Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA.
| |
Collapse
|
32
|
Regulation of IGF-I by IGFBP3 and IGFBP5 during odontoblast differentiation in mice. J Oral Biosci 2019; 61:157-162. [PMID: 31400542 DOI: 10.1016/j.job.2019.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Although intracellular signaling pathways of insulin-like growth factor I (IGF-I) related to the proliferation of dental pulp cells have been investigated, the switching mechanism from cell proliferation to differentiation during odontogenesis remains elusive. This study aimed to elucidate the role of IGF binding protein (IGFBP) 3 and 5 in regulation of IGF-I during odontoblast differentiation in mouse incisors. METHODS The detailed expression patterns of IGF-I, IGF-I receptor (IGF-IR), IGFBP3, and IGFBP5 together with that of an odontoblast differentiation marker, nestin, were examined by immunohistochemistry and/or in situ hybridization using paraffinized sections of TetOP-H2B-GFP mouse incisors at postnatal 4 weeks. RESULTS Undifferentiated dental papilla cells and preodontoblasts (preOB) showed intense IGF-I- and IGF-IRα-positive reactions, and the expression was observed in differentiated odontoblasts, such as immature odontoblasts (iOB) and mature odontoblasts (mOB). IGFBP3/Igfbp3 was transiently expressed in preOB and early iOB, and the intensity of expression gradually reduced with the progression of odontoblast differentiation. In contrast, immunohistochemical analysis for IGFBP5 identified a positive reaction in the undifferentiated dental papilla cells and differentiated odontoblasts, and the expression of Igfbp5 was reduced in the differentiated odontoblasts. CONCLUSION The present study demonstrated the expression patterns of IGF-I, IGF-IR, IGFBP3, and IGFBP5 during odontoblast differentiation in mouse incisors. These results suggested that IGFBP3 regulates the transition from the proliferative to differentiation stage by inhibiting the action of IGF-I on the proliferation of dental papilla cells, and that IGFBP5 plays an important role in the maintenance of the differentiated odontoblasts during tooth development.
Collapse
|
33
|
de Silva HC, Lin MZ, Phillips L, Martin JL, Baxter RC. IGFBP-3 interacts with NONO and SFPQ in PARP-dependent DNA damage repair in triple-negative breast cancer. Cell Mol Life Sci 2019; 76:2015-2030. [PMID: 30725116 PMCID: PMC11105386 DOI: 10.1007/s00018-019-03033-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/09/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022]
Abstract
Women with triple-negative breast cancer (TNBC) are generally treated by chemotherapy but their responsiveness may be blunted by DNA double-strand break (DSB) repair. We previously reported that IGFBP-3 forms nuclear complexes with EGFR and DNA-dependent protein kinase (DNA-PKcs) to modulate DSB repair by non-homologous end-joining (NHEJ) in TNBC cells. To discover IGFBP-3 binding partners involved in chemoresistance through stimulation of DSB repair, we analyzed the IGFBP-3 interactome by LC-MS/MS and confirmed interactions by coimmunoprecipitation and proximity ligation assay. Functional effects were demonstrated by DNA end-joining in vitro and measurement of γH2AX foci. In response to 20 µM etoposide, the DNA/RNA-binding protein, non-POU domain-containing octamer-binding protein (NONO) and its dimerization partner splicing factor, proline/glutamine-rich (SFPQ) formed complexes with IGFBP-3, demonstrated in basal-like TNBC cell lines HCC1806 and MDA-MB-468. NONO binding to IGFBP-3 was also shown in a cell-free biochemical assay. IGFBP-3 complexes with NONO and SFPQ were blocked by inhibiting EGFR with gefitinib or DNA-PKcs with NU7026, and by the PARP inhibitors veliparib and olaparib, which also reduced DNA end-joining activity and delayed the resolution of the γH2AX signal (i.e. inhibited DNA DSB repair). Downregulation of the long noncoding RNA in NHEJ pathway 1 (LINP1) by siRNA also blocked IGFBP-3 interaction with NONO-SFPQ. These findings suggest a PARP-dependent role for NONO and SFPQ in IGFBP-3-dependent DSB repair and the involvement of LINP1 in the complex formation. We propose that targeting of the DNA repair function of IGFBP-3 may enhance chemosensitivity in basal-like TNBC, thus improving patient outcomes.
Collapse
Affiliation(s)
- Hasanthi C de Silva
- Kolling Institute, Royal North Shore Hospital, The University of Sydney, St. Leonards, NSW, 2065, Australia
| | - Mike Z Lin
- Kolling Institute, Royal North Shore Hospital, The University of Sydney, St. Leonards, NSW, 2065, Australia
- Orange Family Medical Centre, 95 Peisley Street, Orange, NSW, 2800, Australia
| | - Leo Phillips
- Kolling Institute, Royal North Shore Hospital, The University of Sydney, St. Leonards, NSW, 2065, Australia
| | - Janet L Martin
- Kolling Institute, Royal North Shore Hospital, The University of Sydney, St. Leonards, NSW, 2065, Australia
| | - Robert C Baxter
- Kolling Institute, Royal North Shore Hospital, The University of Sydney, St. Leonards, NSW, 2065, Australia.
| |
Collapse
|
34
|
Gene expression analysis reveals early dysregulation of disease pathways and links Chmp7 to pathogenesis of spinal and bulbar muscular atrophy. Sci Rep 2019; 9:3539. [PMID: 30837566 PMCID: PMC6401132 DOI: 10.1038/s41598-019-40118-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/04/2019] [Indexed: 01/09/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) results from a CAG repeat expansion within the androgen receptor gene (AR). It is unclear why motor neurons selectively degenerate and there are currently no treatments for this debilitating disease. To uncover the causative genes and pathways involved in motor neuron dysfunction, we undertook transcriptomic profiling of primary embryonic motor neurons from SBMA mice. We show that transcriptional dysregulation occurs early during development in SBMA motor neurons. One gene found to be dysregulated, Chmp7, was also altered in vivo in spinal cord before symptom onset in SBMA mice, and crucially in motor neuron precursor cells derived from SBMA patient stem cells, suggesting that Chmp7 may play a causal role in disease pathogenesis by disrupting the endosome-lysosome system. Furthermore, genes were enriched in SBMA motor neurons in several key pathways including p53, DNA repair, WNT and mitochondrial function. SBMA embryonic motor neurons also displayed dysfunctional mitochondria along with DNA damage, possibly resulting from DNA repair gene dysregulation and/or mitochondrial dysfunction. This indicates that a coordinated dysregulation of multiple pathways leads to development of SBMA. Importantly, our findings suggest that the identified pathways and genes, in particular Chmp7, may serve as potential therapeutic targets in SBMA.
Collapse
|
35
|
Daruich A, Le Rouzic Q, Jonet L, Naud MC, Kowalczuk L, Pournaras JA, Boatright JH, Thomas A, Turck N, Moulin A, Behar-Cohen F, Picard E. Iron is neurotoxic in retinal detachment and transferrin confers neuroprotection. SCIENCE ADVANCES 2019; 5:eaau9940. [PMID: 30662950 PMCID: PMC6326753 DOI: 10.1126/sciadv.aau9940] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/28/2018] [Indexed: 05/12/2023]
Abstract
In retinal detachment (RD), photoreceptor death and permanent vision loss are caused by neurosensory retina separating from the retinal pigment epithelium because of subretinal fluid (SRF), and successful surgical reattachment is not predictive of total visual recovery. As retinal iron overload exacerbates cell death in retinal diseases, we assessed iron as a predictive marker and therapeutic target for RD. In the vitreous and SRF from patients with RD, we measured increased iron and transferrin (TF) saturation that is correlated with poor visual recovery. In ex vivo and in vivo RD models, iron induces immediate necrosis and delayed apoptosis. We demonstrate that TF decreases both apoptosis and necroptosis induced by RD, and using RNA sequencing, pathways mediating the neuroprotective effects of TF are identified. Since toxic iron accumulates in RD, we propose TF supplementation as an adjunctive therapy to surgery for improving the visual outcomes of patients with RD.
Collapse
Affiliation(s)
- Alejandra Daruich
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, 75006 Paris, France
- Department of ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
- Ophthalmology Department, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Quentin Le Rouzic
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, 75006 Paris, France
| | - Laurent Jonet
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, 75006 Paris, France
| | - Marie-Christine Naud
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, 75006 Paris, France
| | - Laura Kowalczuk
- Department of ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Jean-Antoine Pournaras
- Department of ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Jeffrey H. Boatright
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, GA, USA
- Center of Excellence, Atlanta Veterans Administration Medical Center, Decatur, GA, USA
| | - Aurélien Thomas
- Geneva University Hospitals, Unit of Toxicology, CURML, Geneva, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
| | - Natacha Turck
- Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
- Geneva University, Department of Human Protein Science, Geneva, Switzerland
| | - Alexandre Moulin
- Department of ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Francine Behar-Cohen
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, 75006 Paris, France
- Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
- Ophtalmopole, Cochin Hospital, AP-HP, Assistance Publique Hôpitaux de Paris, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France
- Corresponding author.
| | - Emilie Picard
- INSERM, UMRS1138, Team 17, From physiopathology of ocular diseases to clinical development, Université Sorbonne Paris Cité, Centre de Recherche des Cordeliers, 15 rue de l’Ecole de Médecine, 75006 Paris, France
| |
Collapse
|
36
|
Hack NL, Strobel JS, Journey ML, Beckman BR, Lema SC. Response of the insulin-like growth factor-1 (Igf1) system to nutritional status and growth rate variation in olive rockfish (Sebastes serranoides). Comp Biochem Physiol A Mol Integr Physiol 2018; 224:42-52. [DOI: 10.1016/j.cbpa.2018.05.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/28/2018] [Accepted: 05/30/2018] [Indexed: 12/20/2022]
|
37
|
Lan M, Zhu XP, Cao ZY, Liu JM, Lin Q, Liu ZL. Extracellular vesicles-mediated signaling in the osteosarcoma microenvironment: Roles and potential therapeutic targets. J Bone Oncol 2018; 12:101-104. [PMID: 30155405 PMCID: PMC6111053 DOI: 10.1016/j.jbo.2018.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS) is the most common non-hematologic malignant tumor of bone in children. It is usually characterized by a high risk of developing lung metastasis and poor prognosis. Extracellular vesicles (EVs) are cell-derived nanoparticles with a small size of 50–200 nm in diameter. As a communicator, the contents of the EVs secreted via either fusing with lysosomes for degradation and recycling or fusing with the cell plasma membrane into the extracellular environment, which play an important role in regulating the tumor microenvironment of OS and mediating the Wnt/β-catenin and TGF-β signalings. Increasing evidences suggest that EVs have significant role in OS growth, progression, metastasis and drug resistance. In this study, the roles of EVs in the physiology and pathogenesis of OS and the potential attractive therapeutic target for the treatment of OS were reviewed.
Collapse
Affiliation(s)
- Min Lan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Xiao-Ping Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Zhi-Yuan Cao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Jia-Ming Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhi-Li Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| |
Collapse
|
38
|
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) 1-6 bind IGFs but not insulin with high affinity. They were initially identified as serum carriers and passive inhibitors of IGF actions. However, subsequent studies showed that, although IGFBPs inhibit IGF actions in many circumstances, they may also potentiate these actions. IGFBPs are widely expressed in most tissues, and they are flexible endocrine and autocrine/paracrine regulators of IGF activity, which is essential for this important physiological system. More recently, individual IGFBPs have been shown to have IGF-independent actions. Mechanisms underlying these actions include (i) interaction with non-IGF proteins in compartments including the extracellular space and matrix, the cell surface and intracellular space, (ii) interaction with and modulation of other growth factor pathways including EGF, TGF-β and VEGF, and (iii) direct or indirect transcriptional effects following nuclear entry of IGFBPs. Through these IGF-dependent and IGF-independent actions, IGFBPs modulate essential cellular processes including proliferation, survival, migration, senescence, autophagy and angiogenesis. They have been implicated in a range of disorders including malignant, metabolic, neurological and immune diseases. A more complete understanding of their cellular roles may lead to the development of novel IGFBP-based therapeutic opportunities.
Collapse
Affiliation(s)
- L A Bach
- Department of Medicine (Alfred)Monash University, Melbourne, Australia
- Department of Endocrinology and DiabetesAlfred Hospital, Melbourne, Australia
| |
Collapse
|
39
|
Aleksic T, Gray N, Wu X, Rieunier G, Osher E, Mills J, Verrill C, Bryant RJ, Han C, Hutchinson K, Lambert AG, Kumar R, Hamdy FC, Weyer-Czernilofsky U, Sanderson MP, Bogenrieder T, Taylor S, Macaulay VM. Nuclear IGF1R Interacts with Regulatory Regions of Chromatin to Promote RNA Polymerase II Recruitment and Gene Expression Associated with Advanced Tumor Stage. Cancer Res 2018; 78:3497-3509. [PMID: 29735545 PMCID: PMC6031306 DOI: 10.1158/0008-5472.can-17-3498] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/28/2018] [Accepted: 04/26/2018] [Indexed: 01/02/2023]
Abstract
Internalization of ligand-activated type I IGF receptor (IGF1R) is followed by recycling to the plasma membrane, degradation or nuclear translocation. Nuclear IGF1R reportedly associates with clinical response to IGF1R inhibitory drugs, yet its role in the nucleus is poorly characterized. Here, we investigated the significance of nuclear IGF1R in clinical cancers and cell line models. In prostate cancers, IGF1R was predominantly membrane localized in benign glands, while malignant epithelium contained prominent internalized (nuclear/cytoplasmic) IGF1R, and nuclear IGF1R associated significantly with advanced tumor stage. Using ChIP-seq to assess global chromatin occupancy, we identified IGF1R-binding sites at or near transcription start sites of genes including JUN and FAM21, most sites coinciding with occupancy by RNA polymerase II (RNAPol2) and histone marks of active enhancers/promoters. IGF1R was inducibly recruited to chromatin, directly binding DNA and interacting with RNAPol2 to upregulate expression of JUN and FAM21, shown to mediate tumor cell survival and IGF-induced migration. IGF1 also enriched RNAPol2 on promoters containing IGF1R-binding sites. These functions were inhibited by IGF1/II-neutralizing antibody xentuzumab (BI 836845), or by blocking receptor internalization. We detected IGF1R on JUN and FAM21 promoters in fresh prostate cancers that contained abundant nuclear IGF1R, with evidence of correlation between nuclear IGF1R content and JUN expression in malignant prostatic epithelium. Taken together, these data reveal previously unrecognized molecular mechanisms through which IGFs promote tumorigenesis, with implications for therapeutic evaluation of anti-IGF drugs.Significance: These findings reveal a noncanonical nuclear role for IGF1R in tumorigenesis, with implications for therapeutic evaluation of IGF inhibitory drugs. Cancer Res; 78(13); 3497-509. ©2018 AACR.
Collapse
Affiliation(s)
- Tamara Aleksic
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Nicki Gray
- Computational Biology Research Group, University of Oxford, Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
| | - Xiaoning Wu
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Eliot Osher
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Jack Mills
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Clare Verrill
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Richard J Bryant
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Cheng Han
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| | | | - Adam G Lambert
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rajeev Kumar
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | - Thomas Bogenrieder
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
- Department of Urology, University Hospital Grosshadern, Ludwig-Maximilians-University, Marchioninistrasse, Munich, Germany
| | - Stephen Taylor
- Computational Biology Research Group, University of Oxford, Weatherall Institute of Molecular Medicine, Oxford, United Kingdom
| | - Valentine M Macaulay
- Department of Oncology, University of Oxford, Oxford, United Kingdom.
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
40
|
Simabuco FM, Morale MG, Pavan IC, Morelli AP, Silva FR, Tamura RE. p53 and metabolism: from mechanism to therapeutics. Oncotarget 2018; 9:23780-23823. [PMID: 29805774 PMCID: PMC5955117 DOI: 10.18632/oncotarget.25267] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
The tumor cell changes itself and its microenvironment to adapt to different situations, including action of drugs and other agents targeting tumor control. Therefore, metabolism plays an important role in the activation of survival mechanisms to keep the cell proliferative potential. The Warburg effect directs the cellular metabolism towards an aerobic glycolytic pathway, despite the fact that it generates less adenosine triphosphate than oxidative phosphorylation; because it creates the building blocks necessary for cell proliferation. The transcription factor p53 is the master tumor suppressor; it binds to more than 4,000 sites in the genome and regulates the expression of more than 500 genes. Among these genes are important regulators of metabolism, affecting glucose, lipids and amino acids metabolism, oxidative phosphorylation, reactive oxygen species (ROS) generation and growth factors signaling. Wild-type and mutant p53 may have opposing effects in the expression of these metabolic genes. Therefore, depending on the p53 status of the cell, drugs that target metabolism may have different outcomes and metabolism may modulate drug resistance. Conversely, induction of p53 expression may regulate differently the tumor cell metabolism, inducing senescence, autophagy and apoptosis, which are dependent on the regulation of the PI3K/AKT/mTOR pathway and/or ROS induction. The interplay between p53 and metabolism is essential in the decision of cell fate and for cancer therapeutics.
Collapse
Affiliation(s)
- Fernando M. Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mirian G. Morale
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Isadora C.B. Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana P. Morelli
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando R. Silva
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo E. Tamura
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
41
|
Abstract
Insulinlike growth factor (IGF) binding proteins (IGFBPs) 1 to 6 are high-affinity regulators of IGF activity. They generally inhibit IGF actions by preventing binding to the IGF-I receptor but can also enhance their actions under some conditions. Posttranslational modifications such as glycosylation and phosphorylation modulate IGFBP properties, and IGFBP proteolysis results in IGF release. IGFBPs have more recently been shown to have IGF-independent actions. A number of mechanisms are involved, including modulation of other growth factor pathways, nuclear localization and transcriptional regulation, interaction with the sphingolipid pathway, and binding to non-IGF biomolecules in the extracellular space and matrix, on the cell surface and intracellularly. IGFBPs modulate important biological processes, including cell proliferation, survival, migration, senescence, autophagy, and angiogenesis. Their actions have been implicated in growth, metabolism, cancer, stem cell maintenance and differentiation, and immune regulation. Recent studies have shown that epigenetic mechanisms are involved in the regulation of IGFBP abundance. A more complete understanding of IGFBP biology is necessary to further define their cellular roles and determine their therapeutic potential.
Collapse
Affiliation(s)
- Leon A Bach
- Department of Endocrinology and Diabetes, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
He Y, Yuan X, Zhou G, Feng A. Activation of IGF-1/IGFBP-3 signaling by berberine improves intestinal mucosal barrier of rats with acute endotoxemia. Fitoterapia 2018; 124:200-205. [DOI: 10.1016/j.fitote.2017.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 11/06/2017] [Accepted: 11/13/2017] [Indexed: 12/19/2022]
|
43
|
Julovi SM, Martin JL, Baxter RC. Nuclear Insulin-Like Growth Factor Binding Protein-3 As a Biomarker in Triple-Negative Breast Cancer Xenograft Tumors: Effect of Targeted Therapy and Comparison With Chemotherapy. Front Endocrinol (Lausanne) 2018; 9:120. [PMID: 29623068 PMCID: PMC5874320 DOI: 10.3389/fendo.2018.00120] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/09/2018] [Indexed: 12/22/2022] Open
Abstract
Triple-negative breast cancer (TNBC) typically has a worse outcome than other breast cancer subtypes, in part owing to a lack of approved therapeutic targets or prognostic markers. We have previously described an oncogenic pathway in basal-like TNBC cells, initiated by insulin-like growth factor binding protein-3 (IGFBP-3), in which the epidermal growth factor receptor (EGFR) is transactivated by sphingosine-1-phosphate (S1P) resulting from sphingosine kinase (SphK)-1 activation. Oncogenic IGFBP-3 signaling can be targeted by combination treatment with the S1P receptor modulator and SphK inhibitor, fingolimod, and the EGFR kinase inhibitor, gefitinib (F + G). However, the interaction of this treatment with chemotherapy has not been documented. Since we observed nuclear localization of IGFBP-3 in some TNBC tumors, this study aimed to evaluate the prognostic significance of nuclear IGFBP-3 in pre-clinical models of basal-like TNBC treated with F + G and doxorubicin. Orthotopic xenograft tumors were grown in nude mice from the human basal-like TNBC cell lines MDA-MB-468 and HCC1806, and were treated with gefitinib, 25 mg/Kg, plus fingolimod, 5 mg/Kg, 3-times weekly. In some studies, doxorubicin was also administered once weekly for 6 weeks. Tumor tissue proteins were quantitated by immunohistochemistry (IHC). Interaction between doxorubicin and F + G was also studied in proliferation assays in vitro. In both tumor models, tissue staining for IGFBP-3 was predominantly nuclear. Combination of F + G significantly enhanced mouse survival, decreased nuclear IGFBP-3 and Ki67 staining, and increased apoptosis (cleaved caspase-3) staining. Kaplan-Meier survival analysis showed that a high tumor IGFBP-3 IHC score (>median), like a high Ki67 score, was significantly associated with shorter survival time, whereas a high apoptosis score was associated with prolonged survival. Studied in vitro in both cell lines, low-dose doxorubicin that had little effect alone, strongly enhanced the cytostatic effect of low-dose F + G combination. However, in both in vivo models, doxorubicin at maximum-tolerated dose neither inhibited tumor growth when administered alone, nor enhanced the significant inhibitory effect of F + G. We conclude that doxorubicin may not add benefit to the inhibitory effect of F + G unless its dose-limiting toxicity can be overcome. Nuclear IGFBP-3 appears to have potential as a prognostic marker in TNBC and could be evaluated for clinical utility.
Collapse
|
44
|
Oncogenic function of the homeobox A13-long noncoding RNA HOTTIP-insulin growth factor-binding protein 3 axis in human gastric cancer. Oncotarget 2017; 7:36049-36064. [PMID: 27144338 PMCID: PMC5094982 DOI: 10.18632/oncotarget.9102] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/11/2016] [Indexed: 12/11/2022] Open
Abstract
To study the mechanisms of gastric tumorigenesis, we have established CSN cell line from human normal gastric mucosa, and CS12, a tumorigenic and invasive gastric cancer cell line from CSN passages. Many stem cell markers were expressed in both CSN and CS12 cells, but LGR5 and NANOG were expressed only in CS12 cells. Increased expression of homeobox A13 (HoxA13) and its downstream cascades was significant for the tumorigenic activity of CS12 cells, and was associated with recruitment of E2F-1 to HoxA13 promoter accompanied with increased trimethylation of histone H3 lysine 4 (H3K4me3) at the hypomethylated E2F motifs. Knockdown of HoxA13 caused the downregulation of long non-coding RNA HOTTIP and insulin growth factor-binding protein 3 (IGFBP-3) genes, indicating that both were targets of HoxA13. Concurrent regulation of HoxA13-HOTTIP was mediated by the mixed lineage leukemia-WD repeat domain 5 complex, which caused the trimethylation of H3K4 and then stimulated cell proliferation. HoxA13 transactivated the IGFBP-3 promoter through the HOX-binding site. Activation of IGFBP-3 stimulated the oncogenic potential and invasion activity. Increased expression of HoxA13 (63.2%) and IGFBP-3 (28.6%) was detected in human gastric cancer tissues and was found in the gastric cancer data of The Cancer Genome Atlas. Taken together, the HoxA13–HOTTIP–IGFBP-3 cascade is critical for the carcinogenic characteristics of CS12 cells.
Collapse
|
45
|
Minchenko DO, Tsymbal DO, Yavorovsky OP, Solokha NV, Minchenko OH. Expression of genes encoding IGFBPs, SNARK, CD36, and PECAM1 in the liver of mice treated with chromium disilicide and titanium nitride nanoparticles. Endocr Regul 2017; 51:84-95. [PMID: 28609285 DOI: 10.1515/enr-2017-0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE The aim of the present study was to examine the effect of chromium disilicide and titanium nitride nanoparticles on the expression level of genes encoding important regulatory factors (IGFBP1, IGFBP2, IGFBP3, IGFBP4, IGFBP5, SNARK/NUAK2, CD36, and PECAM1/CD31) in mouse liver for evaluation of possible toxic effects of these nanoparticles. METHODS Male mice received 20 mg chromium disilicide nanoparticles (45 nm) and titanium nitride nanoparticles (20 nm) with food every working day for 2 months. The expression of IGFBP1, IGFBP2, IGFBP3, IGFBP4, IGFBP5, SNARK, CD36, and PECAM1 genes in mouse liver was studied by quantitative polymerase chain reaction. RESULTS Treatment of mice with chromium disilicide nanoparticles led to down-regulation of the expression of IGFBP2, IGFBP5, PECAM1, and SNARK genes in the liver in comparison with control mice, with more prominent changes for SNARK gene. At the same time, the expression of IGFBP3 and CD36 genes was increased in mouse liver upon treatment with chromium disilicide nanoparticles. We have also shown that treatment with titanium nitride nanoparticles resulted in down-regulation of the expression of IGFBP2 and SNARK genes in the liver with more prominent changes for SNARK gene. At the same time, the expression of IGFBP3, IGFBP4, and CD36 genes was increased in the liver of mice treated with titanium nitride nanoparticles. Furthermore, the effect of chromium disilicide nanoparticles on IGFBP2 and CD36 genes expression was significantly stronger as compared to titanium nitride nanoparticles. CONCLUSIONS The results of this study demonstrate that chromium disilicide and titanium nitride nanoparticles have variable effects on the expression of IGFBP2, IGFBP3, IGFBP4, IGFBP5, SNARK, CD36, and PECAM1 genes in mouse liver, which may reflect the genotoxic activities of the studied nanoparticles.
Collapse
|
46
|
Abstract
This review based on translational research predicts that the transcription factor p53 is the key effector of all anti-acne therapies. All-trans retinoic acid (ATRA) and isotretinoin (13-cis retinoic acid) enhance p53 expression. Tetracyclines and macrolides via inhibiting p450 enzymes attenuate ATRA degradation, thereby increase p53. Benzoyl peroxide and hydrogen peroxide elicit oxidative stress, which upregulates p53. Azelaic acid leads to mitochondrial damage associated with increased release of reactive oxygen species inducing p53. p53 inhibits the expression of androgen receptor and IGF-1 receptor, and induces the expression of IGF binding protein 3. p53 induces FoxO1, FoxO3, p21 and sestrin 1, sestrin 2, and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), the key inducer of isotretinoin-mediated sebocyte apoptosis explaining isotretinoin's sebum-suppressive effect. Anti-androgens attenuate the expression of miRNA-125b, a key negative regulator of p53. It can thus be concluded that all anti-acne therapies have a common mode of action, i.e., upregulation of the guardian of the genome p53. Immortalized p53-inactivated sebocyte cultures are unfortunate models for studying acne pathogenesis and treatment.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7a, 49076, Osnabrück, Germany.
| |
Collapse
|
47
|
The role of insulin growth factor-1 on the vascular regenerative effect of MAA coated disks and macrophage-endothelial cell crosstalk. Biomaterials 2017; 144:199-210. [PMID: 28841464 DOI: 10.1016/j.biomaterials.2017.08.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/04/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022]
Abstract
The IGF-1 signaling pathway and IGF-1-dependent macrophage/endothelial cell crosstalk was found to be critical features of the vascular regenerative effect displayed by implanted methacrylic acid -co-isodecyl acrylate (MAA-co-IDA; 40% MAA) coated disks in CD1 mice. Inhibition of IGF-1 signaling using AG1024 an IGF1-R tyrosine kinase inhibitor abrogated vessel formation 14 days after disk implantation in a subcutaneous pocket. Explanted tissue had increased arginase 1 expression and reduced iNOS expression consistent with the greater shift from "M1" ("pro-inflammatory") macrophages to "M2" ("pro-angiogenic") macrophages for MAA coated disks relative to control MM (methyl methacrylate-co-IDA) disks; the latter did not generate a vascular response and the polarization shift was muted with AG1024. In vitro, medium conditioned by macrophages (both human dTHP1 cells and mouse bone marrow derived macrophages) had elevated IGF-1 mRNA and protein levels, while the cells had reduced IGF1-R but elevated IGFBP-3 mRNA levels. These cells also had reduced iNOS and elevated Arg1 expression, consistent with the in vivo polarization results, including the inhibitory effects of AG1024. On the other hand, HUVEC exposed to dTHP1 conditioned medium migrated and proliferated faster suggesting that the primary target of the macrophage released IGF-1 was endothelial cells. Although further investigation is warranted, IGF-1 appears to be a key feature underpinning the observed vascularization. Why MAA based materials have this effect remains to be defined, however.
Collapse
|
48
|
Martin JL, Julovi SM, Lin MZ, de Silva HC, Boyle FM, Baxter RC. Inhibition of basal-like breast cancer growth by FTY720 in combination with epidermal growth factor receptor kinase blockade. Breast Cancer Res 2017; 19:90. [PMID: 28778177 PMCID: PMC5545026 DOI: 10.1186/s13058-017-0882-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/12/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND New molecular targets are needed for women with triple-negative breast cancer (TNBC). This pre-clinical study investigated the combination of the EGFR inhibitor gefitinib with the sphingosine kinase (SphK) inhibitor FTY720 (Fingolimod), aiming to block tumorigenic signaling downstream of IGFBP-3, which is abundantly expressed in basal-like TNBC. METHODS In studies of breast cancer cell growth in culture, proliferation was monitored by IncuCyte live-cell imaging, and protein abundance was determined by western blotting. In vivo studies of mammary tumor growth used two models: orthotopic xenograft tumors derived from three basal-like TNBC cell lines, grown in immune-deficient mice, and syngeneic murine 4T1 tumors grown in immune-competent mice. Protein abundance in tumor tissue was assessed by immunohistochemistry. RESULTS Quantitated by live-cell imaging, the inhibitor combination showed synergistic cytostatic activity in basal-like cell lines across several TNBC molecular subtypes, the synergy being decreased by IGFBP-3 downregulation. Suppression of the tumorigenic mediator CD44 by gefitinib was potentiated by FTY720, consistent with CD44 involvement in the targeted pathway. In MDA-MB-468 and HCC1806 orthotopic TNBC xenograft tumors in nude mice, the drug combination inhibited tumor growth and prolonged mouse survival, although this effect was not significant for the gefitinib-resistant cell line HCC70. Combination treatment of murine 4T1 TNBC tumors in syngeneic BALB/c mice was more effective in immune-competent than immune-deficient (nude) mice, and a relative loss of tumor CD3 (T-cell) immunoreactivity caused by FTY720 treatment alone was alleviated by the drug combination, suggesting that, even at an FTY720 dose causing relative lymphopenia, the combination is still effective in an immune-competent setting. Immunohistochemistry of xenograft tumors showed significant enhancement of caspase-3 cleavage and suppression of Ki67 and phospho-EGFR by the drug combination, but SphK1 downregulation occurred only in MDA-MB-468 tumors, so is unlikely to be integral to treatment efficacy. CONCLUSIONS Our data indicate that targeting IGFBP-3-dependent signaling pathways through gefitinib-FTY720 co-therapy may be effective in many basal-like breast cancers, and suggest tissue IGFBP-3 and CD44 measurement as potential biomarkers of treatment efficacy.
Collapse
Affiliation(s)
- Janet L Martin
- Kolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Sohel M Julovi
- Kolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Mike Z Lin
- Kolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.,Present address: Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Hasanthi C de Silva
- Kolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Frances M Boyle
- Patricia Ritchie Centre for Cancer Care and Research, Mater Hospital, North Sydney, NSW, 2065, Australia
| | - Robert C Baxter
- Kolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.
| |
Collapse
|
49
|
Cao Z, Koochekpour S, Strup SE, Kyprianou N. Reversion of epithelial-mesenchymal transition by a novel agent DZ-50 via IGF binding protein-3 in prostate cancer cells. Oncotarget 2017; 8:78507-78519. [PMID: 29108245 PMCID: PMC5667978 DOI: 10.18632/oncotarget.19659] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/05/2017] [Indexed: 01/10/2023] Open
Abstract
Dysregulation of transforming growth factor-β1 (TGF-β1) and insulin-like growth factor (IGF) axis has been linked to reactive stroma dynamics in prostate cancer progression. IGF binding protein-3 (IGFBP3) induction is initiated by stroma remodeling and could represent a potential therapeutic target for prostate cancer. In previous studies a lead quinazoline-based Doxazosin® derivative, DZ-50, impaired prostate tumor growth by targeting proteins involved in focal adhesion, anoikis resistance and epithelial-mesenchymal-transition (EMT). This study demonstrates that DZ-50 increased expression of the epithelial marker E-cadherin, and decreased the mesenchymal marker N-cadherin in human prostate cancer cells. In DU-145 cells, the effect of DZ-50 on EMT towards mesenchymal epithelial transition (MET) was inhibited by talin1 overexpression, a focal adhesion regulator promoting anoikis resistance and tumor invasion. DZ-50 treatment of human prostate cancer cells and cancer-associated fibroblasts (CAFs) downregulated IGFBP3 expression at mRNA and protein level. In TGF-β1 responsive LNCaPTβRII, TGF-β1 reversed DZ-50-induced MET by antagonizing the drug-induced decrease of nuclear IGFBP3. Furthermore, co-culture with CAFs promoted prostate cancer epithelial cell invasion, an effect that was significantly inhibited by DZ-50. Our findings demonstrate that the lead compound, DZ-50, inhibited the invasive properties of prostate cancer epithelial cells by targeting IGFBP3 and mediating EMT conversion to MET. This study integrated the mechanisms underlying the effect of DZ-50 and further supported the therapeutic value of this compound in the treatment of advanced metastatic prostate cancer.
Collapse
Affiliation(s)
- Zheng Cao
- Department of Urology, University of Kentucky, Lexington, KY, USA
| | - Shahriar Koochekpour
- Department of Genetics and Genomic and Urology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Stephen E Strup
- Department of Urology, University of Kentucky, Lexington, KY, USA
| | - Natasha Kyprianou
- Department of Urology, University of Kentucky, Lexington, KY, USA.,Departments of Biochemistry and Toxicology & Cancer Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
50
|
Pickard A, Durzynska J, McCance DJ, Barton ER. The IGF axis in HPV associated cancers. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 772:67-77. [PMID: 28528691 DOI: 10.1016/j.mrrev.2017.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/30/2017] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Human papillomaviruses (HPV) infect and replicate in stratified epithelium at cutaneous and mucosal surfaces. The proliferation and maintenance of keratinocytes, the cells which make up this epithelium, are controlled by a number of growth factor receptors such as the keratinocyte growth factor receptor (KGFR, also called fibroblast growth factor receptor 2b (FGFR2b)), the epithelial growth factor receptor (EGFR) and the insulin-like growth factor receptors 1 and 2 (IGF1R and IGF2R). In this review, we will delineate the mutation, gene transcription, translation and processing of the IGF axis within HPV associated cancers. The IGFs are key for developmental and postnatal growth of almost all tissues; we explore whether this crucial axis has been hijacked by HPV.
Collapse
MESH Headings
- Cell Proliferation
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Keratinocytes/cytology
- Keratinocytes/virology
- Neoplasms/genetics
- Neoplasms/virology
- Papillomaviridae/pathogenicity
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, IGF Type 1
- Receptor, IGF Type 2/genetics
- Receptor, IGF Type 2/metabolism
- Receptors, Somatomedin/genetics
- Receptors, Somatomedin/metabolism
- Somatomedins/genetics
- Somatomedins/metabolism
Collapse
Affiliation(s)
- Adam Pickard
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, UK; Wellcome Centre for Cell Matrix Research, University of Manchester, M13 9PL, UK.
| | - Julia Durzynska
- Department of Molecular Virology, Institute of Experimental Biology, A. Mickiewicz University, ul. Umultowska 89, 61-614, Poznań, Poland; Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Dennis J McCance
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| |
Collapse
|