1
|
Cai X, Sun R, Yang L, Yao N, Sun Y, Zhang G, Ge W, Zhou Y, Gui Z, Wang Y, Zheng H, Xu D, Zhao Y, Nie X, Liu Z, Zhang H, Hu P, Cheng H, Xue Z, Wang J, Yu J, Chen C, Luo D, Zhu J, Liu T, Zhang Y, Wu Q, Guo Q, Chen W, Wang J, Wei W, Lin X, Yao J, Wang G, Peng L, Liu S, Wang Z, Liu H, Wang J, Wu F, Yuan Z, Gong T, Lv Y, Xiang J, Zhu Y, Xie L, Ge M, Guan H, Guo T. Proteomic analysis reveals modulation of key proteins in follicular thyroid cancer progression. Chin Med J (Engl) 2025:00029330-990000000-01557. [PMID: 40394764 DOI: 10.1097/cm9.0000000000003645] [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: 10/15/2024] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Cytopathology cannot be used to reliably distinguish follicular thyroid adenoma (FTA) from follicular thyroid carcinoma (FTC), the second most common form of thyroid cancer, because they exhibit nearly identical cellular morphology. Given the challenges in diagnosis and treatment, this study aims to identify the mechanisms underlying FTC is essential. METHODS Using parallel reaction monitoring-mass spectrometry (PRM-MS) assays, we identified and quantified 94 differentially expressed protein candidates from a retrospective cohort of 1085 FTC and FTA tissue samples from 18 clinical centers. Of these targeted proteins, those with the potential for distinguishing FTC from FTA were prioritized using machine learning. Co-immunoprecipitation (co-IP) and immunofluorescence co-localization assays, as well as gene interference, overexpression, and immunohistochemistry (IHC) experiments, were used to investigate the interactions and cellular functions of selected proteins. RESULTS Using machine learning models and feature selection methods, 30 of the 94 candidates were prioritized as key proteins. Co-IP and immunofluorescence co-localization assays using FTC cell lines revealed interactions among insulin-like growth factor 2 receptor (IGF2R), major vault protei (MVP), histone deacetylase 1 (HDAC1), and histone H1.5 (H1-5). Gene interference and overexpression experiments in FTC-133 cells confirmed the promotional role of these proteins in cell proliferation. IHC assays of patient samples further confirmed elevated expression of these four proteins in FTC compared with that in FTA. CONCLUSIONS Our findings underscore the utility of advanced proteomic techniques in elucidating the molecular underpinnings of FTC, highlighting the potential significance of IGF2R, MVP, HDAC1, and H1-5 in FTC progression, and providing a foundation for the exploration of targeted therapies.
Collapse
Affiliation(s)
- Xue Cai
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Rui Sun
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Liang Yang
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Nan Yao
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Yaoting Sun
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Guangmei Zhang
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Weigang Ge
- Westlake Omics (Hangzhou) Biotechnology Co., Ltd. Hangzhou, Zhejiang 310024, China
| | - Yan Zhou
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Zhiqiang Gui
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Haitao Zheng
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Dong Xu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yongfu Zhao
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, China
| | - Xiu Nie
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zhiyan Liu
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Pingping Hu
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Honghan Cheng
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Zhangzhi Xue
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Jiatong Wang
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Jing Yu
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Dingcun Luo
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Jingqiang Zhu
- Division of Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tong Liu
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Yifeng Zhang
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital; Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai 200072, China
| | - Qijun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Qiaonan Guo
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Wanyuan Chen
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310006, China
| | - Jianbiao Wang
- Department of Head and Neck Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| | - Wenjun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiangfeng Lin
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Jincao Yao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, China
| | - Li Peng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shuyi Liu
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Zhihong Wang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Hanqing Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jiaxi Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Fan Wu
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Tingting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Yangfan Lv
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jingjing Xiang
- Department of Pathology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Yi Zhu
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Lei Xie
- Department of Head and Neck Surgery, Institute of Micro-Invasive Surgery of Zhejiang University, The Affiliated Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| | - Minghua Ge
- Otolaryngology and Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310006, China
| | - Haixia Guan
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Tiannan Guo
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Affiliated Hangzhou First People's Hospital, State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| |
Collapse
|
2
|
Kjeldsen CMN, Oxvig C. The Proteinase PAPP-A has Deep Evolutionary Roots Outside of the IGF System. Genome Biol Evol 2025; 17:evaf042. [PMID: 40084812 PMCID: PMC11925022 DOI: 10.1093/gbe/evaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
The animal pappalysin metalloproteinases, PAPP-A and PAPP-A2, are highly specific regulatory enzymes of the insulin-like growth factor (IGF) system. Cleavage of their only known substrates, a subset of IGF binding proteins (IGFBPs), releases bioactive IGFI and IGFII, thus promoting IGF signaling. Stanniocalcin-1 and -2 (STC1 and STC2) are potent pappalysin inhibitors, completing the STC-PAPP-A-IGFBP-IGF axis. Utilizing homology searches and phylogenetic analyses, we examined the occurrence of pappalysins in the animal kingdom and their functional conservation. This revealed the extensive presence of pappalysins across metazoans, as well as the presence of 3 pappalysins: PAPP-A, PAPP-A2, and a third group of invertebrate pappalysins, which we name invertebrate PAPP-A (invPAPP-A). We show that PAPP-A and PAPP-A2 arose by duplication during early vertebrate evolution. Despite significant evolutionary distance, the domain architecture of the metazoan pappalysins is completely conserved, and several functional domains and motifs are highly conserved across all pappalysins. However, invPAPP-A exists outside the context of IGFBPs, suggesting that the animal pappalysins may have substrates beyond the IGFBPs for PAPP-A and PAPP-A2 that remain to be discovered. Since PAPP-A is an emerging drug target, it is important to understand potential involvement in regulatory systems other than the IGF system, which might be affected upon targeting of PAPP-A.
Collapse
Affiliation(s)
- Caroline M N Kjeldsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus DK-8000 C, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus DK-8000 C, Denmark
| |
Collapse
|
3
|
Pan Y, Xiang Q, Deng K, Anwar MI, Wang L, Wang Y, Liang Q, Shen L, Yang J, Hou Z, Shen W. Engineering IGF2 for Lysosome-targeting chimeras development to target drug-resistant membrane proteins in tumor therapy. Protein Sci 2025; 34:e70051. [PMID: 39969096 PMCID: PMC11837023 DOI: 10.1002/pro.70051] [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: 03/26/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 02/20/2025]
Abstract
Lysosome-targeting chimeras (LYTACs) represent a promising approach for the targeted degradation of membrane proteins. Currently, two primary methods for LYTAC development involve chemically modified antibodies and wild-type insulin-like growth factor 2 (IGF2) fusion proteins (iLYTACs). However, LYTACs necessitate intricate chemical modification processes, while wild-type IGF2 in iLYTAC technology binds to IGF1R, potentially triggering carcinogenesis. To tackle this challenge, we introduce specific IGF2R-binding lysosomal targeting chimeras (sLYTACs), a novel technology utilizing engineered IGF2 mutant fusion antibodies for the degradation of endogenous membrane proteins. Diverging from iLYTACs, sLYTACs exhibit selective binding to IGF2R with increased affinity, significantly bolstering the anti-proliferative impact on drug-resistant tumor cells both in vitro and in vivo. By effectively degrading third-generation tyrosine kinase inhibitor-resistant EGFR mutants, masking binding epitope HER2, and concurrently targeting compensatory receptors interacting with these proteins, sLYTACs show great promise in drug development to overcome bypass signaling and combat drug resistance in tumors.
Collapse
Affiliation(s)
- Yanchao Pan
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Qing Xiang
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Kai Deng
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | | | - Leiming Wang
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Yuan Wang
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Qiulian Liang
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Lirou Shen
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Jing Yang
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Zhongyu Hou
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| | - Weijun Shen
- Translational Research CenterShenzhen Bay LaboratoryShenzhenChina
| |
Collapse
|
4
|
Ruan X, Jin X, Sun F, Pi J, Jinghu Y, Lin X, Zhang N, Chen G. IGF signaling pathway in bone and cartilage development, homeostasis, and disease. FASEB J 2024; 38:e70031. [PMID: 39206513 DOI: 10.1096/fj.202401298r] [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: 06/08/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The skeleton plays a fundamental role in the maintenance of organ function and daily activities. The insulin-like growth factor (IGF) family is a group of polypeptide substances with a pronounced role in osteoblast differentiation, bone development, and metabolism. Disturbance of the IGFs and the IGF signaling pathway is inextricably linked with assorted developmental defects, growth irregularities, and jeopardized skeletal structure. Recent findings have illustrated the significance of the action of the IGF signaling pathway via growth factors and receptors and its interactions with dissimilar signaling pathways (Wnt/β-catenin, BMP, TGF-β, and Hh/PTH signaling pathways) in promoting the growth, survival, and differentiation of osteoblasts. IGF signaling also exhibits profound influences on cartilage and bone development and skeletal homeostasis via versatile cell-cell interactions in an autocrine, paracrine, and endocrine manner systemically and locally. Our review summarizes the role and regulatory function as well as a potentially integrated gene network of the IGF signaling pathway with other signaling pathways in bone and cartilage development and skeletal homeostasis, which in turn provides an enlightening insight into visualizing bright molecular targets to be eligible for designing effective drugs to handle bone diseases and maladies, such as osteoporosis, osteoarthritis, and dwarfism.
Collapse
Affiliation(s)
- Xinyi Ruan
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiuhui Jin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Fuju Sun
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiashun Pi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yihan Jinghu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xinyi Lin
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Nenghua Zhang
- Clinical Laboratory, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
5
|
Rao SS, Kundapura SV, Dey D, Palaniappan C, Sekar K, Kulal A, Ramagopal UA. Cumulative phylogenetic, sequence and structural analysis of Insulin superfamily proteins provide unique structure-function insights. Mol Inform 2024; 43:e202300160. [PMID: 38973776 DOI: 10.1002/minf.202300160] [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: 07/03/2023] [Revised: 01/17/2024] [Accepted: 03/14/2024] [Indexed: 07/09/2024]
Abstract
The insulin superfamily proteins (ISPs), in particular, insulin, IGFs and relaxin proteins are key modulators of animal physiology. They are known to have evolved from the same ancestral gene and have diverged into proteins with varied sequences and distinct functions, but maintain a similar structural architecture stabilized by highly conserved disulphide bridges. The recent surge of sequence data and the structures of these proteins prompted a need for a comprehensive analysis, which connects the evolution of these sequences (427 sequences) in the light of available functional and structural information including representative complex structures of ISPs with their cognate receptors. This study reveals (a) unusually high sequence conservation of IGFs (>90 % conservation in 184 sequences) and provides a possible structure-based rationale for such high sequence conservation; (b) provides an updated definition of the receptor-binding signature motif of the functionally diverse relaxin family members (c) provides a probable non-canonical C-peptide cleavage site in a few insulin sequences. The high conservation of IGFs appears to represent a classic case of resistance to sequence diversity exerted by physiologically important interactions with multiple partners. We also propose a probable mechanism for C-peptide cleavage in a few distinct insulin sequences and redefine the receptor-binding signature motif of the relaxin family. Lastly, we provide a basis for minimally modified insulin mutants with potential therapeutic application, inspired by concomitant changes observed in other insulin superfamily protein members supported by molecular dynamics simulation.
Collapse
Affiliation(s)
- Shrilakshmi Sheshagiri Rao
- Division of Biological Sciences, Poornaprajna Institute of Scientific Research, Poornaprajnapura, 562110, Bidalur (Post), Bengaluru, India
- Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Shankar V Kundapura
- Division of Biological Sciences, Poornaprajna Institute of Scientific Research, Poornaprajnapura, 562110, Bidalur (Post), Bengaluru, India
- Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Debayan Dey
- Division of Biological Sciences, Poornaprajna Institute of Scientific Research, Poornaprajnapura, 562110, Bidalur (Post), Bengaluru, India
- Present address: Department of Biochemistry, Emory University School of Medicine, GA 30322, Atlanta, USA
| | - Chandrasekaran Palaniappan
- Department of Computational and Data Sciences, Indian Institute of Science, 560012, Bangalore, India
- Molecular Biophysics Unit, Indian Institute of Science, 560012, Bangalore, India
| | - Kanagaraj Sekar
- Department of Computational and Data Sciences, Indian Institute of Science, 560012, Bangalore, India
| | - Ananda Kulal
- Division of Biological Sciences, Poornaprajna Institute of Scientific Research, Poornaprajnapura, 562110, Bidalur (Post), Bengaluru, India
| | - Udupi A Ramagopal
- Division of Biological Sciences, Poornaprajna Institute of Scientific Research, Poornaprajnapura, 562110, Bidalur (Post), Bengaluru, India
- Department of Microbiology and FST, School of Science, GITAM University, 530045, Visakhapatnam, India
| |
Collapse
|
6
|
Giri S, Allen KJH, Prabaharan CB, Ramirez JB, Fiore L, Uppalapati M, Dadachova E. Initial insights into the interaction of antibodies radiolabeled with Lutetium-177 and Actinium-225 with tumor microenvironment in experimental human and canine osteosarcoma. Nucl Med Biol 2024; 134-135:108917. [PMID: 38718557 DOI: 10.1016/j.nucmedbio.2024.108917] [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: 02/23/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Osteosarcoma (OS) is a prevalent primary bone cancer affecting both humans and canines. This study describes initial insights into the interaction of the human monoclonal antibody IF3 to an insulin-like growth factor 2 receptor (IGF2R) radiolabeled with either alpha-emitting Actinium-225 (225Ac) or beta-emitting Lutetium-177 (177Lu) radionuclides with the OS cells and tumor microenvironment (TME) in experimental human and canine OS. BASIC PROCEDURES SCID mice bearing canine Gracie or human OS-33 OS tumors were treated with 177Lu- or 225Ac-labeled IF3 antibody, sacrificed at 24, 72 or 168 h post-treatment and their tumors were analyzed by immunohistochemistry (IHC) for the presence of OS cells, various elements of TME as well as for the double DNA strand breaks with γH2AX and caspase 3 assays. MAIN FINDINGS IHC revealed a reduction in IGF2R-positive OS cells and OS stem cell populations post therapy with 225Ac- and 177Lu-labeled IF3 antibody. Notably, radiolabeled IF3 antibody effectively diminished pro-tumorigenic M2 macrophages, highlighting its therapeutic promise. The study also unveiled varied responses of natural killer (NK) cells and M1 macrophages, shedding light on the intricate TME interplay. Time-dependent increase in γ-H2AX staining in canine Gracie and human OS-33 tumors treated with [177Lu]Lu-IF3 and [225Ac]Ac-IF3 was observed at 24 and 72 h post-RIT. PRINCIPAL CONCLUSIONS These findings suggest that radiolabeled antibodies offer a hopeful avenue for personalized OS treatment, emphasizing the importance of understanding their impact on the TME and potential synergies with immunotherapy.
Collapse
Affiliation(s)
- Sabeena Giri
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Chandra Bose Prabaharan
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jonathan Bonet Ramirez
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Luciano Fiore
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Maruti Uppalapati
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
7
|
Yang JL, Yamada-Hunter SA, Labanieh L, Sotillo E, Cheah JS, Roberts DS, Mackall CL, Bertozzi CR, Ting AY. Directed evolution of genetically encoded LYTACs for cell-mediated delivery. Proc Natl Acad Sci U S A 2024; 121:e2320053121. [PMID: 38513100 PMCID: PMC10990137 DOI: 10.1073/pnas.2320053121] [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: 11/15/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Lysosome-targeting chimeras (LYTACs) are a promising therapeutic modality to drive the degradation of extracellular proteins. However, early versions of LYTAC contain synthetic glycopeptides that cannot be genetically encoded. Here, we present our designs for a fully genetically encodable LYTAC (GELYTAC), making our tool compatible with integration into therapeutic cells for targeted delivery at diseased sites. To achieve this, we replaced the glycopeptide portion of LYTACs with the protein insulin-like growth factor 2 (IGF2). After showing initial efficacy with wild-type IGF2, we increased the potency of GELYTAC using directed evolution. Subsequently, we demonstrated that our engineered GELYTAC construct not only secretes from HEK293T cells but also from human primary T-cells to drive the uptake of various targets into receiver cells. Immune cells engineered to secrete GELYTAC thus represent a promising avenue for spatially selective targeted protein degradation.
Collapse
Affiliation(s)
- Jonathan Lee Yang
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Sean A. Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Louai Labanieh
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Bioengineering, Stanford University, Stanford, CA94305
- Parker Institute for Cancer Immunotherapy, San Francisco, CA94305
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Bioengineering, Stanford University, Stanford, CA94305
| | - Joleen S. Cheah
- Department of Biology, Stanford University, Stanford, CA94305
| | - David S. Roberts
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Crystal L. Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Parker Institute for Cancer Immunotherapy, San Francisco, CA94305
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA94305
- Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Carolyn R. Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Alice Y. Ting
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Department of Biology, Stanford University, Stanford, CA94305
- Department of Genetics, Stanford University, Stanford, CA94305
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA94158
| |
Collapse
|
8
|
Braulke T, Carette JE, Palm W. Lysosomal enzyme trafficking: from molecular mechanisms to human diseases. Trends Cell Biol 2024; 34:198-210. [PMID: 37474375 DOI: 10.1016/j.tcb.2023.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023]
Abstract
Lysosomes degrade and recycle macromolecules that are delivered through the biosynthetic, endocytic, and autophagic routes. Hydrolysis of the different classes of macromolecules is catalyzed by about 70 soluble enzymes that are transported from the Golgi apparatus to lysosomes in a mannose 6-phosphate (M6P)-dependent process. The molecular machinery that generates M6P tags for receptor-mediated targeting of lysosomal enzymes was thought to be understood in detail. However, recent studies on the M6P pathway have identified a previously uncharacterized core component, yielded structural insights in known components, and uncovered functions in various human diseases. Here we review molecular mechanisms of lysosomal enzyme trafficking and discuss its relevance for rare lysosomal disorders, cancer, and viral infection.
Collapse
Affiliation(s)
- Thomas Braulke
- Department of Osteology and Biomechanics, Cell Biology of Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan E Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Wilhelm Palm
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
9
|
Tian Y, Miao Y, Guo P, Wang J, Han D. Insulin-like Growth Factor 2-Tagged Aptamer Chimeras (ITACs) Modular Assembly for Targeted and Efficient Degradation of Two Membrane Proteins. Angew Chem Int Ed Engl 2024; 63:e202316089. [PMID: 38059276 DOI: 10.1002/anie.202316089] [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: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/08/2023]
Abstract
Overexpression of pathogenic membrane proteins drives abnormal proliferation and invasion of tumor cells. Various strategies to durably knockdown membrane proteins with heterobifunctional degraders have been successfully developed, including LYTAC, KineTAC, and AbTAC. However, challenges including complicated synthetic procedures and the inability to simultaneously degrade multiple pathogenic proteins still exist. Herein, we developed insulin-like growth factor 2 (IGF2)-tagged aptamer chimeras (ITACs) that link the cell-surface lysosome-targeting receptor IGF2R and membrane proteins of interest (POIs) based on specific recognition of aptamers to the POIs and high-affinity binding of IGF2 to IGF2R. We demonstrated that ITACs exhibit robust degradation efficiency of various membrane proteins in multiple cell lines. Furthermore, systematic studies revealed that a moderate cell-surface IGF2R level is responsible for the excellent degradation performance of ITACs. Importantly, we further established a modular assembly strategy that allows assembly of one IGF2 with two aptamers with precise stoichiometry (dITACs), enabling cooperative and simultaneous degradation of two membrane proteins. This work provides an efficient and facile target membrane protein degradation platform and will shed light on the treatment of diseases related to the overexpression of membrane proteins.
Collapse
Affiliation(s)
- Yuan Tian
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanyan Miao
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Pei Guo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Junyan Wang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Da Han
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
10
|
Nelson D, Thompson KJ, Wang L, Wang Z, Eberts P, Azarin SM, Kalari KR, Kandimalla KK. Pericyte Control of Gene Expression in the Blood-Brain Barrier Endothelium: Implications for Alzheimer's Disease. J Alzheimers Dis 2024; 99:S281-S297. [PMID: 38393902 DOI: 10.3233/jad-230907] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Background A strong body of evidence suggests that cerebrovascular pathologies augment the onset and progression of Alzheimer's disease (AD). One distinctive aspect of this cerebrovascular dysfunction is the degeneration of brain pericytes-often overlooked supporting cells of blood-brain barrier endothelium. Objective The current study investigates the influence of pericytes on gene and protein expressions in the blood-brain barrier endothelium, which is expected to facilitate the identification of pathophysiological pathways that are triggered by pericyte loss and lead to blood-brain barrier dysfunction in AD. Methods Bioinformatics analysis was conducted on the RNA-Seq expression counts matrix (GSE144474), which compared solo-cultured human blood-brain barrier endothelial cells against endothelial cells co-cultured with human brain pericytes in a non-contact model. We constructed a similar cell culture model to verify protein expression using western blots. Results The insulin resistance and ferroptosis pathways were found to be enriched. Western blots of the insulin receptor and heme oxygenase expressions were consistent with those observed in RNA-Seq data. Additionally, we observed more than 5-fold upregulation of several genes associated with neuroprotection, including insulin-like growth factor 2 and brain-derived neurotrophic factor. Conclusions Results suggest that pericyte influence on blood-brain barrier endothelial gene expression confers protection from insulin resistance, iron accumulation, oxidative stress, and amyloid deposition. Since these are conditions associated with AD pathophysiology, they imply mechanisms by which pericyte degeneration could contribute to disease progression.
Collapse
Affiliation(s)
- Doug Nelson
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Kevin J Thompson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Lushan Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Zengtao Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Paulina Eberts
- Department of Chemical Engineering and Materials Science, College of Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, College of Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Krishna R Kalari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
11
|
Yang JL, Yamada-Hunter SA, Labanieh L, Sotillo E, Cheah JS, Roberts DS, Mackall CL, Ting AY, Bertozzi CR. Directed Evolution of Genetically Encoded LYTACs for Cell-Mediated Delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567117. [PMID: 38014030 PMCID: PMC10680704 DOI: 10.1101/2023.11.14.567117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Lysosome-targeting chimeras (LYTACs) are a promising therapeutic modality to drive the degradation of extracellular proteins. However, early versions of LYTAC contain synthetic glycopeptides that cannot be genetically encoded. Here we present our designs for a fully genetically encodable LYTAC (GELYTAC), making our tool compatible with integration into therapeutic cells for targeted delivery at diseased sites. To achieve this, we replaced the glycopeptide portion of LYTACs with the protein insulin like growth factor 2 (IGF2). After showing initial efficacy with wild type IGF2, we increased the potency of GELYTAC using directed evolution. Subsequently, we demonstrated that our engineered GELYTAC construct not only secretes from HEK293T cells but also from human primary T-cells to drive the uptake of various targets into receiver cells. Immune cells engineered to secrete GELYTAC thus represent a promising avenue for spatially-selective targeted protein degradation.
Collapse
Affiliation(s)
- Jonathan Lee Yang
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Sean A. Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Louai Labanieh
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena Sotillo
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joleen S. Cheah
- Departments of Biology, and Genetics Stanford University, Stanford, CA 94305, USA
| | - David S. Roberts
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Crystal L. Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94305, USA
| | - Alice Y. Ting
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Departments of Biology, and Genetics Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA 94158, USA
| | - Carolyn R. Bertozzi
- Department of Chemistry and Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| |
Collapse
|
12
|
Mikitiuk M, Barczyński J, Bielski P, Arciniega M, Tyrcha U, Hec A, Lipińska AD, Rychłowski M, Holak TA, Sitar T. IGF2 Peptide-Based LYTACs for Targeted Degradation of Extracellular and Transmembrane Proteins. Molecules 2023; 28:7519. [PMID: 38005242 PMCID: PMC10673611 DOI: 10.3390/molecules28227519] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Lysosome-targeting chimeras (LYTACs) have recently been developed to facilitate the lysosomal degradation of specific extracellular and transmembrane molecular targets. However, the LYTAC particles described to date are based on glycopeptide conjugates, which are difficult to prepare and produce on a large scale. Here, we report on the development of pure protein LYTACs based on the non-glycosylated IGF2 peptides, which can be readily produced in virtually any facility capable of monoclonal antibody production. These chimeras utilize the IGF2R/CI-M6PR pathway for lysosomal shuttling and, in our illustrative example, target programmed death ligand 1 (PD-L1), eliciting physiological effects analogous to immune checkpoint blockade. Results from in vitro assays significantly exceed the effects of anti-PD-L1 antibodies alone.
Collapse
Affiliation(s)
- Michał Mikitiuk
- Recepton Sp. z o.o., Trzy Lipy 3, 80-172 Gdańsk, Poland
- Department of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland
| | - Jan Barczyński
- Recepton Sp. z o.o., Trzy Lipy 3, 80-172 Gdańsk, Poland
- Department of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland
| | | | | | | | | | - Andrea D. Lipińska
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-307 Gdańsk, Poland
| | - Michał Rychłowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-307 Gdańsk, Poland
| | - Tad A. Holak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland;
| | - Tomasz Sitar
- Recepton Sp. z o.o., Trzy Lipy 3, 80-172 Gdańsk, Poland
| |
Collapse
|
13
|
Xiong D, Yang J, Li D, Wang J. Exploration of Key Immune-Related Transcriptomes Associated with Doxorubicin-Induced Cardiotoxicity in Patients with Breast Cancer. Cardiovasc Toxicol 2023; 23:329-348. [PMID: 37684436 PMCID: PMC10514147 DOI: 10.1007/s12012-023-09806-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023]
Abstract
Based on a few studies, heart failure patients with breast cancer were assessed to find potential biomarkers for doxorubicin-induced cardiotoxicity. However, key immune-related transcriptional markers linked to doxorubicin-induced cardiotoxicity in breast cancer patients have not been thoroughly investigated. We used GSE40447, GSE76314, and TCGA BRCA cohorts to perform this study. Then, we performed various bioinformatics approaches to identify the key immune-related transcriptional markers and their association with doxorubicin-induced cardiotoxicity in patients with breast cancer. We found 255 upregulated genes and 286 downregulated genes in patients with doxorubicin-induced heart failure in breast cancer. We discovered that in patients with breast cancer comorbidity doxorubicin-induced cardiotoxicity, the 58 immunological genes are elevated (such as CPA3, VSIG4, GATA2, RFX2, IL3RA, and LRP1), and the 60 genes are significantly suppressed (such as MS4A1, FCRL1, CD200, FCRLA, FCRL2, and CD79A). Furthermore, we revealed that the immune-related differentially expressed genes (DEGs) are substantially associated with the enrichment of KEGG pathways, including B-cell receptor signaling pathway, primary immunodeficiency, chemokine signaling pathway, hematopoietic cell lineage, cytokine-cytokine receptor interaction, Toll-like receptor signaling pathway, MAPK signaling pathway, focal adhesion, dilated cardiomyopathy, cell adhesion molecule, etc. Moreover, we discovered that the doxorubicin-induced immune-related genes are crucially involved in the protein-protein interaction and gene clusters. The immune-related genes, including IFIT5, XCL1, SPIB, BTLA, MS4A1, CD19, TCL1A, CD83, CD200, FCRLA, CD79A, BIRC3, and IGF2R are significantly associated with a poor survival prognosis of breast cancer patients and showed diagnostic efficacy in patients with breast cancer and heart failure. Molecular docking revealed that the survival-associated genes interact with the doxorubicin with appreciable binding affinity. Finally, we validated the expression level of immune-related genes in breast cancer patients-derived cardiomyocytes with doxorubicin-induced cardiotoxicity and found that the level of RAD9A, HSPA1B, GATA2, IGF2R, CD200, ERCC8, and BCL11A genes are consistently dysregulated. Our findings offered a basis for understanding the mechanism and pathogenesis of the cardiotoxicity caused by doxorubicin in breast cancer patients and predicted the interaction of immune-related potential biomarkers with doxorubicin.
Collapse
Affiliation(s)
- Daiqin Xiong
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Jianhua Yang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Dongfeng Li
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Jie Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China.
| |
Collapse
|
14
|
Sharma T, Kundu N, Kaur S, Shankaraswamy J, Saxena S. Why to target G-quadruplexes using peptides: Next-generation G4-interacting ligands. J Pept Sci 2023; 29:e3491. [PMID: 37009771 DOI: 10.1002/psc.3491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/04/2023]
Abstract
Guanine-rich oligonucleotides existing in both DNA and RNA are able to fold into four-stranded DNA secondary structures via Hoogsteen type hydrogen-bonding, where four guanines self-assemble into a square planar arrangement, which, when stacked upon each other, results in the formation of higher-order structures called G-quadruplexes. Their distribution is not random; they are more frequently present at telomeres, proto-oncogenic promoters, introns, 5'- and 3'-untranslated regions, stem cell markers, ribosome binding sites and so forth and are associated with various biological functions, all of which play a pivotal role in various incurable diseases like cancer and cellular ageing. Several studies have suggested that G-quadruplexes could not regulate biological processes by themselves; instead, various proteins take part in this regulation and can be important therapeutic targets. There are certain limitations in using whole G4-protein for therapeutics purpose because of its high manufacturing cost, laborious structure prediction, dynamic nature, unavailability for oral administration due to its degradation in the gut and inefficient penetration to reach the target site because of the large size. Hence, biologically active peptides can be the potential candidates for therapeutic intervention instead of the whole G4-protein complex. In this review, we aimed to clarify the biological roles of G4s, how we can identify them throughout the genome via bioinformatics, the proteins interacting with G4s and how G4-interacting peptide molecules may be the potential next-generation ligands for targeting the G4 motifs located in biologically important regions.
Collapse
Affiliation(s)
- Taniya Sharma
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Nikita Kundu
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Sarvpreet Kaur
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Jadala Shankaraswamy
- Department of Fruit Science, College of Horticulture, Mojerla, Sri Konda Laxman Telangana State Horticultural University, Budwel, Telangana, India
| | - Sarika Saxena
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
15
|
Štepihar D, Florke Gee RR, Hoyos Sanchez MC, Fon Tacer K. Cell-specific secretory granule sorting mechanisms: the role of MAGEL2 and retromer in hypothalamic regulated secretion. Front Cell Dev Biol 2023; 11:1243038. [PMID: 37799273 PMCID: PMC10548473 DOI: 10.3389/fcell.2023.1243038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
Intracellular protein trafficking and sorting are extremely arduous in endocrine and neuroendocrine cells, which synthesize and secrete on-demand substantial quantities of proteins. To ensure that neuroendocrine secretion operates correctly, each step in the secretion pathways is tightly regulated and coordinated both spatially and temporally. At the trans-Golgi network (TGN), intrinsic structural features of proteins and several sorting mechanisms and distinct signals direct newly synthesized proteins into proper membrane vesicles that enter either constitutive or regulated secretion pathways. Furthermore, this anterograde transport is counterbalanced by retrograde transport, which not only maintains membrane homeostasis but also recycles various proteins that function in the sorting of secretory cargo, formation of transport intermediates, or retrieval of resident proteins of secretory organelles. The retromer complex recycles proteins from the endocytic pathway back to the plasma membrane or TGN and was recently identified as a critical player in regulated secretion in the hypothalamus. Furthermore, melanoma antigen protein L2 (MAGEL2) was discovered to act as a tissue-specific regulator of the retromer-dependent endosomal protein recycling pathway and, by doing so, ensures proper secretory granule formation and maturation. MAGEL2 is a mammalian-specific and maternally imprinted gene implicated in Prader-Willi and Schaaf-Yang neurodevelopmental syndromes. In this review, we will briefly discuss the current understanding of the regulated secretion pathway, encompassing anterograde and retrograde traffic. Although our understanding of the retrograde trafficking and sorting in regulated secretion is not yet complete, we will review recent insights into the molecular role of MAGEL2 in hypothalamic neuroendocrine secretion and how its dysregulation contributes to the symptoms of Prader-Willi and Schaaf-Yang patients. Given that the activation of many secreted proteins occurs after they enter secretory granules, modulation of the sorting efficiency in a tissue-specific manner may represent an evolutionary adaptation to environmental cues.
Collapse
Affiliation(s)
- Denis Štepihar
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX, United States
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Rebecca R. Florke Gee
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX, United States
| | - Maria Camila Hoyos Sanchez
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX, United States
| | - Klementina Fon Tacer
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, TX, United States
| |
Collapse
|
16
|
Characterization of IGF2R Molecular Expression in Canine Osteosarcoma as Part of a Novel Comparative Oncology Approach. Int J Mol Sci 2023; 24:ijms24031867. [PMID: 36768202 PMCID: PMC9916217 DOI: 10.3390/ijms24031867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Progress in prognostic factors, treatments, and outcome for both canine and human osteosarcoma (OS) has been minimal over the last three decades. Surface overexpression of the cation independent mannose-6-phosphate/insulin-like growth factor receptor type 2 (IGF2R) has been proven to occur in human OS cells. Subsequently, radioimmunotherapy (RIT) targeting IGF2R has demonstrated promising preliminary results. The main aims of this study were to investigate the expression of IGF2R in spontaneously occurring canine OS cells using immunohistochemistry (IHC) on archived biopsy samples and to assess its prognostic significance. Thirty-four dogs were included in the study. All cases showed that 80-100% of OS cells stained positive for IGF2R. IGF2R overexpression alone was not shown to have prognostic significance using both visual and quantitative methods of IHC staining intensity. This study has established for the first time the consistent expression of IGF2R in spontaneously occurring canine OS. This comparative oncology approach will allow further investigation into RIT as a novel treatment modality; first in canines and then in humans with OS. In addition, further studies should be performed to assess the true prognostic significance of IGF2R overexpression.
Collapse
|
17
|
Hossain MA, Al Ashik SA, Mahin MR, Al Amin M, Rahman MH, Khan MA, Emran AA. Systems biology and in silico-based analysis of PCOS revealed the risk of metabolic disorders. Heliyon 2022; 8:e12480. [PMID: 36619413 PMCID: PMC9816984 DOI: 10.1016/j.heliyon.2022.e12480] [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: 02/04/2022] [Revised: 07/18/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Background Polycystic ovarian syndrome (PCOS) is a common condition of hyperandrogenism, chronic ovulation, and polycystic ovaries in females during the reproduction and maturation of the ovum. Although PCOS has been associated with metabolic disorders, including type 2 diabetes (T2D), obesity (OBE), and cardiovascular disease (CVD), Causal connection and molecular features are still unknown. Purpose Therefore, we investigated the shared common differentially expressed genes (DEGs), pathways, and networks of associated proteins in PCOS and metabolic diseases with therapeutic intervention. Methods We have used a bioinformatics pipeline to analyze transcriptome data for the polycystic ovarian syndrome (PCOS), type 2 diabetes (T2D), obesity (OBE), and cardiovascular diseases (CVD) in female patients. Then we employed gene-disease association network, gene ontology (GO) and signaling pathway analysis, selection of hub genes from protein-protein interaction (PPI) network, molecular docking, and gold benchmarking approach to screen potential hub proteins. Result We discovered 2225 DEGs in PCOS patients relative to healthy controls and 34, 91, and 205 significant DEGs with T2D, Obesity, and CVD, respectively. Gene Ontology analysis revealed several significant shared and metabolic pathways from signaling pathway analysis. Furthermore, we identified ten potential hub proteins from PPI analysis that may serve as a therapeutic intervention in the future. Finally, we targeted one significant hub protein, IGF2R (PDB ID: 2V5O), out of ten hub proteins based on the Maximal clique centrality (MCC) algorithm and literature review for molecular docking study. Enzastaurin (-12.5), Kaempferol (-9.1), Quercetin (-9.0), and Coumestrol (-8.9) kcal/mol showed higher binding affinity in the molecular docking approach than 19 drug compounds. We have also found that the selected four compounds displayed favorable ADMET properties compared to the native ligand. Conclusion Our in-silico research findings identified a shared molecular etiology between PCOS and metabolic diseases that may suggest new therapeutic targets and warrants future experimental validation of the key targets.
Collapse
Affiliation(s)
- Md. Arju Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1092, Bangladesh
| | - Sheikh Abdullah Al Ashik
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1092, Bangladesh
| | - Moshiur Rahman Mahin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1092, Bangladesh
| | - Md. Al Amin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1092, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia, 7003, Bangladesh
| | - Md. Arif Khan
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1092, Bangladesh
- Department of Biotechnology and Genetic Engineering, University of Development Alternative, 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | - Abdullah Al Emran
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1092, Bangladesh
| |
Collapse
|
18
|
Venkatesan M, Semper C, Skrivergaard S, Di Leo R, Mesa N, Rasmussen MK, Young JF, Therkildsen M, Stogios PJ, Savchenko A. Recombinant production of growth factors for application in cell culture. iScience 2022; 25:105054. [PMID: 36157583 PMCID: PMC9489951 DOI: 10.1016/j.isci.2022.105054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/07/2022] [Accepted: 08/26/2022] [Indexed: 11/04/2022] Open
Abstract
Culturing eukaryotic cells has widespread applications in research and industry, including the emerging field of cell-cultured meat production colloquially referred to as “cellular agriculture”. These applications are often restricted by the high cost of growth medium necessary for cell growth. Mitogenic protein growth factors (GFs) are essential components of growth medium and account for upwards of 90% of the total costs. Here, we present a set of expression constructs and a simplified protocol for recombinant production of functionally active GFs, including FGF2, IGF1, PDGF-BB, and TGF-β1 in Escherichia coli. Using this E. coli expression system, we produced soluble GF orthologs from species including bovine, chicken, and salmon. Bioactivity analysis revealed orthologs with improved performance compared to commercially available alternatives. We estimated that the production cost of GFs using our methodology will significantly reduce the cost of cell culture medium, facilitating low-cost protocols tailored for cultured meat production and tissue engineering. Developed methodology for low-cost production of soluble, bioactive GFs Purified GFs were active on NIH-3T3 and bovine satellite cells Some GF orthologs outperformed commercially sourced GFs Production of GFs using these methods can foster significant cost savings
Collapse
Affiliation(s)
- Meenakshi Venkatesan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Cameron Semper
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Rosa Di Leo
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Nathalie Mesa
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | | | | | | | - Peter J Stogios
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada
| | - Alexei Savchenko
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON M5S 3E8, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
19
|
Neville MC, Webb PG, Baumgartner HK, Bitler BG. Claudin-4 localization in epithelial ovarian cancer. Heliyon 2022; 8:e10862. [PMID: 36237976 PMCID: PMC9552118 DOI: 10.1016/j.heliyon.2022.e10862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/06/2022] [Accepted: 09/28/2022] [Indexed: 11/19/2022] Open
Abstract
Claudin-4, a protein with the structure of classic claudins most often found in cell-cell junctions, is frequently overexpressed in epithelial cancers where its localization has not been studied. In this study we aimed to find out where this membrane protein is localized in an ovarian tumor model, OVCAR3 cells, that express high levels of the protein. Immunohistochemical studies showed claudin-4 staining in a perinuclear region, at most plasma membranes and in cytoplasmic puncta. Native claudin-4 did not overlap with phosphorylated claudin-4, which was partially located in focal adhesions. Using claudin-4 BioID technology we confirmed that large amounts of claudin-4 are localized to the Golgi compartment, including in dispersed Golgi in cells where claudin-4 is partially knocked down and in dividing cells. Claudin-4 appears to be present in the vicinity of several types of cell-cell junctions, but there is no evidence that it forms tight junctions in these tumor cells. Both claudin-4, the Golgi marker GM130, and the plasma membrane receptor Notch2 were found in dispersed Golgi in dividing cells. This definition of the cellular architecture of claudin-4 should provide a framework for better understanding of the function of claudin-4 in tumor cells and its molecular interactions.
Collapse
Affiliation(s)
- Margaret C. Neville
- Departments of Obstetrics and Gynecology and Physiology and Biophysics, University of Colorado School of Medicine, Aurora, CO, 80845, USA
- Corresponding author.
| | - Patricia G. Webb
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80845, USA
| | - Heidi K. Baumgartner
- University of Colorado Anschutz Medical Campus, 2700 E. 19th Ave., Aurora, CO, 80045, USA
| | - Benjamin G. Bitler
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado Denver Anschutz Medical Campus, Mail Stop 8613, 12700 E. 19 Ave., Aurora, CO, 80045, USA
| |
Collapse
|
20
|
IGF2: Development, Genetic and Epigenetic Abnormalities. Cells 2022; 11:cells11121886. [PMID: 35741015 PMCID: PMC9221339 DOI: 10.3390/cells11121886] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 02/07/2023] Open
Abstract
In the 30 years since the first report of parental imprinting in insulin-like growth factor 2 (Igf2) knockout mouse models, we have learnt much about the structure of this protein, its role and regulation. Indeed, many animal and human studies involving innovative techniques have shed light on the complex regulation of IGF2 expression. The physiological roles of IGF-II have also been documented, revealing pleiotropic tissue-specific and developmental-stage-dependent action. Furthermore, in recent years, animal studies have highlighted important interspecies differences in IGF-II function, gene expression and regulation. The identification of human disorders due to impaired IGF2 gene expression has also helped to elucidate the major role of IGF-II in growth and in tumor proliferation. The Silver-Russell and Beckwith-Wiedemann syndromes are the most representative imprinted disorders, as they constitute both phenotypic and molecular mirrors of IGF2-linked abnormalities. The characterization of patients with either epigenetic or genetic defects altering IGF2 expression has confirmed the central role of IGF-II in human growth regulation, particularly before birth, and its effects on broader body functions, such as metabolism or tumor susceptibility. Given the long-term health impact of these rare disorders, it is important to understand the consequences of IGF2 defects in these patients.
Collapse
|
21
|
Abstract
The functional mass of kidney tissue in an adult is an important determinant of human health. Kidney formation during development is an essential determinant of the final nephron endowment of the adult organ, and no evidence has been reported that mice or humans are able to generate new nephrons after the developmental period. Mechanisms controlling organ growth after development are essential to establish the final adult organ size. The potential for organ growth is maintained in adult life and the size of one kidney may be significantly increased by loss of the contralateral kidney. The mouse has provided a model system for investigators to critically explore genetic, cell biological, and hormonal control of developmental and juvenile kidney growth. This article reviews three basic aspects of kidney size regulation: (1) Mechanisms that control nephron formation and how these are altered by the cessation of nephrogenesis at the end of the developmental period. (2) Applicability of the general model for growth hormone-insulin like growth factor control to kidney growth both pre- and postnatally. (3) Commonalities between mechanisms of juvenile kidney growth and the compensatory growth that is stimulated in adult life by reduction of kidney mass. Understanding the mechanisms that determine set-points for cell numbers and size in the kidney may inform ongoing efforts to generate kidney tissue from stem cells.
Collapse
Affiliation(s)
- Leif Oxburgh
- The Rogosin Institute, New York, NY, United States.
| |
Collapse
|
22
|
Tran V, O’Neill HC. Role of SVEP1 in Stroma-Dependent Hematopoiesis In vitro. Front Cell Dev Biol 2022; 9:760480. [PMID: 35174156 PMCID: PMC8841349 DOI: 10.3389/fcell.2021.760480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
Study of the microenvironment that supports hematopoietic stem cell (HSC) development in vivo is very difficult involving small numbers of interacting cells which are usually not well defined. While much is known about HSC niches located within the bone marrow in terms of contributing cell types and signalling molecules, very little is known about equivalent niches within spleen. Extramedullary hematopoiesis in spleen contributes myeloid cells important in the mobilisation of an immune response. As a result, it is important to develop in vitro models to identify the cells which constitute HSC niches in spleen and to identify the regulatory molecules supporting myeloid cell development. Studies described here document a model system to study the maintenance and differentiation of HSC by splenic stromal cells in vitro. The splenic stromal lines 5G3 and 3B5 differ in hematopoietic support capacity. SVEP1 and IGF2 are molecules of interest specifically expressed by 5G3 stroma. Gene knockdown technology using shRNA plasmids has been used to reduce gene expression in 5G3 and to determine specific effects on myeloid cell development following co-culture with overlaid hematopoietic progenitors in vitro. Knockdown of Svep1 gave specific inhibition of a dendritic cell (DC) population described previously in spleen (L-DC). Knockdown of Igf2 resulted in loss of production of a minor subset of conventional (c) DC. SVEP1 is now considered a marker of mesenchymal stromal cells with osteogenic differentiative capacity reflective of perivascular stromal cells. The power of this in vitro model is evidenced by the fact that it has been used to define SVEP1 as a specific adhesion molecule that regulates the hematopoietic process dependent on stromal niche interaction. The identification of stromal cells and molecules that contribute to the hematopoietic process in spleen, brings us closer to the realm of therapeutically regulating hematopoiesis in vivo, and to inhibiting niches which support cancer stem cells.
Collapse
Affiliation(s)
- Vinson Tran
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Helen C. O’Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
- *Correspondence: Helen C. O’Neill,
| |
Collapse
|
23
|
Ferrao Blanco MN, Domenech Garcia H, Legeai-Mallet L, van Osch GJVM. Tyrosine kinases regulate chondrocyte hypertrophy: promising drug targets for Osteoarthritis. Osteoarthritis Cartilage 2021; 29:1389-1398. [PMID: 34284112 DOI: 10.1016/j.joca.2021.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.
Collapse
Affiliation(s)
- M N Ferrao Blanco
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - H Domenech Garcia
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - L Legeai-Mallet
- Université de Paris, INSERM U1163, Institut Imagine, Paris, France.
| | - G J V M van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
24
|
Weber F, Casalini T, Valentino G, Brülisauer L, Andreas N, Koeberle A, Kamradt T, Contini A, Luciani P. Targeting transdifferentiated hepatic stellate cells and monitoring the hepatic fibrogenic process by means of IGF2R-specific peptides designed in silico. J Mater Chem B 2021; 9:2092-2106. [PMID: 33595041 DOI: 10.1039/d0tb02372h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The lack of accurate and easily applicable methods for the diagnosis of liver fibrosis, a disease characterized by an accumulation of the extracellular matrix released by activated hepatic stellate cells (HSCs), has been a major limitation for the clinical management of liver diseases. The identification of biomarkers specific to liver microstructure alterations, combined with a non-invasive optical imaging modality, could guide clinicians towards a therapeutic strategy. In this study, structural information of the insulin-like growth factor 2 receptor (IGF2R), an overexpressed protein on activated HSCs, was used for in silico screening of novel IGF2R-specific peptide ligands. Molecular dynamics simulations, followed by computational alanine scanning of the IGF2R/IGF2 complex, led to the identification of a putative peptide sequence containing the most relevant amino acids for the receptor-ligand interaction (IGF2 E12-C21). The Residue Scan tool, implemented in the MOE software, was then used to optimize the binding affinity of this sequence by amino acid mutations. The designed peptides and their associated scrambled sequences were fluorescently labelled and their binding affinity to LX-2 cells (model for activated human HSCs) was tested using flow cytometry and confocal microscopy. In vitro binding was verified for all sequences (KD ≤ 13.2 μM). With respect to the putative binding sequence, most mutations led to an increased affinity. All sequences have shown superior binding compared to their associated scrambled sequences. Using HPLC, all peptides were tested in vitro for their proteolytic resistance and showed a stability of ≥60% intact after 24 h at 37 °C in 50% v/v FBS. In view of their prospective diagnostic application, a comparison of binding affinity was performed in perpetuated and quiescent-like LX-2 cells. Furthermore, the IGF2R expression for different cell phenotypes was analysed by a quantitative mass spectrometric approach. Our peptides showed increased binding to the perpetuated cell state, indicating their good selectivity for the diagnostically relevant phenotype. In summary, the increased binding affinity of our peptides towards perpetuated LX-2 cells, as well as the satisfactory proteolytic stability, proves that the in silico designed sequences offer a new potential strategy for the targeting of hepatic fibrosis.
Collapse
Affiliation(s)
- Florian Weber
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland. and Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Tommaso Casalini
- Institute of Mechanical Engineering and Material Technology, Department of Innovative Technology, SUPSI, Manno, Switzerland and Institute for Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Gina Valentino
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland. and Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Lorine Brülisauer
- Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Nico Andreas
- Institute of Immunology, Jena University Hospital, Jena, Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria and Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Thomas Kamradt
- Institute of Immunology, Jena University Hospital, Jena, Germany
| | - Alessandro Contini
- Dipartimento di Scienze Farmaceutiche-Sezione di Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milano, Italy
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland. and Department of Pharmaceutical Technology, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
25
|
Novel Human Antibodies to Insulin Growth Factor 2 Receptor (IGF2R) for Radioimmunoimaging and Therapy of Canine and Human Osteosarcoma. Cancers (Basel) 2021; 13:cancers13092208. [PMID: 34064450 PMCID: PMC8124616 DOI: 10.3390/cancers13092208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Osteosarcoma (OS) is the most common type of bone cancer and mainly affects children, teens and young adults. The overall survival rate is ~67%, but patients with distant metastases have poor prognosis. Insulin growth factor 2 receptor (IGF2R) is a protein that has been shown to be expressed widely in human patient-derived OS cells and is a suitable for target for monoclonal antibody-based therapies. Given the similarities between canine and human OS, IGF2R is also overexpressed in canine OS. Towards the goal of one-health approach, we generated human antibodies that bind with similar affinities to IGF2R expressed in human, murine and canine tissues. We demonstrate tumor accumulation of radiolabeled antibodies in mice bearing human and canine patients derived tumors. Therefore, these antibodies show promise for development into the agents for radioimmunoimaging and radioimmunotherapy of OS in human and canine patients. Abstract Etiological and genetic drivers of osteosarcoma (OS) are not well studied and vary from one tumor to another; making it challenging to pursue conventional targeted therapy. Recent studies have shown that cation independent mannose-6-phosphate/insulin-like growth factor-2 receptor (IGF2R) is consistently overexpressed in almost all of standard and patient-derived OS cell lines, making it an ideal therapeutic target for development of antibody-based drugs. Monoclonal antibodies, targeting IGF2R, can be conjugated with alpha- or beta-emitter radionuclides to deliver cytocidal doses of radiation to target IGF2R expression in OS. This approach known as radioimmunotherapy (RIT) can therefore be developed as a novel treatment for OS. In addition, OS is one of the common cancers in companion dogs and very closely resembles human OS in clinical presentation and molecular aberrations. In this study, we have developed human antibodies that cross-react with similar affinities to IGF2R proteins of human, canine and murine origin. We used naïve and synthetic antibody Fab-format phage display libraries to develop antibodies to a conserved region on IGF2R. The generated antibodies were radiolabeled and characterized in vitro and in vivo using human and canine OS patient-derived tumors in SCID mouse models. We demonstrate specific binding to IGF2R and tumor uptake in these models, as well as binding to tumor tissue of canine OS patients, making these antibodies suitable for further development of RIT for OS
Collapse
|
26
|
Gully BS, Venugopal H, Fulcher AJ, Fu Z, Li J, Deuss FA, Llerena C, Heath WR, Lahoud MH, Caminschi I, Rossjohn J, Berry R. The cryo-EM structure of the endocytic receptor DEC-205. J Biol Chem 2020; 296:100127. [PMID: 33257321 PMCID: PMC7948739 DOI: 10.1074/jbc.ra120.016451] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 11/06/2022] Open
Abstract
DEC-205 (CD205), a member of the macrophage mannose receptor protein family, is the prototypic endocytic receptor of dendritic cells, whose ligands include phosphorothioated cytosine-guanosine oligonucleotides, a motif often seen in bacterial or viral DNA. However, despite growing biological and clinical significance, little is known about the structural arrangement of this receptor or any of its family members. Here, we describe the 3.2 Å cryo-EM structure of human DEC-205, thereby illuminating the structure of the mannose receptor protein family. The DEC-205 monomer forms a compact structure comprising two intercalated rings of C-type lectin-like domains, where the N-terminal cysteine-rich and fibronectin domains reside at the central intersection. We establish a pH-dependent oligomerization pathway forming tetrameric DEC-205 using solution-based techniques and ultimately solved the 4.9 Å cryo-EM structure of the DEC-205 tetramer to identify the unfurling of the second lectin ring which enables tetramer formation. Furthermore, we suggest the relevance of this oligomerization pathway within a cellular setting, whereby cytosine-guanosine binding appeared to disrupt this cell-surface oligomer. Accordingly, we provide insight into the structure and oligomeric assembly of the DEC-205 receptor.
Collapse
Affiliation(s)
- Benjamin S Gully
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.
| | - Hariprasad Venugopal
- Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Victoria, Australia
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, Victoria, Australia
| | - Zhihui Fu
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jessica Li
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Felix A Deuss
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Carmen Llerena
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Mireille H Lahoud
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Irina Caminschi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom.
| | - Richard Berry
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
27
|
Understanding IGF-II Action through Insights into Receptor Binding and Activation. Cells 2020; 9:cells9102276. [PMID: 33053840 PMCID: PMC7601145 DOI: 10.3390/cells9102276] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023] Open
Abstract
The insulin-like growth factor (IGF) system regulates metabolic and mitogenic signaling through an intricate network of related receptors and hormones. IGF-II is one of several hormones within this system that primarily regulates mitogenic functions and is especially important during fetal growth and development. IGF-II is also found to be overexpressed in several cancer types, promoting growth and survival. It is also unique in the IGF system as it acts through both IGF-1R and insulin receptor isoform A (IR-A). Despite this, IGF-II is the least investigated ligand of the IGF system. This review will explore recent developments in IGF-II research including a structure of IGF-II bound to IGF-1R determined using cryo-electron microscopy (cryoEM). Comparisons are made with the structures of insulin and IGF-I bound to their cognate receptors. Finally discussed are outstanding questions in the mechanism of action of IGF-II with the goal of developing antagonists of IGF action in cancer.
Collapse
|
28
|
Zheng Y, Sun Q, Xu X, Wang W. Novel peptide derived from IGF-2 displays anti-angiogenic activity in vitro and inhibits retinal angiogenesis in a model of oxygen-induced retinopathy. Clin Exp Ophthalmol 2020; 48:1261-1275. [PMID: 33026147 DOI: 10.1111/ceo.13864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Retinopathy of prematurity (ROP), a major cause of significant visual morbidity and blindness in preterm infants, is closely related to pathological angiogenesis. The aim of the study is to evaluate the effect of a new 12-aa peptide (named peptide CW-703) from human insulin-like growth factor-2, against angiogenesis in ROP. METHODS In order to evaluate the inhibitory effect of CW-703 on the proliferation, migration, tube formation and apoptosis of human umbilical vein endothelial cells (ScienCell) in vitro, we used MTS assays, a modified Boyden chamber, Matrigel system and TUNEL assays. Effects in vivo were assayed using chorioallantoic membrane assays and oxygen-induced retinopathy (OIR) models in mice. We also performed eletrophysiological and histologic examinations to evaluate the possible toxicity of the peptide. Real-time PCR, ELISA and western blotting were used to elucidate the mechanism of CW-703. RESULTS CW-703 inhibited angiogenesis in vitro by suppressing endothelial cell proliferation, migration and tube formation. CW-703 also prevented angiogenesis in chicken chorioallantoic membrane assays and OIR assays in mice. No evident functional or morphologic abnormalities in neuroretina after CW-703 injection were revealed in electrophysiological tests and histological examinations. Moreover, we elucidated that CW-703 competed for binding to IGF-1R and inhibited angiogenesis by inhibiting IGF-1R/PI3K/AKT activation and downregulating vascular endothelial growth factor expression. CONCLUSION The novel peptide CW-703 may act as an effective inhibitor of ocular pathologic angiogenesis, especially in treating ROP.
Collapse
Affiliation(s)
- Ying Zheng
- Department of Ophthalmology, Shanghai General Hospital affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Qiao Sun
- Department of Ophthalmology, Shanghai General Hospital affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Weijun Wang
- Department of Ophthalmology, Shanghai General Hospital affiliated to Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
29
|
Olson LJ, Misra SK, Ishihara M, Battaile KP, Grant OC, Sood A, Woods RJ, Kim JJP, Tiemeyer M, Ren G, Sharp JS, Dahms NM. Allosteric regulation of lysosomal enzyme recognition by the cation-independent mannose 6-phosphate receptor. Commun Biol 2020; 3:498. [PMID: 32908216 PMCID: PMC7481795 DOI: 10.1038/s42003-020-01211-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/11/2020] [Indexed: 01/09/2023] Open
Abstract
The cation-independent mannose 6-phosphate receptor (CI-MPR, IGF2 receptor or CD222), is a multifunctional glycoprotein required for normal development. Through the receptor's ability to bind unrelated extracellular and intracellular ligands, it participates in numerous functions including protein trafficking, lysosomal biogenesis, and regulation of cell growth. Clinically, endogenous CI-MPR delivers infused recombinant enzymes to lysosomes in the treatment of lysosomal storage diseases. Although four of the 15 domains comprising CI-MPR's extracellular region bind phosphorylated glycans on lysosomal enzymes, knowledge of how CI-MPR interacts with ~60 different lysosomal enzymes is limited. Here, we show by electron microscopy and hydroxyl radical protein footprinting that the N-terminal region of CI-MPR undergoes dynamic conformational changes as a consequence of ligand binding and different pH conditions. These data, coupled with X-ray crystallography, surface plasmon resonance and molecular modeling, allow us to propose a model explaining how high-affinity carbohydrate binding is achieved through allosteric domain cooperativity.
Collapse
Affiliation(s)
- Linda J Olson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| | - Sandeep K Misra
- Department of BioMolecular Sciences, University of Mississippi, Oxford, MS, 38677, USA
| | - Mayumi Ishihara
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Kevin P Battaile
- IMCA-CAT, Hauptman-Woodward Medical Research Institute, Argonne, IL, USA
- New York Structural Biology Center, New York City, NY, 10027, USA
| | - Oliver C Grant
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Amika Sood
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Robert J Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Jung-Ja P Kim
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Joshua S Sharp
- Department of BioMolecular Sciences, University of Mississippi, Oxford, MS, 38677, USA
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
30
|
Potalitsyn P, Selicharová I, Sršeň K, Radosavljević J, Marek A, Nováková K, Jiráček J, Žáková L. A radioligand binding assay for the insulin-like growth factor 2 receptor. PLoS One 2020; 15:e0238393. [PMID: 32877466 PMCID: PMC7467306 DOI: 10.1371/journal.pone.0238393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/14/2020] [Indexed: 01/04/2023] Open
Abstract
Insulin-like growth factors 2 and 1 (IGF2 and IGF1) and insulin are closely related hormones that are responsible for the regulation of metabolic homeostasis, development and growth of the organism. Physiological functions of insulin and IGF1 are relatively well-studied, but information about the role of IGF2 in the body is still sparse. Recent discoveries called attention to emerging functions of IGF2 in the brain, where it could be involved in processes of learning and memory consolidation. It was also proposed that these functions could be mediated by the receptor for IGF2 (IGF2R). Nevertheless, little is known about the mechanism of signal transduction through this receptor. Here we produced His-tagged domain 11 (D11), an IGF2-binding element of IGF2R; we immobilized it on the solid support through a well-defined sandwich, consisting of neutravidin, biotin and synthetic anti-His-tag antibodies. Next, we prepared specifically radiolabeled [125I]-monoiodotyrosyl-Tyr2-IGF2 and optimized a sensitive and robust competitive radioligand binding assay for determination of the nanomolar binding affinities of hormones for D11 of IGF2. The assay will be helpful for the characterization of new IGF2 mutants to study the functions of IGF2R and the development of new compounds for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Pavlo Potalitsyn
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Irena Selicharová
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Kryštof Sršeň
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Jelena Radosavljević
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Marek
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Kateřina Nováková
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Jiráček
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| | - Lenka Žáková
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
31
|
Bochel AJ, Williams C, McCoy AJ, Hoppe HJ, Winter AJ, Nicholls RD, Harlos K, Jones EY, Berger I, Hassan AB, Crump MP. Structure of the Human Cation-Independent Mannose 6-Phosphate/IGF2 Receptor Domains 7-11 Uncovers the Mannose 6-Phosphate Binding Site of Domain 9. Structure 2020; 28:1300-1312.e5. [PMID: 32877646 DOI: 10.1016/j.str.2020.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/13/2020] [Accepted: 08/07/2020] [Indexed: 11/29/2022]
Abstract
The cation-independent mannose 6-phosphate (M6P)/Insulin-like growth factor-2 receptor (CI-MPR/IGF2R) is an ∼300 kDa transmembrane protein responsible for trafficking M6P-tagged lysosomal hydrolases and internalizing IGF2. The extracellular region of the CI-MPR has 15 homologous domains, including M6P-binding domains (D) 3, 5, 9, and 15 and IGF2-binding domain 11. We have focused on solving the first structures of human D7-10 within two multi-domain constructs, D9-10 and D7-11, and provide the first high-resolution description of the high-affinity M6P-binding D9. Moreover, D9 stabilizes a well-defined hub formed by D7-11 whereby two penta-domains intertwine to form a dimeric helical-type coil via an N-glycan bridge on D9. Remarkably the D7-11 structure matches an IGF2-bound state of the receptor, suggesting this may be an intrinsically stable conformation at neutral pH. Interdomain clusters of histidine and proline residues may impart receptor rigidity and play a role in structural transitions at low pH.
Collapse
Affiliation(s)
- Alice J Bochel
- School of Chemistry, Cantock's Close, University of Bristol, Bristol BS8 1TS, UK
| | - Christopher Williams
- School of Chemistry, Cantock's Close, University of Bristol, Bristol BS8 1TS, UK; BrisSynBio, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK
| | - Airlie J McCoy
- Cambridge Institute for Medical Research, Department of Haematology, University of Cambridge, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| | - Hans-Jürgen Hoppe
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK; dAInomics Ltd, 66 High Street, Bassingbourn Royston SG8 5LF, UK
| | - Ashley J Winter
- School of Chemistry, Cantock's Close, University of Bristol, Bristol BS8 1TS, UK
| | - Ryan D Nicholls
- School of Chemistry, Cantock's Close, University of Bristol, Bristol BS8 1TS, UK
| | - Karl Harlos
- Cancer Research UK Receptor Structure Research Group, Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - E Yvonne Jones
- Cancer Research UK Receptor Structure Research Group, Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Imre Berger
- School of Biochemistry, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - A Bassim Hassan
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK.
| | - Matthew P Crump
- School of Chemistry, Cantock's Close, University of Bristol, Bristol BS8 1TS, UK; BrisSynBio, Life Sciences Building, Tyndall Avenue, Bristol BS8 1TQ, UK.
| |
Collapse
|
32
|
Wang R, Qi X, Schmiege P, Coutavas E, Li X. Marked structural rearrangement of mannose 6-phosphate/IGF2 receptor at different pH environments. SCIENCE ADVANCES 2020; 6:eaaz1466. [PMID: 32095534 PMCID: PMC7015683 DOI: 10.1126/sciadv.aaz1466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/02/2019] [Indexed: 05/08/2023]
Abstract
Many cell surface receptors internalize their ligands and deliver them to endosomes, where the acidic pH causes the ligand to dissociate. The liberated receptor returns to the cell surface in a process called receptor cycling. The structural basis for pH-dependent ligand dissociation is not well understood. In some receptors, the ligand binding domain is composed of multiple repeated sequences. The insulin-like growth factor 2 receptor (IGF2R) contains 15 β strand-rich repeat domains. The overall structure and the mechanism by which IGF2R binds IGF2 and releases it are unknown. We used cryo-EM to determine the structures of the IGF2R at pH 7.4 with IGF2 bound and at pH 4.5 in the ligand-dissociated state. The results reveal different arrangements of the receptor in different pH environments mediated by changes in the interactions between the repeated sequences. These results have implications for our understanding of ligand release from receptors in endocytic compartments.
Collapse
Affiliation(s)
- Rong Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaofeng Qi
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philip Schmiege
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elias Coutavas
- Laboratory of Cell Biology, The Rockefeller University, New York, NY 10065, USA
| | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Corresponding author.
| |
Collapse
|
33
|
Jafari E, Gheysarzadeh A, Mahnam K, Shahmohammadi R, Ansari A, Bakhtyari H, Mofid MR. In silico interaction of insulin-like growth factor binding protein 3 with insulin-like growth factor 1. Res Pharm Sci 2018; 13:332-342. [PMID: 30065766 PMCID: PMC6040160 DOI: 10.4103/1735-5362.235160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP-3) is a vital protein exist in circulation which interacts with high affinity to insulin-like growth factor (IGFs) altering their activities. Therefore, the interaction between IGFs and IGFBP-3 has a key role altering large spectrum of activities such as cell cycle progression, proliferation and apoptosis. Despite decades of research, the crystal structure of IGFBP-3 has not been identified possibly due to some technical challenge in its crystallizing. The three-dimensional (3D) structure of IGFBP-3 was predicted using homology modeling, Phyre2, and molecular dynamic. Its interaction with IGF-1 was also identified by HADDOCK software. IGFBP-3 has the most identity with other IGFBPs in N and C-domain; however, its linker domain has lower identity. Our data predicted that IGF-1 structurally interacts with N-domain and linker domain of IGFBP-3. Some conserved residues of IGFBP-3 such as Glu33, Arg36, Gly39, Arg60, Arg66, Asn109, and Ile146 interacts with Glu3, Asp12, Phe16, Gly19, Asp20, Arg21, and Glu58 of IGF-1. In addition, our data predict that the linker domain has a loop structure which covers post translational modification and interacts with IGF-1. The phosphorylation of Ser111 in linker domain, which previously has been shown to induce apoptosis make a repulsive force interrupting this interaction to IGF-1, which enables IGFBP-3 to induce apoptosis. The present study suggests that the linker domain has a key role in recognition of IGFBP-3 with IGF-1.
Collapse
Affiliation(s)
- Elham Jafari
- Department of Medicinal Chemistry, Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Ali Gheysarzadeh
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Karim Mahnam
- Biology Department, Faculty of Sciences, Shahrekord University, Shahrekord, I.R. Iran
| | | | - Amir Ansari
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Hadi Bakhtyari
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Reza Mofid
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
34
|
Takifugu rubripes cation independent mannose 6-phosphate receptor: Cloning, expression and functional characterization of the IGF-II binding domain. Int J Biol Macromol 2018; 113:59-65. [DOI: 10.1016/j.ijbiomac.2018.01.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 01/30/2018] [Indexed: 11/22/2022]
|
35
|
Wang K, Wu P, Yang Q, Chen D, Zhou J, Jiang A, Ma J, Tang Q, Xiao W, Jiang Y, Zhu L, Li X, Tang G. Detection of Selection Signatures in Chinese Landrace and Yorkshire Pigs Based on Genotyping-by-Sequencing Data. Front Genet 2018; 9:119. [PMID: 29686696 PMCID: PMC5900008 DOI: 10.3389/fgene.2018.00119] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
The domestic pigs have been undergone intense selection pressures for these development of interested traits following domestication and modern breeding. This has altered many traits in most of pig breeds, such as growth rate, body weight, fertility, and immunity. Thus, the objectives of this study were to (1) detect these selection signatures and identify the candidate genes that show evidences of recent artificial selection at the level of whole genome, (2) be beneficial to understand the relationship between genomic structure and phenotypic diversity, and (3) highlight the key roles of these candidate genes in growth and development in the two breeds. The data consisted of total raw number of 345570 single nucleotide polymorphisms (SNPs) in 1200 individuals from the Chinese Landrace pigs (L, n = 600) and Yorkshire pigs (Y, n = 600). Based on these SNPs data, two complementary methods, population differentiation (Fst) and composite likelihood ratio test (CLR), were carried out to detect the selection signatures in this study. A total of 540 potential selection regions (50 kb) which contained 111 candidate genes were detected for Landrace-Yorkshire pair (L-Y) by Fst. In addition, 73 and 125 candidate genes were found for Landrace pigs and Yorkshire pigs by CLR test based on 321 and 628 potential selection regions, respectively. Some candidate genes are associated with important traits and signaling pathways including the ACACA, MECR, COL11A1, GHR, IGF1R, IGF2R, IFNG, and MTOR gene. The ACACA and MECR gene are related to fatty acid biosynthesis. The COL11A1 gene is essential for the development of the normal differentiation. The GHR, IGF1R, and IGF2R gene are significant candidate genes which play major roles in the growth and development in animals. The IFNG gene is associated with some aspects of immune response. The MTOR gene regulates many signaling pathways and signaling transduction pathway.
Collapse
Affiliation(s)
- Kai Wang
- *Correspondence: Kai Wang, Guoqing Tang,
| | | | | | | | | | | | | | | | | | | | | | | | - Guoqing Tang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
36
|
Xu Y, Kong GKW, Menting JG, Margetts MB, Delaine CA, Jenkin LM, Kiselyov VV, De Meyts P, Forbes BE, Lawrence MC. How ligand binds to the type 1 insulin-like growth factor receptor. Nat Commun 2018; 9:821. [PMID: 29483580 PMCID: PMC5826941 DOI: 10.1038/s41467-018-03219-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/29/2018] [Indexed: 12/28/2022] Open
Abstract
Human type 1 insulin-like growth factor receptor is a homodimeric receptor tyrosine kinase that signals into pathways directing normal cellular growth, differentiation and proliferation, with aberrant signalling implicated in cancer. Insulin-like growth factor binding is understood to relax conformational restraints within the homodimer, initiating transphosphorylation of the tyrosine kinase domains. However, no three-dimensional structures exist for the receptor ectodomain to inform atomic-level understanding of these events. Here, we present crystal structures of the ectodomain in apo form and in complex with insulin-like growth factor I, the latter obtained by crystal soaking. These structures not only provide a wealth of detail of the growth factor interaction with the receptor’s primary ligand-binding site but also indicate that ligand binding separates receptor domains by a mechanism of induced fit. Our findings are of importance to the design of agents targeting IGF-1R and its partner protein, the human insulin receptor. The human type 1 insulin-like growth factor receptor (IGF-1R) is important for normal human growth and development. Here, the authors present the crystal structures of the IGF-1R ectodomain both in its apo form and in complex with its ligand insulin-like growth factor I and discuss the receptor activation mechanism.
Collapse
Affiliation(s)
- Yibin Xu
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia
| | - Geoffrey K-W Kong
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Monash Macromolecular Crystallisation Facility, 11 Chancellors Walk, Clayton Campus, Monash University, Clayton, VIC, 3800, Australia
| | - John G Menting
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Mai B Margetts
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Carlie A Delaine
- Discipline of Medical Biochemistry, College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, SA, 5042, Australia
| | - Lauren M Jenkin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Vladislav V Kiselyov
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Pierre De Meyts
- Department of Cell Signaling, de Duve Institute, B-1200, Brussels, Belgium.,Department of Stem Cell Research, Novo Nordisk A/S, 2760, Måløv, Denmark.,De Meyts R&D Consulting, Avenue Reine Astrid 42, B-1950, Kraainem, Belgium
| | - Briony E Forbes
- Discipline of Medical Biochemistry, College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, SA, 5042, Australia
| | - Michael C Lawrence
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Royal Parade, Parkville, VIC, 3050, Australia.
| |
Collapse
|
37
|
Vishwamitra D, George SK, Shi P, Kaseb AO, Amin HM. Type I insulin-like growth factor receptor signaling in hematological malignancies. Oncotarget 2018; 8:1814-1844. [PMID: 27661006 PMCID: PMC5352101 DOI: 10.18632/oncotarget.12123] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022] Open
Abstract
The insulin-like growth factor (IGF) signaling system plays key roles in the establishment and progression of different types of cancer. In agreement with this idea, substantial evidence has shown that the type I IGF receptor (IGF-IR) and its primary ligand IGF-I are important for maintaining the survival of malignant cells of hematopoietic origin. In this review, we discuss current understanding of the role of IGF-IR signaling in cancer with a focus on the hematological neoplasms. We also address the emergence of IGF-IR as a potential therapeutic target for the treatment of different types of cancer including plasma cell myeloma, leukemia, and lymphoma.
Collapse
Affiliation(s)
- Deeksha Vishwamitra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suraj Konnath George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hesham M Amin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
38
|
Glidden MD, Aldabbagh K, Phillips NB, Carr K, Chen YS, Whittaker J, Phillips M, Wickramasinghe NP, Rege N, Swain M, Peng Y, Yang Y, Lawrence MC, Yee VC, Ismail-Beigi F, Weiss MA. An ultra-stable single-chain insulin analog resists thermal inactivation and exhibits biological signaling duration equivalent to the native protein. J Biol Chem 2017; 293:47-68. [PMID: 29114035 DOI: 10.1074/jbc.m117.808626] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/12/2017] [Indexed: 12/12/2022] Open
Abstract
Thermal degradation of insulin complicates its delivery and use. Previous efforts to engineer ultra-stable analogs were confounded by prolonged cellular signaling in vivo, of unclear safety and complicating mealtime therapy. We therefore sought an ultra-stable analog whose potency and duration of action on intravenous bolus injection in diabetic rats are indistinguishable from wild-type (WT) insulin. Here, we describe the structure, function, and stability of such an analog, a 57-residue single-chain insulin (SCI) with multiple acidic substitutions. Cell-based studies revealed native-like signaling properties with negligible mitogenic activity. Its crystal structure, determined as a novel zinc-free hexamer at 2.8 Å, revealed a native insulin fold with incomplete or absent electron density in the C domain; complementary NMR studies are described in the accompanying article. The stability of the analog (ΔGU 5.0(±0.1) kcal/mol at 25 °C) was greater than that of WT insulin (3.3(±0.1) kcal/mol). On gentle agitation, the SCI retained full activity for >140 days at 45 °C and >48 h at 75 °C. These findings indicate that marked resistance to thermal inactivation in vitro is compatible with native duration of activity in vivo Further, whereas WT insulin forms large and heterogeneous aggregates above the standard 0.6 mm pharmaceutical strength, perturbing the pharmacokinetic properties of concentrated formulations, dynamic light scattering, and size-exclusion chromatography revealed only limited SCI self-assembly and aggregation in the concentration range 1-7 mm Such a combination of favorable biophysical and biological properties suggests that SCIs could provide a global therapeutic platform without a cold chain.
Collapse
Affiliation(s)
- Michael D Glidden
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106
| | - Khadijah Aldabbagh
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Nelson B Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kelley Carr
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yen-Shan Chen
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Jonathan Whittaker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Manijeh Phillips
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | | | - Nischay Rege
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Mamuni Swain
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yi Peng
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106
| | - Yanwu Yang
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael C Lawrence
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vivien C Yee
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Faramarz Ismail-Beigi
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Michael A Weiss
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106; Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
39
|
Poondla V, Chikati R, Kallubai M, Chennupati V, Subramanyam R, Obulam VSR. Characterization and molecular modeling of polygalacturonase isoforms from Saccharomyces cerevisiae. 3 Biotech 2017; 7:285. [PMID: 28828292 DOI: 10.1007/s13205-017-0912-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 08/02/2017] [Indexed: 10/19/2022] Open
Abstract
Earlier, low-temperature-active polygalacturonase isoforms from Saccharomyces cerevisiae PVK4 were isolated and purified. Substrate specificity of polygalacturonase isoforms indicated high affinity for pectins and very low enzyme activity towards non-pectic polysaccharides. To characterize the polygalacturonase isoforms, biochemical, spectral, and in silico approaches were used. The apparent Km and Vmax values for hydrolysis of pectin and galacturonic acid were 0.31 mg/ml and 3.15 mmol min/mg, respectively. Interestingly, the polygalacturonase isoforms were found to be more stable at optimal pH and temperature of 4.5 and 40 °C, respectively. These isoforms were reacted with different metal ions; Cd2+ and Ni2+ severely inhibited the enzyme activity, while Mg2+, Zn2+, Cd2+, Fe2+ Cu2+, and Ni2+ inhibited to a lesser extent, which clearly demonstrated that variations in enzyme activity were due to their differential binding affinity of metal ions. Furthermore, decrease in the viscosity of polygalacturonic acid and citrus pectin by these isoforms was approximately four and six times higher than the rate of release of reducing sugars. This indicates that polygalacturonase isoforms have an endo-mode of action. In addition to the above, thermostability of purified polygalacturonase isoforms was studied by circular dichroism and fluorescence spectroscopy. Circular dichroism showed 18% alpha helix and 57% beta sheets at pH 5, while at pH 7, 8, and 9 there was an increase of random coil. Fluorescence studies revealed small conformational changes, which were observed at 30-50 °C, while unfolding transition region was noticed between 60 and 70 °C. The purified enzyme fractions were analyzed by MALDI-TOF MS. Finally, 3D model structures for isoenzymes of polygalacturonase of S. cerevisiae were generated and validated as good quality models, which are also suitable for molecular interaction studies.
Collapse
|
40
|
Wang Y, MacDonald RG, Thinakaran G, Kar S. Insulin-Like Growth Factor-II/Cation-Independent Mannose 6-Phosphate Receptor in Neurodegenerative Diseases. Mol Neurobiol 2017; 54:2636-2658. [PMID: 26993302 PMCID: PMC5901910 DOI: 10.1007/s12035-016-9849-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
Abstract
The insulin-like growth factor II/mannose 6-phosphate (IGF-II/M6P) receptor is a multifunctional single transmembrane glycoprotein. Recent studies have advanced our understanding of the structure, ligand-binding properties, and trafficking of the IGF-II/M6P receptor. This receptor has been implicated in a variety of important cellular processes including growth and development, clearance of IGF-II, proteolytic activation of enzymes, and growth factor precursors, in addition to its well-known role in the delivery of lysosomal enzymes. The IGF-II/M6P receptor, distributed widely in the central nervous system, has additional roles in mediating neurotransmitter release and memory enhancement/consolidation, possibly through activating IGF-II-related intracellular signaling pathways. Recent studies suggest that overexpression of the IGF-II/M6P receptor may have an important role in regulating the levels of transcripts and proteins involved in the development of Alzheimer's disease (AD)-the prevalent cause of dementia affecting the elderly population in our society. It is reported that IGF-II/M6P receptor overexpression can increase the levels/processing of amyloid precursor protein leading to the generation of β-amyloid peptide, which is associated with degeneration of neurons and subsequent development of AD pathology. Given the significance of the receptor in mediating the transport and functioning of the lysosomal enzymes, it is being considered for therapeutic delivery of enzymes to the lysosomes to treat lysosomal storage disorders. Notwithstanding these results, additional studies are required to validate and fully characterize the function of the IGF-II/M6P receptor in the normal brain and its involvement in various neurodegenerative disorders including AD. It is also critical to understand the interaction between the IGF-II/M6P receptor and lysosomal enzymes in neurodegenerative processes, which may shed some light on developing approaches to detect and prevent neurodegeneration through the dysfunction of the receptor and the endosomal-lysosomal system.
Collapse
Affiliation(s)
- Y Wang
- Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2M8, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - R G MacDonald
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - G Thinakaran
- Departments of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - S Kar
- Department of Psychiatry, University of Alberta, Edmonton, AB, T6G 2M8, Canada.
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB, T6G 2M8, Canada.
| |
Collapse
|
41
|
Drew DA, Chin SM, Gilpin KK, Parziale M, Pond E, Schuck MM, Stewart K, Flagg M, Rawlings CA, Backman V, Carolan PJ, Chung DC, Colizzo FP, Freedman M, Gala M, Garber JJ, Huttenhower C, Kedrin D, Khalili H, Kwon DS, Markowitz SD, Milne GL, Nishioka NS, Richter JM, Roy HK, Staller K, Wang M, Chan AT. ASPirin Intervention for the REDuction of colorectal cancer risk (ASPIRED): a study protocol for a randomized controlled trial. Trials 2017; 18:50. [PMID: 28143522 PMCID: PMC5286828 DOI: 10.1186/s13063-016-1744-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Although aspirin is recommended for the prevention of colorectal cancer, the specific individuals for whom the benefits outweigh the risks are not clearly defined. Moreover, the precise mechanisms by which aspirin reduces the risk of cancer are unclear. We recently launched the ASPirin Intervention for the REDuction of colorectal cancer risk (ASPIRED) trial to address these uncertainties. METHODS/DESIGN ASPIRED is a prospective, double-blind, multidose, placebo-controlled, biomarker clinical trial of aspirin use in individuals previously diagnosed with colorectal adenoma. Individuals (n = 180) will be randomized in a 1:1:1 ratio to low-dose (81 mg/day) or standard-dose (325 mg/day) aspirin or placebo. At two study visits, participants will provide lifestyle, dietary and biometric data in addition to urine, saliva and blood specimens. Stool, grossly normal colorectal mucosal biopsies and cytology brushings will be collected during a flexible sigmoidoscopy without bowel preparation. The study will examine the effect of aspirin on urinary prostaglandin metabolites (PGE-M; primary endpoint), plasma inflammatory markers (macrophage inhibitory cytokine-1 (MIC-1)), colonic expression of transcription factor binding (transcription factor 7-like 2 (TCF7L2)), colonocyte gene expression, including hydroxyprostaglandin dehydrogenase 15-(NAD) (HPGD) and those that encode Wnt signaling proteins, colonic cellular nanocytology and oral and gut microbial composition and function. DISCUSSION Aspirin may prevent colorectal cancer through multiple, interrelated mechanisms. The ASPIRED trial will scrutinize these pathways and investigate putative mechanistically based risk-stratification biomarkers. TRIAL REGISTRATION This protocol is registered with the U.S. National Institutes of Health trial registry, ClinicalTrials.gov, under the identifier NCT02394769 . Registered on 16 March 2015.
Collapse
Affiliation(s)
- David A. Drew
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Samantha M. Chin
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Katherine K. Gilpin
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Melanie Parziale
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Emily Pond
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Madeline M. Schuck
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Kathleen Stewart
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Meaghan Flagg
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA USA
| | | | - Vadim Backman
- McCormick School of Engineering, Northwestern University, Evanston, IL USA
| | - Peter J. Carolan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Daniel C. Chung
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Francis P. Colizzo
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | | | - Manish Gala
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - John J. Garber
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Dmitriy Kedrin
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Hamed Khalili
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Douglas S. Kwon
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA USA
| | - Sanford D. Markowitz
- Case Western Reserve University School of Medicine and University Hospitals of Cleveland, Cleveland, OH USA
| | - Ginger L. Milne
- Eicosanoid Core Laboratory, Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Norman S. Nishioka
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - James M. Richter
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Hemant K. Roy
- Section of Gastroenterology, Boston Medical Center, Boston, MA USA
| | - Kyle Staller
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Molin Wang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
- Broad Institute, Cambridge, MA USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
- Division of Gastroenterology and Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, GRJ-825C, Boston, MA 02114 USA
| |
Collapse
|
42
|
Papaioannou A, Kuyucak S, Kuncic Z. Elucidating the Activation Mechanism of the Insulin-Family Proteins with Molecular Dynamics Simulations. PLoS One 2016; 11:e0161459. [PMID: 27548502 PMCID: PMC4993506 DOI: 10.1371/journal.pone.0161459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022] Open
Abstract
The insulin-family proteins bind to their own receptors, but insulin-like growth factor II (IGF-II) can also bind to the A isoform of the insulin receptor (IR-A), activating unique and alternative signaling pathways from those of insulin. Although extensive studies of insulin have revealed that its activation is associated with the opening of the B chain-C terminal (BC-CT), the activation mechanism of the insulin-like growth factors (IGFs) still remains unknown. Here, we present the first comprehensive study of the insulin-family proteins comparing their activation process and mechanism using molecular dynamics simulations to reveal new insights into their specificity to the insulin receptor. We have found that all the proteins appear to exhibit similar stochastic dynamics in their conformational change to an active state. For the IGFs, our simulations show that activation involves two opening locations: the opening of the BC-CT section away from the core, similar to insulin; and the additional opening of the BC-CT section away from the C domain. Furthermore, we have found that these two openings occur simultaneously in IGF-I, but not in IGF-II, where they can occur independently. This suggests that the BC-CT section and the C domain behave as a unified domain in IGF-I, but as two independent domains in IGF-II during the activation process, implying that the IGFs undergo different activation mechanisms for receptor binding. The probabilities of the active and inactive states of the proteins suggest that IGF-II is hyperactive compared to IGF-I. The hinge residue and the hydrophobic interactions in the core are found to play a critical role in the stability and activity of IGFs. Overall, our simulations have elucidated the crucial differences and similarities in the activation mechanisms of the insulin-family proteins, providing new insights into the molecular mechanisms responsible for the observed differences between IGF-I and IGF-II in receptor binding.
Collapse
Affiliation(s)
- Anastasios Papaioannou
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Physics, University of Sydney, Sydney, NSW, Australia
- * E-mail: (AP); (ZK)
| | - Serdar Kuyucak
- School of Physics, University of Sydney, Sydney, NSW, Australia
| | - Zdenka Kuncic
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Physics, University of Sydney, Sydney, NSW, Australia
- * E-mail: (AP); (ZK)
| |
Collapse
|
43
|
Frago S, Nicholls RD, Strickland M, Hughes J, Williams C, Garner L, Surakhy M, Maclean R, Rezgui D, Prince SN, Zaccheo OJ, Ebner D, Sanegre S, Yu S, Buffa FM, Crump MP, Hassan AB. Functional evolution of IGF2:IGF2R domain 11 binding generates novel structural interactions and a specific IGF2 antagonist. Proc Natl Acad Sci U S A 2016; 113:E2766-75. [PMID: 27140600 PMCID: PMC4878476 DOI: 10.1073/pnas.1513023113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Among the 15 extracellular domains of the mannose 6-phosphate/insulin-like growth factor-2 receptor (M6P/IGF2R), domain 11 has evolved a binding site for IGF2 to negatively regulate ligand bioavailability and mammalian growth. Despite the highly evolved structural loops of the IGF2:domain 11 binding site, affinity-enhancing AB loop mutations suggest that binding is modifiable. Here we examine the extent to which IGF2:domain 11 affinity, and its specificity over IGF1, can be enhanced, and we examine the structural basis of the mechanistic and functional consequences. Domain 11 binding loop mutants were selected by yeast surface display combined with high-resolution structure-based predictions, and validated by surface plasmon resonance. We discovered previously unidentified mutations in the ligand-interacting surface binding loops (AB, CD, FG, and HI). Five combined mutations increased rigidity of the AB loop, as confirmed by NMR. When added to three independently identified CD and FG loop mutations that reduced the koff value by twofold, these mutations resulted in an overall selective 100-fold improvement in affinity. The structural basis of the evolved affinity was improved shape complementarity established by interloop (AB-CD) and intraloop (FG-FG) side chain interactions. The high affinity of the combinatorial domain 11 Fc fusion proteins functioned as ligand-soluble antagonists or traps that depleted pathological IGF2 isoforms from serum and abrogated IGF2-dependent signaling in vivo. An evolved and reengineered high-specificity M6P/IGF2R domain 11 binding site for IGF2 may improve therapeutic targeting of the frequent IGF2 gain of function observed in human cancer.
Collapse
Affiliation(s)
- Susana Frago
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Ryan D Nicholls
- Department of Organic and Biological Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Madeleine Strickland
- Department of Organic and Biological Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Jennifer Hughes
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Christopher Williams
- Department of Organic and Biological Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Lee Garner
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Mirvat Surakhy
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Rory Maclean
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Dellel Rezgui
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Stuart N Prince
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Oliver J Zaccheo
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Daniel Ebner
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, United Kingdom
| | - Sabina Sanegre
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Sheng Yu
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Francesca M Buffa
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Matthew P Crump
- Department of Organic and Biological Chemistry, School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom;
| | - Andrew Bassim Hassan
- Tumour Growth Control Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom;
| |
Collapse
|
44
|
Brohus M, Gorbunova V, Faulkes CG, Overgaard MT, Conover CA. The Insulin-Like Growth Factor System in the Long-Lived Naked Mole-Rat. PLoS One 2015; 10:e0145587. [PMID: 26694858 PMCID: PMC4694111 DOI: 10.1371/journal.pone.0145587] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/04/2015] [Indexed: 11/29/2022] Open
Abstract
Naked mole-rats (Heterocephalus glaber) (NMRs) are the longest living rodents known. They show negligible senescence, and are resistant to cancers and certain damaging effects associated with aging. The insulin-like growth factors (IGFs) have pluripotent actions, influencing growth processes in virtually every system of the body. They are established contributors to the aging process, confirmed by the demonstration that decreased IGF signaling results in life-extending effects in a variety of species. The IGFs are likewise involved in progression of cancers by mediating survival signals in malignant cells. This report presents a full characterization of the IGF system in the NMR: ligands, receptors, IGF binding proteins (IGFBPs), and IGFBP proteases. A particular emphasis was placed on the IGFBP protease, pregnancy-associated plasma protein-A (PAPP-A), shown to be an important lifespan modulator in mice. Comparisons of IGF-related genes in the NMR with human and murine sequences indicated no major differences in essential parts of the IGF system, including PAPP-A. The protease was shown to possess an intact active site despite the report of a contradictory genome sequence. Furthermore, PAPP-A was expressed and translated in NMRs cells and retained IGF-dependent proteolytic activity towards IGFBP-4 and IGF-independent activity towards IGFBP-5. However, experimental data suggest differential regulatory mechanisms for PAPP-A expression in NMRs than those described in humans and mice. This overall description of the IGF system in the NMR represents an initial step towards elucidating the complex molecular mechanisms underlying longevity, and how these animals have evolved to ensure a delayed and healthy aging process.
Collapse
Affiliation(s)
- Malene Brohus
- The Division of Endocrinology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States of America
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers Vej 7H, DK-9220 Aalborg Oe, Denmark
| | - Vera Gorbunova
- University of Rochester, Department of Biology, 434 Hutchinson Hall, River Campus, Rochester, New York, 14627, United States of America
| | - Chris G. Faulkes
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Michael T. Overgaard
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers Vej 7H, DK-9220 Aalborg Oe, Denmark
| | - Cheryl A. Conover
- The Division of Endocrinology, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States of America
- * E-mail:
| |
Collapse
|
45
|
D'Alessio C, Dahms NM. Glucosidase II and MRH-domain containing proteins in the secretory pathway. Curr Protein Pept Sci 2015; 16:31-48. [PMID: 25692846 DOI: 10.2174/1389203716666150213160438] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 08/25/2014] [Accepted: 11/13/2014] [Indexed: 12/24/2022]
Abstract
N-glycosylation in the endoplasmic reticulum (ER) consists of the transfer of a preassembled glycan conserved among species (Glc3Man9GlcNAc2) from a lipid donor to a consensus sequence within a nascent protein that is entering the ER. The protein-linked glycans are then processed by glycosidases and glycosyltransferases in the ER producing specific structures that serve as signalling molecules for the fate of the folding glycoprotein: to stay in the ER during the folding process, to be retrotranslocated to the cytosol for proteasomal degradation if irreversibly misfolded, or to pursue transit through the secretory pathway as a mature glycoprotein. In the ER, each glycan signalling structure is recognized by a specific lectin. A domain similar to that of the mannose 6-phosphate receptors (MPRs) has been identified in several proteins of the secretory pathway. These include the beta subunit of glucosidase II (GII), a key enzyme in the early processing of the transferred glycan that removes middle and innermost glucoses and is involved in quality control of glycoprotein folding in the ER (QC), the lectins OS-9 and XTP3-B, proteins involved in the delivery of ER misfolded proteins to degradation (ERAD), the gamma subunit of the Golgi GlcNAc-1-phosphotransferase, an enzyme involved in generating the mannose 6-phosphate (M6P) signal for sorting acidic hydrolases to lysosomes, and finally the MPRs that deliver those hydrolytic enzymes to the lysosome. Each of the MRH-containing proteins recognizes a different signalling N-glycan structure. Three-dimensional structures of some of the MRH domains have been solved, providing the basis to understand recognition mechanisms.
Collapse
Affiliation(s)
| | - Nancy M Dahms
- Laboratory of Glycobiology, Fundación Instituto Leloir - Instituto de Investigaciones Bioquimicas de Buenos Aires-CONICET, Av. Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina, and School of Sciences, University of Buenos Aires, C1428EHA, Buenos Aires, Argentina.
| |
Collapse
|
46
|
Abstract
The insulin/insulin-like signaling and target of rapamycin (IIS/TOR) network regulates lifespan and reproduction, as well as metabolic diseases, cancer, and aging. Despite its vital role in health, comparative analyses of IIS/TOR have been limited to invertebrates and mammals. We conducted an extensive evolutionary analysis of the IIS/TOR network across 66 amniotes with 18 newly generated transcriptomes from nonavian reptiles and additional available genomes/transcriptomes. We uncovered rapid and extensive molecular evolution between reptiles (including birds) and mammals: (i) the IIS/TOR network, including the critical nodes insulin receptor substrate (IRS) and phosphatidylinositol 3-kinase (PI3K), exhibit divergent evolutionary rates between reptiles and mammals; (ii) compared with a proxy for the rest of the genome, genes of the IIS/TOR extracellular network exhibit exceptionally fast evolutionary rates; and (iii) signatures of positive selection and coevolution of the extracellular network suggest reptile- and mammal-specific interactions between members of the network. In reptiles, positively selected sites cluster on the binding surfaces of insulin-like growth factor 1 (IGF1), IGF1 receptor (IGF1R), and insulin receptor (INSR); whereas in mammals, positively selected sites clustered on the IGF2 binding surface, suggesting that these hormone-receptor binding affinities are targets of positive selection. Further, contrary to reports that IGF2R binds IGF2 only in marsupial and placental mammals, we found positively selected sites clustered on the hormone binding surface of reptile IGF2R that suggest that IGF2R binds to IGF hormones in diverse taxa and may have evolved in reptiles. These data suggest that key IIS/TOR paralogs have sub- or neofunctionalized between mammals and reptiles and that this network may underlie fundamental life history and physiological differences between these amniote sister clades.
Collapse
|
47
|
Olson LJ, Castonguay AC, Lasanajak Y, Peterson FC, Cummings RD, Smith DF, Dahms NM. Identification of a fourth mannose 6-phosphate binding site in the cation-independent mannose 6-phosphate receptor. Glycobiology 2015; 25:591-606. [PMID: 25573276 DOI: 10.1093/glycob/cwv001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/05/2015] [Indexed: 11/12/2022] Open
Abstract
The 300 kDa cation-independent mannose 6-phosphate receptor (CI-MPR) plays an essential role in lysosome biogenesis by targeting ∼ 60 different phosphomannosyl-containing acid hydrolases to the lysosome. This type I membrane glycoprotein has a large extracellular region comprised of 15 homologous domains. Two mannose 6-phosphate (M6P) binding sites have been mapped to domains 3 and 9, whereas domain 5 binds preferentially to the phosphodiester, M6P-N-acetylglucosamine (GlcNAc). A structure-based sequence alignment predicts that the C-terminal domain 15 contains three out of the four conserved residues identified as essential for carbohydrate recognition by domains 3, 5 and 9 of the CI-MPR, but lacks two cysteine residues that are predicted to form a disulfide bond. To determine whether domain 15 of the CI-MPR has lectin activity and to probe its carbohydrate-binding specificity, truncated forms of the CI-MPR were tested for binding to acid hydrolases with defined N-glycans in surface plasmon resonance analyses, and used to interrogate a phosphorylated glycan microarray. The results show that a construct encoding domains 14-15 binds both M6P and M6P-GlcNAc with similar affinity (Kd = 13 and 17 μM, respectively). Site-directed mutagenesis studies demonstrate the essential role of the conserved Tyr residue in domain 15 for phosphomannosyl binding. A structural model of domain 15 was generated that predicted an Arg residue to be in the binding pocket and mutagenesis studies confirmed its important role in carbohydrate binding. Together, these results show that the CI-MPR contains a fourth carbohydrate-recognition site capable of binding both phosphomonoesters and phosphodiesters.
Collapse
Affiliation(s)
- Linda J Olson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alicia C Castonguay
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yi Lasanajak
- National Center for Functional Glycomics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Richard D Cummings
- National Center for Functional Glycomics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - David F Smith
- National Center for Functional Glycomics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
48
|
van Veggel KM, Huits RMHG, Donker GH, Lentjes EGWM, van Doorn J. Column chromatographic characterization of complex formation of pro-IGF-II isoforms with acid labile subunit and IGF-binding proteins associated with non-islet cell tumour induced hypoglycaemia. Growth Horm IGF Res 2014; 24:233-238. [PMID: 25174810 DOI: 10.1016/j.ghir.2014.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/06/2014] [Accepted: 08/06/2014] [Indexed: 12/01/2022]
Abstract
OBJECTIVE AND DESIGN Non-islet cell tumour induced hypoglycaemia (NICTH) is a paraneoplastic phenomenon that is associated with the formation of several isoforms of pro-insulin like growth factor 2 (pro-IGF-II), or so called "big" IGF-II. Disturbance of ternary complex formation by big IGF-II is assumed to be a crucial early event in the pathogenic cascade of hypoglycaemia. By size-exclusion chromatography, we investigated complex formation by adding different naturally occurring isoforms of pro-IGF-II to pooled normal adult serum. Results were compared with the analysis of the serum from a patient with NICTH. RESULTS Gel filtration experiments with the serum of a patient with NICTH demonstrated that ternary complex formation was severely compromised. The various forms of pro-IGF-II did not induce a shift of IGF-binding protein 3 (IGFBP-3) from 150kD towards smaller binary complexes in the normal adult serum, suggesting that they did not interfere with the interaction between the acid labile subunit and IGFBP-3. Instead, unglycosylated recombinant pro-IGF-II[1-104] was capable of forming a 150kD complex. In contrast, predominantly glycosylated and unglycosylated pro-IGF-II[1-87] eluted in the free unbound form. We showed that mature IGF-II and isoforms of pro-IGF-II were able to phosphorylate the IGF-I receptors of MC7 cells, albeit to a markedly lesser extent than IGF-I. When the patient's serum was tested in this system, the IGF-I receptor phosphorylation activity was considerably less than that in sera from age matched healthy individuals. CONCLUSION We postulate that, alongside the presence of big IGF-II in the circulation, additional steps are required to stimulate the release of IGF-II and pro-IGF-II isoforms from IGFBPs in vivo. These factors may be proteases, that are present in the local environment of the tumour and in insulin-sensitive tissues.
Collapse
Affiliation(s)
- K M van Veggel
- Dept. Internal Medicine, Dr. Horacio E. Oduber Hospital, Oranjestad, Aruba; Dept. Internal Medicine, Rijnland Hospital, Leiderdorp, The Netherlands.
| | - R M H G Huits
- Dept. Internal Medicine, Dr. Horacio E. Oduber Hospital, Oranjestad, Aruba; Center for Infectious Diseases, ID4C, Military Hospital Queen Astrid, Brussels, Belgium
| | - G H Donker
- Dept. Clinical Chemistry and Hematology, Laboratory of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E G W M Lentjes
- Dept. Clinical Chemistry and Hematology, Laboratory of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J van Doorn
- Dept. Clinical Chemistry and Hematology, Laboratory of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands; Dept. Medical Genetics, Section Metabolic Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
49
|
Martínez P, Sales Fidalgo PA, Felip E. Ganitumab for the treatment of small-cell lung cancer. Expert Opin Investig Drugs 2014; 23:1423-32. [PMID: 25189625 DOI: 10.1517/13543784.2014.951434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Small-cell lung cancer (SCLC) accounts for 15 - 20% of all lung cancer cases with few advances made in the systemic treatment and outcomes for extensive-stage SCLC. Many strategies have been evaluated over the past 15 years but none of these approaches has resulted in improved survival rates for patients with SCLC. The IGF receptor (IGF-R) pathway represents a potential actionable target in SCLC patients. Indeed, the IGF-R pathway is involved in cancer development and progression and regulates different vital processes including fetal development, growth and metabolism. AREAS COVERED This review provides an overview of insulin inhibitors and the strategies undertaken in recent years with SCLC. Specifically, the article discusses ganitumab and its applicability to SCLC patients. EXPERT OPINION At present, there is a lack of therapeutic choices for patients with advanced SCLC. Unfortunately, ganitumab, administered in combination with chemotherapy, demonstrated no clinical activity in patients with SCLC, although it could have utility with other cancers. Furthermore, insulin inhibitors may have some utility in the treatment of SCLC and further studies are required to identify subsets of patients most likely to benefit from their use. The authors also believe that it is important to determine the exact role of the IGF pathway in the pathogenesis and propagation of SCLC.
Collapse
Affiliation(s)
- Pablo Martínez
- Vall d´Hebron University Hospital and Vall d´Hebron Institute of Oncology, Medical Oncology Department , Barcelona , Spain
| | | | | |
Collapse
|
50
|
Lund J, Søndergaard MT, Conover CA, Overgaard MT. Heparin-binding mechanism of the IGF2/IGF-binding protein 2 complex. J Mol Endocrinol 2014; 52:345-55. [PMID: 24604839 DOI: 10.1530/jme-13-0184] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
IGF1 and IGF2 are potent stimulators of diverse cellular activities such as differentiation and mitosis. Six IGF-binding proteins (IGFBP1-IGFBP6) are primary regulators of IGF half-life and receptor availability. Generally, the binding of IGFBPs inhibits IGF receptor activation. However, it has been shown that IGFBP2 in complex with IGF2 (IGF2/IGFBP2) stimulates osteoblast function in vitro and increases skeletal mass in vivo. IGF2 binding to IGFBP2 greatly increases the affinity for 2- or 3-carbon O-sulfated glycosaminoglycans (GAGs), e.g. heparin and heparan sulfate, which is hypothesized to preferentially and specifically target the IGF2/IGFBP2 complex to the bone matrix. In order to obtain a more detailed understanding of the interactions between the IGF2/IGFBP2 complex and GAGs, we investigated heparin-binding properties of IGFBP2 and the IGF2/IGFBP2 complex in a quantitative manner. For this study, we mutated key positively charged residues within the two heparin-binding domains (HBDs) in IGFBP2 and in one potential HBD in IGF2. Using heparin affinity chromatography, we demonstrate that the two IGFBP2 HBDs contribute differentially to GAG binding in free IGFBP2 and the IGF2/IGFBP2 protein complex. Moreover, we identify a significant contribution from the HBD in IGF2 to the increased IGF2/IGFBP2 heparin affinity. Using molecular modeling, we present a novel model for the IGF2/IGFBP2 interaction with heparin where all three proposed HBDs constitute a positively charged and surface-exposed area that would serve to promote the increased heparin affinity of the complex compared with free intact IGFBP2.
Collapse
Affiliation(s)
- Jacob Lund
- Department of BiotechnologyChemistry, and Environmental Engineering, Aalborg University, Sohngaardsholmsvej 49, DK-9000 Aalborg, DenmarkDivision of EndocrinologyEndocrine Research Unit, Mayo Clinic, Rochester, Minnesota, USA
| | - Mads T Søndergaard
- Department of BiotechnologyChemistry, and Environmental Engineering, Aalborg University, Sohngaardsholmsvej 49, DK-9000 Aalborg, DenmarkDivision of EndocrinologyEndocrine Research Unit, Mayo Clinic, Rochester, Minnesota, USA
| | - Cheryl A Conover
- Department of BiotechnologyChemistry, and Environmental Engineering, Aalborg University, Sohngaardsholmsvej 49, DK-9000 Aalborg, DenmarkDivision of EndocrinologyEndocrine Research Unit, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael T Overgaard
- Department of BiotechnologyChemistry, and Environmental Engineering, Aalborg University, Sohngaardsholmsvej 49, DK-9000 Aalborg, DenmarkDivision of EndocrinologyEndocrine Research Unit, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|