1
|
Kumar P, Mishra T, Sanyam, Mondal A, Basu S. Triphenylamine-Naphthalimide-Based "On-Off-On" AIEgen for Imaging Golgi Apparatus and Endoplasmic Reticulum. ACS APPLIED BIO MATERIALS 2025; 8:1524-1532. [PMID: 39835412 DOI: 10.1021/acsabm.4c01722] [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] [Indexed: 01/22/2025]
Abstract
Golgi apparatus (GA) and endoplasmic reticulum (ER) are two of the interesting subcellular organelles that are critical for protein synthesis, folding, processing, post-translational modifications, and secretion. Consequently, dysregulation in GA and ER and cross-talk between them are implicated in numerous diseases including cancer. As a result, simultaneous visualization of the GA and ER in cancer cells is extremely crucial for developing cancer therapeutics. To address this, herein, we have designed and synthesized a 1,8-napthalimide-based small molecule (AIE-GA-ER) consisting of phenylsulfonamide as Golgi-ER homing and triphenylamine-napthalimide as aggregation-induced emission (AIE) triggering moieties. AIE-GA-ER exhibited remarkable "on-off-on" AIE properties in THF/water binary solvent system due to aggregated "on-state" in pure THF and 80% water in THF. Molecular dynamic simulations and density functional theory (DFT) calculations exhibited the underlying mechanism of the emissive property of AIE-GA-ER to be the interplay between intramolecular charge transfer (ICT) stabilization and aggregation in THF, DMSO, and water. AIE-GA-ER efficiently homed into the GA and ER of HCT-116 colon cancer cells within 15-30 min as well as noncancerous human retinal epithelial pigment cells (RPE-1) within 3 h with minimum toxicity. This AIEgen has the potential to illuminate the Golgi apparatus and ER simultaneously in cancer cells to understand the chemical biology of their cross-talk for next-generation cancer therapeutics.
Collapse
Affiliation(s)
- Phanindra Kumar
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Tripti Mishra
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Sanyam
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Anirban Mondal
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| | - Sudipta Basu
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
2
|
Liu Z, Liu Y, Kang X, Li L, Xiang Y. Subcellular Organelle Targeting as a Novel Approach to Combat Tumor Metastasis. Pharmaceutics 2025; 17:198. [PMID: 40006565 PMCID: PMC11859411 DOI: 10.3390/pharmaceutics17020198] [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: 12/31/2024] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor metastasis, the spread of cancer cells from the primary site to distant organs, remains a formidable challenge in oncology. Central to this process is the involvement of subcellular organelles, which undergo significant functional and structural changes during metastasis. Targeting these specific organelles offers a promising avenue for enhanced drug delivery and metastasis therapeutic efficacy. This precision increases the potency and reduces potential off-target effects. Moreover, by understanding the role of each organelle in metastasis, treatments can be designed to disrupt the metastatic process at multiple stages, from cell migration to the establishment of secondary tumors. This review delves deeply into tumor metastasis processes and their connection with subcellular organelles. In order to target these organelles, biomembranes, cell-penetrating peptides, localization signal peptides, aptamers, specific small molecules, and various other strategies have been developed. In this review, we will elucidate targeting delivery strategies for each subcellular organelle and look forward to prospects in this domain.
Collapse
Affiliation(s)
- Zefan Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Yang Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Xin Kang
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
| | - Yucheng Xiang
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
3
|
Yin H, Jiang D, Li Y, Chen W, Zhang J, Yang X, Hu J, Wei H. KDELR1 regulates chondrosarcoma drug resistance and malignant behavior through Intergrin-Hippo-YAP1 axis. Cell Death Dis 2024; 15:928. [PMID: 39715773 DOI: 10.1038/s41419-024-07264-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Chondrosarcoma (CS) is the second most common primary bone malignancy, known for its unique transcriptional landscape that renders most CS subtypes resistant to chemotherapy, including neoadjuvant chemotherapy commonly used in osteosarcoma (OS) treatment. Understanding the transcriptional landscape of CS and the mechanisms by which key genes contribute to chemotherapy resistance could be a crucial step in overcoming this challenge. To address this, we developed a single-cell transcriptional map of CS, comparing it with OS and normal cancellous bone. Our analysis revealed a specific increase in KDEL receptor 1 (KDELR1) expression in CS, which was closely associated with CS prognosis, tumor aggressiveness, and drug resistance. KDELR1 plays a key role in regulating membrane protein processing and secretion, as well as contributing to tumor extracellular matrix (ECM) formation and drug resistance. Further investigation using mass spectrometry proteomics and transcriptomics uncovered KDELR1's involvement in modulating the Hippo-YAP pathway activity in CS cells. The KDELR1-Integrin-PLCγ-YAP1 axis emerges as a critical process mediating drug resistance and malignant behavior in CS, offering novel insights and potential therapeutic targets for CS treatment.
Collapse
Affiliation(s)
- Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85 Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Dongjie Jiang
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Yongai Li
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85 Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Wenjun Chen
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Jie Zhang
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Xinghai Yang
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| | - Jinbo Hu
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| | - Haifeng Wei
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
4
|
Liu F, Li Z, Jing J, Zhang X. A Golgi-targeted fluorescent probe for monitoring polarity dynamic during programmed cell death. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 322:124810. [PMID: 39002471 DOI: 10.1016/j.saa.2024.124810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Programmed cell death (PCD) is a controlled form of cell death and it plays an essential role in maintaining homeostasis. Golgi apparatus works as the hotspot during the early event of PCD and Golgi polarity, a vital microenvironment factor, can be regarded as an indicator of physiological status. Combined Golgi-targeted group phenylsulfonamide as electron acceptor group and triphenylamine as electron donor group, a novel Golgi-targeted fluorescent probe GTO had been developed. GTO showed good sensitivity and selectivity to polarity and its remarkable photostability makes it potentially useful for long-term cellular monitoring. In practice, GTO demonstrated good cell permeability and Golgi targeting capabilities. According to our results, GTO was applied to reveal the polarity increase during the early event of PCD and the encouraging results illustrated that GTO was an imaging tool for monitoring polarity in Golgi apparatus and the exploration in early diagnosis and drug discovery.
Collapse
Affiliation(s)
- Feiran Liu
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photo-electronic/Electrophotonic Conversion Materials, Analytical and Testing Center, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Zichun Li
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photo-electronic/Electrophotonic Conversion Materials, Analytical and Testing Center, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Jing Jing
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photo-electronic/Electrophotonic Conversion Materials, Analytical and Testing Center, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, PR China.
| | - Xiaoling Zhang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key Laboratory of Photo-electronic/Electrophotonic Conversion Materials, Analytical and Testing Center, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, PR China; School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, PR China.
| |
Collapse
|
5
|
Shi M, Fu Z, Pan W, Wang K, Liu X, Li N, Tang B. A Golgi Apparatus-Targeted Photothermal Agent with Protein Anchoring for Enhanced Cancer Photothermal Therapy. Adv Healthc Mater 2024; 13:e2303749. [PMID: 38483042 DOI: 10.1002/adhm.202303749] [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: 11/02/2023] [Revised: 03/06/2024] [Indexed: 03/23/2024]
Abstract
The Golgi apparatus (GA) is central in shuttling proteins from the endoplasmic reticulum to different cellular areas. Therefore, targeting the GA to precisely destroy its proteins through local heat could induce apoptosis, offering a potential avenue for effective cancer therapy. Herein, a GA-targeted photothermal agent based on protein anchoring is introduced for enhanced photothermal therapy of tumor through the modification of near-infrared molecular dye with maleimide derivative and benzene sulfonamide. The photothermal agent can actively target the GA and covalently anchor to its sulfhydryl proteins, thereby increasing its retention within the GA. Under laser irradiation, the heat generated by the photothermal agent efficiently disrupts sulfhydryl proteins in situ, leading to GA dysfunction and ultimately inducing cell apoptosis. In vivo experiments demonstrate that the photothermal agent can precisely treat tumors and significantly reduce side effects.
Collapse
Affiliation(s)
- Mingwan Shi
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
| | - Zhongliang Fu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
| | - Kaiye Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
| | - Xiaohan Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Jinan, 250014, P. R. China
- Laoshan Laboratory, Qingdao, 266237, P. R. China
| |
Collapse
|
6
|
Nedelcu A, Mocan T, Sabau LI, Matea CT, Tabaran F, Pop T, Delcea C, Mosteanu O, Mocan L. In vitro photothermal therapy of pancreatic cancer mediated by immunoglobulin G-functionalized silver nanoparticles. Sci Rep 2024; 14:14417. [PMID: 38909066 PMCID: PMC11193743 DOI: 10.1038/s41598-024-63142-4] [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: 12/20/2023] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Pancreatic cancer is one of the most aggressive forms of cancer, and treatment options are limited. One therapeutic approach is to use nanoparticles to deliver the active agent directly to pancreatic cancer cells. Nanoparticles can be designed to specifically target cancer cells, minimizing damage to healthy tissues. Silver nanoparticles have the unique ability to absorb light, especially in the near-infrared (NIR) region. In this study, silver nanoparticles functionalized with IgG molecules were synthesized and administered to pancreatic cancer cell lines. Subsequently, the cells were photo-excited using a 2 W 808 nm laser and further examined in PANC-1 pancreatic cancer cell lines. Flow cytometry and confocal microscopy combined with immunochemical staining were used to examine the interaction between photo-excited silver nanoparticles and pancreatic cancer cells. The photothermal therapy based on IgG-functionalized silver nanoparticles in pancreatic cancer induces dysfunction in the Golgi apparatus, leading to the activation of the caspase-3 apoptotic pathway and ultimately resulting in cellular apoptosis. These findings suggest that our proposed IgG nanoparticle laser treatment could emerge as a novel approach for the therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Andreea Nedelcu
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania
| | - Teodora Mocan
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania.
- Physiology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania.
| | - Lavinia Ioana Sabau
- Physiology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
| | - Cristian Tudor Matea
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania
- Department of Biosciences, University of Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria
| | - Flaviu Tabaran
- Department of Pathology, Sciences and Veterinary Medicine, University of Agricultural, ClujNapoca, Romania
| | - Teodora Pop
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
- Department of Gastroenterology, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
| | - Cristian Delcea
- Department of Forensic Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania.
| | - Ofelia Mosteanu
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
- Department of Gastroenterology, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania
| | - Lucian Mocan
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, ClujNapoca, Romania.
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, ClujNapoca, Romania.
| |
Collapse
|
7
|
Keller T, Trinks N, Brand J, Trippmacher S, Stahlhut P, Albrecht K, Papastavrou G, Koepsell H, Sauer M, Groll J. Design of Nanohydrogels for Targeted Intracellular Drug Transport to the Trans-Golgi Network. Adv Healthc Mater 2023; 12:e2201794. [PMID: 36739269 PMCID: PMC11469190 DOI: 10.1002/adhm.202201794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/27/2023] [Indexed: 02/06/2023]
Abstract
Nanohydrogels combine advantages of hydrogels and nanoparticles. In particular, they represent promising drug delivery systems. Nanogel synthesis by oxidative condensation of polyglycidol prepolymers, that are modified with thiol groups, results in crosslinking by disulfide bonds. Hereby, biomolecules like the antidiabetic peptide RS1-reg, derived from the regulatory protein RS1 of the Na+ -D-glucose cotransporter SGLT1, can be covalently bound by cysteine residues to the nanogel in a hydrophilic, stabilizing environment. After oral uptake, the acid-stable nanogels protect their loading during gastric passage from proteolytic degradation. Under alkaline conditions in small intestine the nanohydrogels become mucoadhesive, pass the intestinal mucosa and are taken up into small intestinal enterocytes by endocytosis. Using Caco-2 cells as a model for small intestinal enterocytes, by confocal laser scanning microscopy and structured illumination microscopy, the colocalization of fluorescent-labeled RS1-reg with markers of endosomes, lysosomes, and trans-Golgi-network after uptake with polyglycidol-based nanogels formed by precipitation polymerization is demonstrated. This indicates that RS1-reg follows the endosomal pathway. In the following, the design of bespoken nanohydrogels for specific targeting of RS1-reg to its site of action at the trans-Golgi network is described that might also represent a way of targeted transport for other drugs to their targets at the Golgi apparatus.
Collapse
Affiliation(s)
- Thorsten Keller
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Nora Trinks
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jessica Brand
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Steffen Trippmacher
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Krystyna Albrecht
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Georg Papastavrou
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Hermann Koepsell
- Institute of Anatomy and Cell BiologyUniversity of WürzburgKoellikerstraße 697070WürzburgGermany
| | - Markus Sauer
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| |
Collapse
|
8
|
Qiao L, Sinha S, Abd El‐Hafeez AA, Lo I, Midde KK, Ngo T, Aznar N, Lopez‐Sanchez I, Gupta V, Farquhar MG, Rangamani P, Ghosh P. A circuit for secretion-coupled cellular autonomy in multicellular eukaryotic cells. Mol Syst Biol 2023; 19:e11127. [PMID: 36856068 PMCID: PMC10090951 DOI: 10.15252/msb.202211127] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 03/02/2023] Open
Abstract
Cancers represent complex autonomous systems, displaying self-sufficiency in growth signaling. Autonomous growth is fueled by a cancer cell's ability to "secrete-and-sense" growth factors (GFs): a poorly understood phenomenon. Using an integrated computational and experimental approach, here we dissect the impact of a feedback-coupled GTPase circuit within the secretory pathway that imparts secretion-coupled autonomy. The circuit is assembled when the Ras-superfamily monomeric GTPase Arf1, and the heterotrimeric GTPase Giαβγ and their corresponding GAPs and GEFs are coupled by GIV/Girdin, a protein that is known to fuel aggressive traits in diverse cancers. One forward and two key negative feedback loops within the circuit create closed-loop control, allow the two GTPases to coregulate each other, and convert the expected switch-like behavior of Arf1-dependent secretion into an unexpected dose-response alignment behavior of sensing and secretion. Such behavior translates into cell survival that is self-sustained by stimulus-proportionate secretion. Proteomic studies and protein-protein interaction network analyses pinpoint GFs (e.g., the epidermal GF) as key stimuli for such self-sustenance. Findings highlight how the enhanced coupling of two biological switches in cancer cells is critical for multiscale feedback control to achieve secretion-coupled autonomy of growth factors.
Collapse
Affiliation(s)
- Lingxia Qiao
- Department of Mechanical and Aerospace Engineering, Jacob's School of EngineeringUniversity of California San DiegoLa JollaCAUSA
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Amer Ali Abd El‐Hafeez
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
- Present address:
Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer InstituteCairo UniversityCairoEgypt
| | - I‐Chung Lo
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Krishna K Midde
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Tony Ngo
- Skaggs School of Pharmacy and Pharmaceutical ScienceUniversity of California San DiegoLa JollaCAUSA
| | - Nicolas Aznar
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Inmaculada Lopez‐Sanchez
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Vijay Gupta
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Marilyn G Farquhar
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, Jacob's School of EngineeringUniversity of California San DiegoLa JollaCAUSA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
- Moores Comprehensive Cancer CenterUniversity of California San DiegoLa JollaCAUSA
- Department of Medicine, School of MedicineUniversity of California San DiegoLa JollaCAUSA
- Veterans Affairs Medical CenterLa JollaCAUSA
| |
Collapse
|
9
|
Geng J, Wang J, Wang H. Emerging Landscape of Cell-Penetrating Peptide-Mediated Organelle Restoration and Replacement. ACS Pharmacol Transl Sci 2023; 6:229-244. [PMID: 36798470 PMCID: PMC9926530 DOI: 10.1021/acsptsci.2c00229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Indexed: 01/18/2023]
Abstract
Organelles are specialized subunits within a cell membrane that perform specific roles or functions, and their dysfunction can lead to a variety of pathophysiologies including developmental defects, aging, and diseases (cancer, cardiovascular and neurodegenerative diseases). Recent studies have shown that cell-penetrating peptide (CPP)-based pharmacological therapies delivered to organelles or even directly resulting in organelle replacement can restore cell function and improve or prevent disease. In this review, we summarized the current developments in the precise delivery of exogenous cargoes via CPPs at the organelle level, CPP-mediated organelle delivery, and discuss their feasibility as next-generation targeting strategies for the diagnosis and treatment of diseases at the organelle level.
Collapse
Affiliation(s)
- Jingping Geng
- Department
of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang443002, China
- Interdisciplinary
Laboratory of Molecular Biology and Biophysics, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097Warszawa, Poland
| | - Jing Wang
- Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland21215, United States
| | - Hu Wang
- Department
of Microbiology and Immunology, Medical School, China Three Gorges University, Yichang443002, China
- Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland21215, United States
| |
Collapse
|
10
|
Jia TT, Zhang Y, Hou JT, Niu H, Wang S. H 2S-based fluorescent imaging for pathophysiological processes. Front Chem 2023; 11:1126309. [PMID: 36778034 PMCID: PMC9911449 DOI: 10.3389/fchem.2023.1126309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Hydrogen sulfide (H2S), as an important endogenous signaling molecule, plays a vital role in many physiological processes. The abnormal behaviors of hydrogen sulfide in organisms may lead to various pathophysiological processes. Monitoring the changes in hydrogen sulfide is helpful for pre-warning and treating these pathophysiological processes. Fluorescence imaging techniques can be used to observe changes in the concentration of analytes in organisms in real-time. Therefore, employing fluorescent probes imaging to investigate the behaviors of hydrogen sulfide in pathophysiological processes is vital. This paper reviews the design strategy and sensing mechanisms of hydrogen sulfide-based fluorescent probes, focusing on imaging applications in various pathophysiological processes, including neurodegenerative diseases, inflammation, apoptosis, oxidative stress, organ injury, and diabetes. This review not only demonstrates the specific value of hydrogen sulfide fluorescent probes in preclinical studies but also illuminates the potential application in clinical diagnostics.
Collapse
Affiliation(s)
- Tong-Tong Jia
- College of Chemistry and Chemical Engineering, Luoyang Normal University, Luoyang, China
| | - Yuanyuan Zhang
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Ji-Ting Hou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Huawei Niu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Shan Wang
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Ingle J, Sengupta P, Basu S. Illuminating Sub-Cellular Organelles by Small Molecule AIEgens. Chembiochem 2023; 24:e202200370. [PMID: 36161823 DOI: 10.1002/cbic.202200370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/25/2022] [Indexed: 01/05/2023]
Abstract
Sub-cellular organelles play a critical role in a myriad biological phenomena. Consequently, organelle structures and functions are invariably highjacked in diverse diseases including metabolic disorders, aging, and cancer. Hence, illuminating organelle dynamics is crucial in understanding the diseased states as well as developing organelle-targeted next generation therapeutics. In this review, we outline the novel small molecules which show remarkable aggregation-induced emission (AIE) properties due to restriction in intramolecular motion (RIM). We outline the examples of small molecules developed to image organelles like mitochondria, endoplasmic reticulum (ER), Golgi, lysosomes, nucleus, cell membrane and lipid droplets. These AIEgens have tremendous potential for next-generation phototherapy.
Collapse
Affiliation(s)
- Jaypalsing Ingle
- Discipline of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| | - Poulomi Sengupta
- Department of Chemistry, Indrashil University, Rajpur, Kadi, Mehsana, Gujarat, 382740, India
| | - Sudipta Basu
- Discipline of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| |
Collapse
|
12
|
Li RS, Wen C, Huang CZ, Li N. Functional molecules and nano-materials for the Golgi apparatus-targeted imaging and therapy. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
13
|
Liu C, Zhu H, Zhang Y, Su M, Liu M, Zhang X, Wang X, Rong X, Wang K, Li X, Zhu B. Recent advances in Golgi-targeted small-molecule fluorescent probes. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Tan W, Zhang Q, Quiñones-Frías MC, Hsu AY, Zhang Y, Rodal A, Hong P, Luo HR, Xu B. Enzyme-Responsive Peptide Thioesters for Targeting Golgi Apparatus. J Am Chem Soc 2022; 144:6709-6713. [PMID: 35404599 PMCID: PMC9069992 DOI: 10.1021/jacs.2c02238] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The Golgi apparatus (GA) is the hub of intracellular trafficking, but selectively targeting GA remains a challenge. We show an unconventional types of peptide thioesters, consisting of an aminoethyl thioester and acting as substrates of thioesterases, for instantly targeting the GA of cells. The peptide thioesters, above or below their critical micelle concentrations, enter cells mainly via caveolin-mediated endocytosis or macropinocytosis, respectively. After being hydrolyzed by GA-associated thioesterases, the resulting thiopeptides form dimers and accumulate in the GA. After saturating the GA, the thiopeptides are enriched in the endoplasmic reticulum (ER). Their buildup in ER and GA disrupts protein trafficking, thus leading to cell death via multiple pathways. The peptide thioesters target the GA of a wide variety of cells, including human, murine, and Drosophila cells. Changing d-diphenylalanine to l-diphenylalanine in the peptide maintains the GA-targeting ability. In addition, targeting GA redirects protein (e.g., NRAS) distribution. This work illustrates a thioesterase-responsive and redox-active molecular platform for targeting the GA and controlling cell fates.
Collapse
Affiliation(s)
- Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | | | - Alan Y. Hsu
- Department of Pathology, Harvard Medical School and Department of Laboratory Medicine, Children’s Hospital Boston and Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Yichi Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Avital Rodal
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Pengyu Hong
- Department of Computer Science, Brandeis University, Waltham, MA 02453, USA
| | - Hongbo R. Luo
- Department of Pathology, Harvard Medical School and Department of Laboratory Medicine, Children’s Hospital Boston and Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| |
Collapse
|
15
|
Niu Z, Yang F, Li H, Wang J, Ni Q, Ma C, Zhu H, Chang H, Zhou X, Lu J, Gao H. MCT4 Promotes Hepatocellular Carcinoma Progression by Upregulating TRAPPC5 Gene. J Hepatocell Carcinoma 2022; 9:289-300. [PMID: 35425722 PMCID: PMC9005128 DOI: 10.2147/jhc.s352948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/30/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Zheyu Niu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Faji Yang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Hongguang Li
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Jianlu Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Qingqiang Ni
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Chaoqun Ma
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Huaqiang Zhu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Hong Chang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Xu Zhou
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Jun Lu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- Correspondence: Jun Lu; Hengjun Gao, Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital affiliated to Shandong University, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China, Email ;
| | - Hengjun Gao
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
16
|
Sequential Drug Delivery in Targeted Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14030573. [PMID: 35335949 PMCID: PMC8949551 DOI: 10.3390/pharmaceutics14030573] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer is a major public health problem and one of the leading causes of death. However, traditional cancer therapy may damage normal cells and cause side effects. Many targeted drug delivery platforms have been developed to overcome the limitations of the free form of therapeutics and biological barriers. The commonly used cancer cell surface targets are CD44, matrix metalloproteinase-2, folate receptors, etc. Once the drug enters the cell, active delivery of the drug molecule to its final destination is still preferred. The subcellular targeting strategies include using glucocorticoid receptors for nuclear targeting, negative mitochondrial membrane potential and N-acetylgalactosaminyltransferase for Golgi apparatus targeting, etc. Therefore, the most effective way to deliver therapeutic agents is through a sequential drug delivery system that simultaneously achieves cellular- and subcellular-level targeting. The dual-targeting delivery holds great promise for improving therapeutic effects and overcoming drug resistance. This review classifies sequential drug delivery systems based on final targeted organelles. We summarize different targeting strategies and mechanisms and gave examples of each case.
Collapse
|
17
|
Cerrato CP, Langel Ü. An update on cell-penetrating peptides with intracellular organelle targeting. Expert Opin Drug Deliv 2022; 19:133-146. [PMID: 35086398 DOI: 10.1080/17425247.2022.2034784] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Cell-penetrating peptide (CPP) technologies represent an important strategy to address drug delivery to specific intracellular compartments by covalent conjugation to targeting sequences, potentially enabling strategies to combat most diseases. AREAS COVERED This updated review article provides an overview of current intracellular organelle targeting by CPP. The targeting strategies of CPP and CPP/cargo complexes to specific cells or intracellular organelles are summarized, and the review provides an update on the current data for their pharmacological and therapeutical applications. EXPERT OPINION Targeted drug delivery is moving from the level of tissue or specific pathogenic cell to the level of specific organelle that is the target of the drug, an important aspect in drug design and development. Organelle-targeted drug delivery results in improved efficacy, ability to control mode of action, reduction of undesired toxicities and side effects, and possibility to overcome drug resistance mechanisms.
Collapse
Affiliation(s)
| | - Ülo Langel
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.,Laboratory of Molecular Biotechnology, Institute of Technology, University of Tartu, Tartu, Estonia
| |
Collapse
|
18
|
Rong X, Liu C, Li M, Zhu H, Zhang Y, Su M, Wang X, Li X, Wang K, Yu M, Sheng W, Zhu B. An Integrated Fluorescent Probe for Ratiometric Detection of Glutathione in the Golgi Apparatus and Activated Organelle-Targeted Therapy. Anal Chem 2021; 93:16105-16112. [PMID: 34797641 DOI: 10.1021/acs.analchem.1c03836] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cancer is a serious threat to human health, and there is an urgent need to develop new treatment methods to overcome it. Organelle targeting therapy, as a highly effective and less toxic side effect treatment strategy, has great research significance and development prospects. Being an essential organelle, the Golgi apparatus plays a particularly major role in the growth of cancer cells. Acting as an indispensable and highly expressed antioxidant in cancer cells, glutathione (GSH) also contributes greatly during the Golgi oxidative stress. Therefore, it counts for much to track the changes of GSH concentration in Golgi for monitoring the occurrence and development of tumor cells, and exploring Golgi-targeted therapy is also extremely important for effective treatment of cancer. In this work, we designed and synthesized a simple Golgi-targeting fluorescent probe GT-GSH for accurately detecting GSH. The probe GT-GSH reacting with GSH decomposes toxic substances to Golgi, thereby killing cancer cells. At the same time, the ratiometric fluorescent probe can detect the concentration changes of GSH in Golgi stress with high sensitivity and selectivity in living cells. Therefore, such a GSH-responsive fluorescent probe with a Golgi-targeted therapy effect gives a new method for accurate treatment of cancer.
Collapse
Affiliation(s)
- Xiaodi Rong
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Caiyun Liu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Mingzhu Li
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Hanchuang Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Yan Zhang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Meijun Su
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Xin Wang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Xiwei Li
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Kun Wang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Miaohui Yu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Baocun Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| |
Collapse
|
19
|
Li RS, Liu J, Shi H, Hu PP, Wang Y, Gao PF, Wang J, Jia M, Li H, Li YF, Mao C, Li N, Huang CZ. Transformable Helical Self-Assembly for Cancerous Golgi Apparatus Disruption. NANO LETTERS 2021; 21:8455-8465. [PMID: 34569805 DOI: 10.1021/acs.nanolett.1c03112] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Golgi apparatus is a major subcellular organelle responsible for drug resistance. Golgi apparatus-targeted nanomechanical disruption provides an attractive approach for killing cancer cells by multimodal mechanism and avoiding drug resistance. Inspired by the poisonous twisted fibrils in Alzheimer's brain tissue and enhanced rigidity of helical structure in nature, we designed transformable peptide C6RVRRF4KY that can self-assemble into nontoxic nanoparticles in aqueous medium but transformed into left-handed helical fibrils (L-HFs) after targeting and furin cleavage in the Golgi apparatus of cancer cells. The L-HFs can mechanically disrupt the Golgi apparatus membrane, resulting in inhibition of cytokine secretion, collapse of the cellular structure, and eventually death of cancer cells. Repeated stimulation of the cancers by the precursors causes no acquired drug resistance, showing that mechanical disruption of subcellular organelle is an excellent strategy for cancer therapy without drug resistance. This nanomechanical disruption concept should also be applicable to multidrug-resistant bacteria and viruses.
Collapse
Affiliation(s)
- Rong Sheng Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Institute of Analytical Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Jiahui Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Hu Shi
- School of Chemistry and Chemical Engineering and Institute of Molecular Science, Shanxi University, Taiyuan 030006, P.R. China
| | - Ping Ping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P.R. China
| | - Yao Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Peng Fei Gao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Jian Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| | - Moye Jia
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Hongwei Li
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Yuan Fang Li
- Key Laboratory of Luminescence and Real-Time Analytical System, Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P.R. China
| | - Chengde Mao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907 United States
| | - Na Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Institute of Analytical Chemistry, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Cheng Zhi Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, P.R. China
| |
Collapse
|
20
|
Zhang L, Qu Z, Song A, Yang J, Yu J, Zhang W, Zhuang C. Garlic oil blocks tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis by inducing phase II drug-metabolizing enzymes. Food Chem Toxicol 2021; 157:112581. [PMID: 34562529 DOI: 10.1016/j.fct.2021.112581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/03/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer caused one-quarter of all cancer deaths that was more than other cancers. Chemoprevention is a potential strategy to reducing lung cancer incidence and death, and the effective chemopreventive agents are needed. We investigated the efficacy and mechanism of garlic oil (GO), the garlic product, in the chemoprevention of tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung cancer in A/J mice and MRC-5 cell models in the present study. As a result, it was demonstrated that GO significantly inhibited the NNK-induced lung cancer in vivo and protected MRC-5 cells from NNK-induced cell damage. GO could induce the expressions of the phase II drug-metabolizing enzymes, including NAD(P)H: quinone oxidoreductase 1 (NQO-1), glutathione S-transferase alpha 1 (GSTA1), and antioxidative enzymes heme oxygenase-1 (HO-1). These results supported the potential of GO as a novel candidate agent for the chemoprevention of tobacco carcinogens induced lung cancer.
Collapse
Affiliation(s)
- Lei Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Zhuo Qu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Aiwei Song
- Montverde Academy Shanghai, 508 South Hanqing Road, Shanghai, 201201, China
| | - Jianhong Yang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Wannian Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| | - Chunlin Zhuang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
21
|
Shiwani HA, Elfaki MY, Memon D, Ali S, Aziz A, Egom EE. Updates on sphingolipids: Spotlight on retinopathy. Biomed Pharmacother 2021; 143:112197. [PMID: 34560541 DOI: 10.1016/j.biopha.2021.112197] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 02/05/2023] Open
Abstract
The sphingolipids ceramide (Cer), ceramide-1-phosphate (C1P), sphingosine (Sph), and sphingosine-1-phosphate (S1P)) are key signaling molecules that regulate many patho-biological processes. During the last decade, they have gained increasing attention since they may participate in important and numerous retinal processes, such as neuronal survival and death, proliferation and migration of neuronal and vascular cells, inflammation, and neovascularization. Cer for instance has emerged as a key mediator of inflammation and death of neuronal and retinal pigment epithelium cells in experimental models of retinopathies such as glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa. S1P may have opposite biological actions, preventing photoreceptor and ganglion cell degeneration but also promoting inflammation, fibrosis, and neovascularization in AMD, glaucoma, and pro-fibrotic disorders. Alterations in Cer, S1P, and ceramide 1- phosphate may also contribute to uveitis. Furthermore, use of inhibitors that either prevent Cer increase or modulate S1P signaling, such as Myriocin, desipramine, and Fingolimod (FTY720), have been shown to preserve neuronal viability and retinal function. Collectively, the expanding role for these sphingolipids in the modulation of vital processes in retina cell types and in their dysregulation in retinal degenerations makes them attractive therapeutic targets.
Collapse
Affiliation(s)
- Haaris A Shiwani
- Department of Ophthalmology, Royal Preston Hospital, United Kingdom.
| | | | - Danyal Memon
- Department of Cardiology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Suhayb Ali
- Department of Acute Medicine, Ulster Hospital, Belfast, United Kingdom
| | - Abdul Aziz
- Department of Respiratory Medicine, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - Emmanuel E Egom
- Institut du Savoir Montfort (ISM), Hôpital Montfort, University of Ottawa, Ottawa, ON, Canada; Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon.
| |
Collapse
|
22
|
Zhan Z, Liu Z, Lai J, Zhang C, Chen Y, Huang H. Anticancer Effects and Mechanisms of OSW-1 Isolated From Ornithogalum saundersiae: A Review. Front Oncol 2021; 11:747718. [PMID: 34631585 PMCID: PMC8496766 DOI: 10.3389/fonc.2021.747718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/06/2021] [Indexed: 01/07/2023] Open
Abstract
For centuries, cancer has been a lingering dark cloud floating on people's heads. With rapid population growth and aging worldwide, cancer incidence and mortality are growing rapidly. Despite major advances in oncotherapy including surgery, radiation and chemical therapy, as well as immunotherapy and targeted therapy, cancer is expected be the leading cause of premature death in this century. Nowadays, natural compounds with potential anticancer effects have become an indispensable natural treasure for discovering clinically useful agents and made remarkable achievements in cancer chemotherapy. In this regards, OSW-1, which was isolated from the bulbs of Ornithogalum saundersiae in 1992, has exhibited powerful anticancer activities in various cancers. However, after almost three decades, OSW-1 is still far from becoming a real anticancer agent for its anticancer mechanisms remain unclear. Therefore, in this review we summarize the available evidence on the anticancer effects and mechanisms of OSW-1 in vitro and in vivo, and some insights for researchers who are interested in OSW-1 as a potential anticancer drug. We conclude that OSW-1 is a potential candidate for anticancer drugs and deserves further study.
Collapse
Affiliation(s)
| | | | | | | | - Yong Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Haiyan Huang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Choi K, Yang A, Baek J, Jeong H, Kang Y, Baek W, Kim JC, Kang M, Choi M, Ham Y, Son MJ, Han SB, Kim J, Jang JH, Ahn JS, Shen H, Woo SH, Kim JH, Cho S. Regulation of Survival Motor Neuron Gene Expression by Calcium Signaling. Int J Mol Sci 2021; 22:ijms221910234. [PMID: 34638572 PMCID: PMC8508836 DOI: 10.3390/ijms221910234] [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: 07/28/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 11/21/2022] Open
Abstract
Spinal muscular atrophy (SMA) is caused by homozygous survival of motor neurons 1 (SMN1) gene deletion, leaving a duplicate gene, SMN2, as the sole source of SMN protein. However, a defect in SMN2 splicing, involving exon 7 skipping, results in a low level of functional SMN protein. Therefore, the upregulation of SMN protein expression from the SMN2 gene is generally considered to be one of the best therapeutic strategies to treat SMA. Most of the SMA drug discovery is based on synthetic compounds, and very few natural compounds have been explored thus far. Here, we performed an unbiased mechanism-independent and image-based screen of a library of microbial metabolites in SMA fibroblasts using an SMN-specific immunoassay. In doing so, we identified brefeldin A (BFA), a well-known inhibitor of ER-Golgi protein trafficking, as a strong inducer of SMN protein. The profound increase in SMN protein was attributed to, in part, the rescue of the SMN2 pre-mRNA splicing defect. Intriguingly, BFA increased the intracellular calcium concentration, and the BFA-induced exon 7 inclusion of SMN2 splicing, was abrogated by the depletion of intracellular calcium and by the pharmacological inhibition of calcium/calmodulin-dependent kinases (CaMKs). Moreover, BFA considerably reduced the expression of Tra2-β and SRSF9 proteins in SMA fibroblasts and enhanced the binding of PSF and hnRNP M to an exonic splicing enhancer (ESE) of exon 7. Together, our results demonstrate a significant role for calcium and its signaling on the regulation of SMN splicing, probably through modulating the expression/activity of splicing factors.
Collapse
Affiliation(s)
- Kwangman Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- Department of Medical Biotechnology, SoonChunHyang University, Asan 31538, Korea
| | - Ansook Yang
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea;
| | - Jiyeon Baek
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea;
| | - Hyejeong Jeong
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea;
| | - Yura Kang
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea; (Y.K.); (W.B.)
- Cancer Molecular Biology Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Woosun Baek
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea; (Y.K.); (W.B.)
- Cancer Molecular Biology Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Joon-Chul Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.-C.K.); (M.-J.S.)
| | - Mingu Kang
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea;
| | - Miri Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea;
| | - Youngwook Ham
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea; (J.-H.J.); (J.S.A.)
| | - Min-Jeong Son
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.-C.K.); (M.-J.S.)
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea;
| | - Janghwan Kim
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Jae-Hyuk Jang
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea; (J.-H.J.); (J.S.A.)
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea
| | - Jong Seog Ahn
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea; (J.-H.J.); (J.S.A.)
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea
| | - Haihong Shen
- Gwangju Institute of Science and Technology, School of life Sciences, Gwangju 61005, Korea;
| | - Sun-Hee Woo
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.-C.K.); (M.-J.S.)
- Correspondence: (S.-H.W.); (J.H.K.); (S.C.); Tel.: +82-42-821-5924 (S.-H.W.); +82-31-920-2204 (J.H.K.); +82-43-240-6105 (S.C.); Fax: +82-42-823-6566 (S.-H.W.); +82-31-920-2006 (J.H.K.); +82-43-240-6159 (S.C)
| | - Jong Heon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Korea; (Y.K.); (W.B.)
- Cancer Molecular Biology Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Korea
- Correspondence: (S.-H.W.); (J.H.K.); (S.C.); Tel.: +82-42-821-5924 (S.-H.W.); +82-31-920-2204 (J.H.K.); +82-43-240-6105 (S.C.); Fax: +82-42-823-6566 (S.-H.W.); +82-31-920-2006 (J.H.K.); +82-43-240-6159 (S.C)
| | - Sungchan Cho
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Korea; (K.C.); (A.Y.); (J.B.); (H.J.); (M.K.); (M.C.); (Y.H.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea; (J.-H.J.); (J.S.A.)
- Correspondence: (S.-H.W.); (J.H.K.); (S.C.); Tel.: +82-42-821-5924 (S.-H.W.); +82-31-920-2204 (J.H.K.); +82-43-240-6105 (S.C.); Fax: +82-42-823-6566 (S.-H.W.); +82-31-920-2006 (J.H.K.); +82-43-240-6159 (S.C)
| |
Collapse
|
24
|
Qiao L, Sinha S, El-hafeez AAA, Lo I, Midde KK, Ngo T, Aznar N, Lopez-sanchez I, Gupta V, Farquhar MG, Rangamani P, Ghosh P. A Circuit for Secretion-coupled Cellular Autonomy in Multicellular Eukaryotes.. [DOI: 10.1101/2021.03.18.436048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
ABSTRACTCancers represent complex autonomous systems, displaying self-sufficiency in growth signaling. Autonomous growth is fueled by a cancer cell’s ability to ‘secrete-and-sense’ growth factors: a poorly understood phenomenon. Using an integrated systems and experimental approach, here we dissect the impact of a feedback-coupled GTPase circuit within the secretory pathway that imparts secretion-coupled autonomy. The circuit is assembled when the Ras-superfamily monomeric GTPase Arf1, and the heterotrimeric GTPase Giαβγ and their corresponding GAPs and GEFs are coupled by GIV/Girdin, a protein that is known to fuel aggressive traits in diverse cancers. One forward and two key negative feedback loops within the circuit create closed-loop control (CLC), allow the two GTPases to coregulate each other, and convert the expected switch-like behavior of Arf1-dependent secretion into an unexpected dose response alignment behavior of sensing and secretion. Such behavior translates into cell survival that is self-sustained by stimulus-proportionate secretion. Proteomic studies and protein-protein interaction network analyses pinpoint growth factors (e.g., the epidermal growth factor; EGF) as a key stimuli for such self-sustenance. Findings highlight how enhanced coupling of two biological switches in cancer cells is critical for multiscale feedback control to achieve secretion-coupled autonomy of growth factors.SYNOPSIS IMAGESTANDFIRST TEXTThis work defines the inner workings of a Golgi-localized molecular circuitry comprised of coupled GTPases, which empowers cells to achieve self-sufficiency in growth factor signaling by creating a secrete-and-sense autocrine loop.HIGHLIGHTS/MAIN FINDINGSModeling and experimental approaches were used to dissect a coupled GTPase circuit.Coupling enables closed loop feedback and mutual control of GTPases.Coupling generates dose response alignment behavior of sensing and secretion of growth factors.Coupling is critical for multiscale feedback control to achieve secretion-coupled autonomy.
Collapse
|
25
|
Andraos C, Gulumian M. Intracellular and extracellular targets as mechanisms of cancer therapy by nanomaterials in relation to their physicochemical properties. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1680. [PMID: 33111484 PMCID: PMC7988657 DOI: 10.1002/wnan.1680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022]
Abstract
Cancer nanomedicine has evolved in recent years and is only expected to increase due to the ease with which nanomaterials (NMs) may be manipulated to the advantage of the cancer patient. The success of nanomedicine is dependent on the cell death mechanism, which in turn is dependent on the organelle initially targeted. The success of cancer nanomedicine is also dependent on other cellular mechanisms such as the induction of autophagy dysfunction, manipulation of the tumor microenvironment (TME) and secretome or induction of host immune responses. Current cancer phototherapies for example, photothermal- or photodynamic therapies as well as radio enhancement also form a major part of cancer nanomedicine. In general, cancer nanomedicine may be grouped into those NMs exhibiting inherent anti-cancer properties that is, self-therapeutic NMs (Group 1), NMs leading to localization of phototherapies or radio-enhancement (Group 2), and NMs as nanocarriers in the absence or presence of external radiation (Group 3). The recent advances of these three groups, together with their advantages and disadvantages as well as their cellular mechanisms and ultimate outcomes are summarized in this review. By exploiting these different intracellular mechanisms involved in initiating cell death pathways, it is possible to synthesize NMs that may have the desirable characteristics to maximize their efficacy in cancer therapy. Therefore, a summary of these important physicochemical characteristics is also presented that need to be considered for optimal cancer cell targeting and initiation of mechanisms that will lead to cancerous cell death. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Charlene Andraos
- Toxicology DepartmentNational Institute for Occupational HealthJohannesburgSouth Africa
| | - Mary Gulumian
- Toxicology DepartmentNational Institute for Occupational HealthJohannesburgSouth Africa
- Haematology and Molecular Medicine DepartmentUniversity of the WitwatersrandJohannesburgSouth Africa
- Water Research Group, Unit for Environmental Sciences and ManagementNorth West UniversityPotchefstroomSouth Africa
| |
Collapse
|
26
|
Khan AA, Allemailem KS, Almatroudi A, Almatroodi SA, Alsahli MA, Rahmani AH. Novel strategies of third level (Organelle-specific) drug targeting: An innovative approach of modern therapeutics. J Drug Deliv Sci Technol 2021; 61:102315. [DOI: 10.1016/j.jddst.2020.102315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Choi NE, Lee JY, Park EC, Lee JH, Lee J. Recent Advances in Organelle-Targeted Fluorescent Probes. Molecules 2021; 26:E217. [PMID: 33406634 PMCID: PMC7795030 DOI: 10.3390/molecules26010217] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/27/2022] Open
Abstract
Recent advances in fluorescence imaging techniques and super-resolution microscopy have extended the applications of fluorescent probes in studying various cellular processes at the molecular level. Specifically, organelle-targeted probes have been commonly used to detect cellular metabolites and transient chemical messengers with high precision and have become invaluable tools to study biochemical pathways. Moreover, several recent studies reported various labeling strategies and novel chemical scaffolds to enhance target specificity and responsiveness. In this review, we will survey the most recent reports of organelle-targeted fluorescent probes and assess their general strategies and structural features on the basis of their target organelles. We will discuss the advantages of the currently used probes and the potential challenges in their application as well as future directions.
Collapse
Affiliation(s)
| | | | | | | | - Jiyoun Lee
- Department of Next-Generation Applied Science, and Global Medical Science, Sungshin University, Seoul 01133, Korea; (N.-E.C.); (J.-Y.L.); (E.-C.P.); (J.-H.L.)
| |
Collapse
|
28
|
Lenkavska L, Tomkova S, Horvath D, Huntosova V. Searching for combination therapy by clustering methods: Stimulation of PKC in Golgi apparatus combined with hypericin induced PDT. Photodiagnosis Photodyn Ther 2020; 31:101813. [PMID: 32442674 DOI: 10.1016/j.pdpdt.2020.101813] [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: 03/16/2020] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
Cancer cell metabolism is a very attractive target for anticancer treatments. This work focuses on protein kinase C (PKC) signaling in the U87 MG glioma. By means of western blot, fluorescence and time-resolved fluorescence microscopy the correlation between the Golgi apparatus (GA), lysosomes and mitochondria were evaluated. The known regulators of PKC were applied to cancer cells. Phorbol myristate acetate (PMA) was chosen as the activator of PKC. Gö6976, hypericin and rottlerin, the inhibitors of PKCα and PKCδ were selected as well. Stabilization, destabilization processes occurring in cells allow classification of observations into several groups. Multiple versions of hierarchical cluster analysis have been applied and similarities have been found between organelles and PKC regulators. The method identified GA as an extraordinary organelle whose functionality is significantly influenced by PKC regulators as well as oxidative stress. Therefore, combination therapy has been designed according to the results of the cluster analysis. Furthermore, the efficacy of photodynamic therapy mediated by hypericin, and the consequent apoptosis, was significantly increased during the treatment. To our knowledge, this is the first demonstration of the effectiveness of the clustering in the given area.
Collapse
Affiliation(s)
- Lenka Lenkavska
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, 041 54, Kosice, Slovakia.
| | - Silvia Tomkova
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, 041 54, Kosice, Slovakia.
| | - Denis Horvath
- Center for Interdisciplinary Biosciences, Technology and Innovation Park, P.J. Safarik University in Kosice, 041 54, Kosice, Slovakia.
| | - Veronika Huntosova
- Center for Interdisciplinary Biosciences, Technology and Innovation Park, P.J. Safarik University in Kosice, 041 54, Kosice, Slovakia.
| |
Collapse
|
29
|
He Z, Zhang Y, Khan AR, Ji J, Yu A, Zhai G. A novel progress of drug delivery system for organelle targeting in tumour cells. J Drug Target 2020; 29:12-28. [PMID: 32698651 DOI: 10.1080/1061186x.2020.1797051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
At present, malignant tumours have become one of the most serious diseases that endanger human health. According to a survey on causes of death in Chinese population in early 1990s, the malignant tumours were the second leading cause of death. In the treatment of tumours, the ideal situation is that drugs should target and accumulate at tumour sites and destroy tumour cells specifically, without affecting normal cells and stem cells with regenerative capacity. This requires drugs to be specifically transported to the target organs, tissues, cells, and even specific organelles, like mitochondria, nuclei, lysosomes, endoplasmic reticulum (ER), and Golgi apparatus (GA). The nano drug delivery system can not only protect drugs from degradation but also facilitate functional modification and targeted drug delivery to the tumour site. This article mainly reviews the targeting of nano drug delivery systems to tumour cytoplasmic matrix, nucleus, mitochondria, ER, and lysosomes. Organelle-specific drug delivery system will be a major mean of targeting drug delivery with lower toxicity, less dosage and higher drug concentration in tumour cells.
Collapse
Affiliation(s)
- Zhijing He
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Yanan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Abdur Rauf Khan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Aihua Yu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
30
|
Zhu H, Liu C, Liang C, Tian B, Zhang H, Zhang X, Sheng W, Yu Y, Huang S, Zhu B. A new phenylsulfonamide-based Golgi-targeting fluorescent probe for H 2S and its bioimaging applications in living cells and zebrafish. Chem Commun (Camb) 2020; 56:4086-4089. [PMID: 32162641 DOI: 10.1039/d0cc00282h] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have synthesized a simple Golgi-targeting H2S fluorescent probe which can detect endogenous and exogenous H2S in cells and zebrafish. In addition, this probe provides a new chemical tool for the detailed study of generation pathways of H2S under Golgi stress response.
Collapse
Affiliation(s)
- Hanchuang Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhu H, Liang C, Cai X, Zhang H, Liu C, Jia P, Li Z, Yu Y, Zhang X, Sheng W, Zhu B. Rational Design of a Targetable Fluorescent Probe for Visualizing H2S Production under Golgi Stress Response Elicited by Monensin. Anal Chem 2019; 92:1883-1889. [DOI: 10.1021/acs.analchem.9b04009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Hanchuang Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Changxu Liang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Xinyu Cai
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Hanming Zhang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Caiyun Liu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Pan Jia
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Zilu Li
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Yamin Yu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Xue Zhang
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| | - Wenlong Sheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Baocun Zhu
- School of Water Conservancy and Environment, University of Jinan, Jinan 250022, China
| |
Collapse
|
32
|
Harisa GI, Faris TM. Direct Drug Targeting into Intracellular Compartments: Issues, Limitations, and Future Outlook. J Membr Biol 2019; 252:527-539. [PMID: 31375855 DOI: 10.1007/s00232-019-00082-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022]
Abstract
Intracellular compartment drug delivery is a promising strategy for the treatment of diseases. By this way, medicines can delivered to particular intracellular compartments. This maximizes the therapeutic efficacy and safety of medicines, particularly of anticancer and antiviral drugs. Intracellular compartment drug delivery is either indirectly by targeting of cell nucleus as central compartment of the cell or directly through the targeting of compartments itself. Drugs or nanoshuttles labeled with compartment's localization signal represent a smart tactic for subcellular compartment targeting. There are several boundaries prevent the arrival of shuttles to the specified intracellular compartments. These boundaries include selective permeability of biomembranes, efflux transporters, and lysosomes. The utilization of specific ligands during design of drug delivery nanoshuttles permits the targeting of specified intracellular compartment. Therefore drugs targeting could correct the diseases associated organelles. This review highlights the direct targeting of the medicines into subcellular compartment as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.
- Department of Biochemistry, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Tarek M Faris
- Department of Pharmaceutics, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
33
|
Rim DE, Yoo HJ, Lee JH, Kwon OJ, Jeong SW. Role of GS28 in sodium nitroprusside-induced cell death in cervical carcinoma cells. J Biochem Mol Toxicol 2019; 33:e22348. [PMID: 31066958 DOI: 10.1002/jbt.22348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 01/11/2023]
Abstract
Golgi S-nitro-N-acetylpenicillamine receptor complex 1 (GS28) has been implicated in Golgi vesicle transport. We examined the role of GS28 and its molecular mechanisms in sodium nitroprusside (SNP)-induced cell death using GS28 siRNA (siGS28)-transfected HeLa cells. Significant inhibition of cytotoxicity was observed in the cells treated with SNP, and photodegraded SNP showed equal cytotoxicity to SNP. Pretreatment with an ERK inhibitor or siErk1 cotransfection blocked the inhibition in cytotoxicity. Additionally, increased phosphorylation of ERK was maintained in the cells treated with SNP, and Nrf2 level was dependent on ERK phosphorylation. However, pretreatment with a pan-caspase inhibitor had no effect on cytotoxicity or procaspase-3 level. Pretreatment with an autophagy inhibitor or siATG5 cotransfection blocked the inhibition of cytotoxicity. The changes of LC3 corresponded to that in siErk1-cotransfected cells. These data suggest that GS28 has an inductive role in SNP-induced cell death via inhibition of ERK, leading to inhibition of autophagic processes in HeLa cells.
Collapse
Affiliation(s)
- Do Eun Rim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Jae Yoo
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Oh-Joo Kwon
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong-Whan Jeong
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
34
|
Gurram B, Li M, Fan J, Wang J, Peng X. Near-infrared fluorescent probe for fast track of cyclooxygenase-2 in Golgi apparatus in cancer cells. Front Chem Sci Eng 2019. [DOI: 10.1007/s11705-019-1796-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Wodlej C, Riedl S, Rinner B, Leber R, Drechsler C, Voelker DR, Choi JY, Lohner K, Zweytick D. Interaction of two antitumor peptides with membrane lipids - Influence of phosphatidylserine and cholesterol on specificity for melanoma cells. PLoS One 2019; 14:e0211187. [PMID: 30682171 PMCID: PMC6347193 DOI: 10.1371/journal.pone.0211187] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
R-DIM-P-LF11-322 and DIM-LF11-318, derived from the cationic human host defense peptide lactoferricin show antitumor activity against human melanoma. While R-DIM-P-LF11-322 interacts specifically with cancer cells, the non-specific DIM-LF11-318 exhibits as well activity against non-neoplastic cells. Recently we have shown that cancer cells expose the negatively charged lipid phosphatidylserine (PS) in the outer leaflet of the plasma membrane, while non-cancer cells just expose zwitterionic or neutral lipids, such as phosphatidylcholine (PC) or cholesterol. Calorimetric and zeta potential studies with R-DIM-P-LF11-322 and cancer-mimetic liposomes composed of PS, PC and cholesterol indicate that the cancer-specific peptide interacts specifically with PS. Cholesterol, however, reduces the effectiveness of the peptide. The non-specific DIM-LF11-318 interacts with PC and PS. Cholesterol does not affect its interaction. The dependence of activity of R-DIM-P-LF11-322 on the presence of exposed PS was also confirmed in vitro upon PS depletion of the outer leaflet of cancer cells by the enzyme PS-decarboxylase. Further corresponding to model studies, cholesterol depleted melanoma plasma membranes showed increased sensitivity to R-DIM-P-LF11-322, whereas activity of DIM-LF11-318 was unaffected. Microscopic studies using giant unilamellar vesicles and melanoma cells revealed strong changes in lateral distribution and domain formation of lipids upon addition of both peptides. Whereas R-DIM-P-LF11-322 enters the cancer cell specifically via PS and reaches an intracellular organelle, the Golgi, inducing mitochondrial swelling and apoptosis, DIM-LF11-318 kills rapidly and non-specifically by lysis of the plasma membrane. In conclusion, the specific interaction of R-DIM-P-LF11-322 with PS and sensitivity to cholesterol seem to modulate its specificity for cancer membranes.
Collapse
Affiliation(s)
- Christina Wodlej
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Sabrina Riedl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Beate Rinner
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Regina Leber
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Carina Drechsler
- BIOSS and Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg i. Br., Germany
| | - Dennis R Voelker
- Department of Medicine, National Jewish Health, Denver CO, United States of America
| | - Jae-Yeon Choi
- Department of Medicine, National Jewish Health, Denver CO, United States of America
| | - Karl Lohner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Dagmar Zweytick
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
36
|
Kendrick-Williams LL, Harth E. Second-Generation Nanosponges: Nanonetworks in Controlled Dimensions via Backbone Ketoxime and Alkoxyamine Cross-Links for Controlled Release. Macromolecules 2018. [DOI: 10.1021/acs.macromol.8b02244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Laken L. Kendrick-Williams
- Department of Chemistry, Center of Excellence in Polymer Chemistry (CEPC), University of Houston, 3585 Cullen Blvd., Houston, Texas 77030, United States
- Department of Chemistry, Vanderbilt University, 7665 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Eva Harth
- Department of Chemistry, Center of Excellence in Polymer Chemistry (CEPC), University of Houston, 3585 Cullen Blvd., Houston, Texas 77030, United States
| |
Collapse
|
37
|
Qin H, Li W, Sun Y, Bao Y, Sun L, Song Z, Zheng L, Zhao Y, Li Y. 20(S)-25-methoxyl-dammarane-3β,12β,20-triol attenuates endoplasmic reticulum stress via ERK/MAPK signaling pathway. Eur J Pharmacol 2018; 836:75-82. [DOI: 10.1016/j.ejphar.2018.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 11/27/2022]
|
38
|
Weissmann M, Bhattacharya U, Feld S, Hammond E, Ilan N, Vlodavsky I. The heparanase inhibitor PG545 is a potent anti-lymphoma drug: Mode of action. Matrix Biol 2018; 77:58-72. [PMID: 30096360 DOI: 10.1016/j.matbio.2018.08.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/17/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022]
Abstract
It is now well recognized that heparanase, an endo-β-D-glucuronidase capable of cleaving heparan sulfate (HS) side chains at a limited number of sites, promotes tumorigenesis by diverse mechanisms. Compelling evidence strongly implies that heparanase is a viable target for cancer therapy, thus encouraging the development of heparanase inhibitors as anti-cancer therapeutics. Here, we examined the efficacy and mode of action of PG545, an HS-mimetic heparanase inhibitor, in human lymphoma. We found that PG545 exhibits a strong anti-lymphoma effect, eliciting lymphoma cell apoptosis. Notably, this anti-lymphoma effect involves ER stress response that was accompanied by increased autophagy. The persistent ER stress evoked by PG545 is held responsible for cell apoptosis because apoptotic cell death was attenuated by an inhibitor of PERK, a molecular effector of ER stress. Importantly, PG545 had no such apoptotic effect on naïve splenocytes, further encouraging the development of this compound as anti-lymphoma drug. Surprisingly, we found that PG545 also elicits apoptosis in lymphoma cells that are devoid of heparanase activity (i.e., Raji), indicating that the drug also exerts heparanase-independent function(s) that together underlie the high potency of PG545 in preclinical cancer models.
Collapse
Affiliation(s)
- Marina Weissmann
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Udayan Bhattacharya
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Sari Feld
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | - Neta Ilan
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
39
|
Luchsinger C, Aguilar M, Burgos PV, Ehrenfeld P, Mardones GA. Functional disruption of the Golgi apparatus protein ARF1 sensitizes MDA-MB-231 breast cancer cells to the antitumor drugs Actinomycin D and Vinblastine through ERK and AKT signaling. PLoS One 2018; 13:e0195401. [PMID: 29614107 PMCID: PMC5882166 DOI: 10.1371/journal.pone.0195401] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/21/2018] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence indicates that the Golgi apparatus plays active roles in cancer, but a comprehensive understanding of its functions in the oncogenic transformation has not yet emerged. At the same time, the Golgi is becoming well recognized as a hub that integrates its functions of protein and lipid biosynthesis to signal transduction for cell proliferation and migration in cancer cells. Nevertheless, the active function of the Golgi apparatus in cancer cells has not been fully evaluated as a target for combined treatment. Here, we analyzed the effect of perturbing the Golgi apparatus on the sensitivity of the MDA-MB-231 breast cancer cell line to the drugs Actinomycin D and Vinblastine. We disrupted the function of ARF1, a protein necessary for the homeostasis of the Golgi apparatus. We found that the expression of the ARF1-Q71L mutant increased the sensitivity of MDA-MB-231 cells to both Actinomycin D and Vinblastine, resulting in decreased cell proliferation and cell migration, as well as in increased apoptosis. Likewise, the combined treatment of cells with Actinomycin D or Vinblastine and Brefeldin A or Golgicide A, two disrupting agents of the ARF1 function, resulted in similar effects on cell proliferation, cell migration and apoptosis. Interestingly, each combined treatment had distinct effects on ERK1/2 and AKT signaling, as indicated by the decreased levels of either phospho-ERK1/2 or phospho-AKT. Our results suggest that disruption of Golgi function could be used as a strategy for the sensitization of cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Charlotte Luchsinger
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Marcelo Aguilar
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Patricia V. Burgos
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Center for Cell Biology and Biomedicine (CEBICEM), School of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela Ehrenfeld
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Department of Anatomy, Histology and Pathology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Gonzalo A. Mardones
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Center for Cell Biology and Biomedicine (CEBICEM), School of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- * E-mail:
| |
Collapse
|
40
|
An integrated approach to infer cross-talks between intracellular protein transport and signaling pathways. BMC Bioinformatics 2018. [PMID: 29536825 PMCID: PMC5850946 DOI: 10.1186/s12859-018-2036-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background The endomembrane system, known as secretory pathway, is responsible for the synthesis and transport of protein molecules in cells. Therefore, genes involved in the secretory pathway are essential for the cellular development and function. Recent scientific investigations show that ER and Golgi apparatus may provide a convenient drug target for cancer therapy. On the other hand, it is known that abundantly expressed genes in different cellular organelles share interconnected pathways and co-regulate each other activities. The cross-talks among these genes play an important role in signaling pathways, associated to the regulation of intracellular protein transport. Results In the present study, we device an integrated approach to understand these complex interactions. We analyze gene perturbation expression profiles, reconstruct a directed gene interaction network and decipher the regulatory interactions among genes involved in protein transport signaling. In particular, we focus on expression signatures of genes involved in the secretory pathway of MCF7 breast cancer cell line. Furthermore, network biology analysis delineates these gene-centric cross-talks at the level of specific modules/sub-networks, corresponding to different signaling pathways. Conclusions We elucidate the regulatory connections between genes constituting signaling pathways such as PI3K-Akt, Ras, Rap1, calcium, JAK-STAT, EFGR and FGFR signaling. Interestingly, we determine some key regulatory cross-talks between signaling pathways (PI3K-Akt signaling and Ras signaling pathway) and intracellular protein transport. Electronic supplementary material The online version of this article (10.1186/s12859-018-2036-2) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Lu XS, Qiao YB, Li Y, Yang B, Chen MB, Xing CG. Preclinical study of cinobufagin as a promising anti-colorectal cancer agent. Oncotarget 2018; 8:988-998. [PMID: 27894091 PMCID: PMC5352212 DOI: 10.18632/oncotarget.13519] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 10/28/2016] [Indexed: 12/21/2022] Open
Abstract
Here, we assessed the anti-colorectal cancer (CRC) cell activity of cinobufagin (CBG). We found that CBG exerted potent cytotoxic and anti-proliferative activity against CRC lines (HCT-116 and HT-29) and primary human CRC cells. Meanwhile, it activated apoptosis, and disrupted cell-cycle progression in the cells. At the signaling level, CBG treatment in CRC cells provoked endoplasmic reticulum stress (ER stress), the latter was evidenced by caspase-12 activation, CHOP expression, as well as PERK and IRE1 phosphorylations. Contrarily, the ER stress inhibitor salubrinal, the caspase-12 inhibitor and CHOP shRNA remarkably attenuated CBG-induced CRC cell death and apoptosis. Further, CBG in-activated mammalian target or rapamycin complex 1 (mTORC1), which appeared responsible for proliferation inhibition in CRC cells. Introduction of a constitutively-active S6K1 (“ca-S6K1”) restored proliferation of CBG-treated CRC cells. Finally, CBG intraperitoneal injection suppressed HCT-116 xenograft tumor growth in the nude mice. CHOP upregulation and mTORC1 in-activation were also noticed in CBG-treated HCT-116 tumors. The results of this preclinical study suggest that CBG could be tested as promising anti-CRC agent.
Collapse
Affiliation(s)
- Xing-Sheng Lu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of General Surgery, Suzhou Municipal Hospital, Suzhou, China
| | - Yin-Biao Qiao
- Department of Hepatobiliary Surgery, The Third Hospital Affiliated to Soochow University, Changzhou City, Jiangsu, China
| | - Ya Li
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bo Yang
- Department of Hepatobiliary Surgery, The Third Hospital Affiliated to Soochow University, Changzhou City, Jiangsu, China
| | - Min-Bin Chen
- Department of Medical Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Chun-Gen Xing
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
42
|
Wang F, Guan X, Yang J, He W, Wei Y, Chen H, Li Y. Differential Expression and Significance of Endoplasmic Reticulum Golgi Intermediate Compartment 1 in Precancerous Gastric Lesions and Gastric Cancer. Am J Med Sci 2018; 355:228-234. [DOI: 10.1016/j.amjms.2017.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/18/2017] [Accepted: 11/06/2017] [Indexed: 02/06/2023]
|
43
|
Endoplasmic reticulum-Golgi intermediate compartment protein 3 knockdown suppresses lung cancer through endoplasmic reticulum stress-induced autophagy. Oncotarget 2018; 7:65335-65347. [PMID: 27588471 PMCID: PMC5323159 DOI: 10.18632/oncotarget.11678] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 08/08/2016] [Indexed: 12/19/2022] Open
Abstract
Trafficking from the endoplasmic reticulum (ER) to the Golgi apparatus is elevated in cancer cells. Therefore, proteins of the ER-Golgi intermediate compartment (ERGIC) attract significant attention as targets for cancer treatment. Enhanced cancer cell growth and epithelial-mesenchymal transition by ERGICs correlates with poor-prognosis of lung cancer. This prompted us to assess whether knockdown of ERGIC3 may decrease lung cancer growth. To test the hypothesis, the effects of ERGIC3 short hairpin RNA (shERGIC3) on ER stress-induced cell death and lung tumorigenesis were investigated both in vitro and in vivo. Knockdown of ERGIC3 led to ER stress-induced autophagic cell death and suppression of proliferation in the A549 human lung cancer cell-line. Moreover, non-invasive aerosol-delivery of shERGIC3 using the biocompatible carrier glycerol propoxylate triacrylate and spermine (GPT-SPE) inhibited lung tumorigenesis in the K-rasLA1 murine model of lung cancer. Our data suggest that suppression of ERGIC3 could provide a framework for the development of effective lung cancer therapies.
Collapse
|
44
|
Yu RY, Xing L, Cui PF, Qiao JB, He YJ, Chang X, Zhou TJ, Jin QR, Jiang HL, Xiao Y. Regulating the Golgi apparatus by co-delivery of a COX-2 inhibitor and Brefeldin A for suppression of tumor metastasis. Biomater Sci 2018; 6:2144-2155. [DOI: 10.1039/c8bm00381e] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Herein, celecoxib (CLX) and brefeldin A (BFA) were encapsulated into the biocompatible polymer PLGA-PEG to form nanoparticles that act on the Golgi apparatus to treat metastatic breast cancer.
Collapse
|
45
|
Amanullah A, Upadhyay A, Joshi V, Mishra R, Jana NR, Mishra A. Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog Neurobiol 2017; 159:1-38. [DOI: 10.1016/j.pneurobio.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
|
46
|
Li L, Yang W, Yue H, He F, Xu S, Wei Q, Chen P, Peng Q, Deng S, Long P. Expression of NIBP and its clinical significance in human early colorectal cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8633-8639. [PMID: 31966720 PMCID: PMC6965483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 06/20/2017] [Indexed: 06/10/2023]
Abstract
AIM To investigate the expression of NIBP and its clinical significance in early colorectal cancer. PATIENTS AND METHODS With immunohistochemistry, the expression of NIBP was detected in 23 patients of early colorectal cancer tissues, 102 patients of invasive colorectal cancer tissues, 32 patients of adenoma and 20 patients of normal tissues. The relationship between NIBP expression and clinicopathological characteristic of colorectal cancer were also analyzed. RESULT We found that the positive rates of NIBP was higher in early colorectal cancer tissues (82.6%, 19/23) than those in adenomas and normal tissues (x2=29.07, P<0.05), but not significant than those in invasive colorectal cancer (x2=1.79, P>0.05). Positivity for T1N0M0, T2N0M0, II, III and IV was 82.6% (19/23), 80.0% (4/5), 78.0% (32/41), 63.6% (21/33), 56.5% (13/23), respectively. With the increase in TNM stage, the positive rate of NIBP decreased, the positive rate of T1N0M0 is highest than other TNM stages, but no statistically significant (P>0.05). CONCLUSION These results suggested that NIBP is highly expressed in human early colorectal cancer tissues. NIBP might involve in the tumorigenesis and probably serve as a new marker for human early colorectal cancer.
Collapse
Affiliation(s)
- Lifu Li
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Wenjuan Yang
- Department of The Fifth Hepatology, The Ninth Hospital of NanchangJiangxi Province, China
| | - Hui Yue
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Fengjian He
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Shenghao Xu
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Qingzhu Wei
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Peisheng Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Qianqian Peng
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Sanhua Deng
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| | - Peiqi Long
- Department of Gastroenterology, The Third Affiliated Hospital of Southern Medical UniversityNo. 183 Zhongshan Road West, Guangzhou, Guangdong Province, China
| |
Collapse
|
47
|
Ignashkova TI, Gendarme M, Peschk K, Eggenweiler HM, Lindemann RK, Reiling JH. Cell survival and protein secretion associated with Golgi integrity in response to Golgi stress-inducing agents. Traffic 2017; 18:530-544. [PMID: 28485883 DOI: 10.1111/tra.12493] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 12/29/2022]
Abstract
The Golgi apparatus is part of the secretory pathway and of central importance for modification, transport and sorting of proteins and lipids. ADP-ribosylation factors, whose activation can be blocked by brefeldin A (BFA), play a major role in functioning of the Golgi network and regulation of membrane traffic and are also involved in proliferation and migration of cancer cells. Due to high cytotoxicity and poor bioavailability, BFA has not passed the preclinical stage of drug development. Recently, AMF-26 and golgicide A have been described as novel inhibitors of the Golgi system with antitumor or bactericidal properties. We provide here further evidence that AMF-26 closely mirrors the mode of action of BFA but is less potent. Using several human cancer cell lines, we studied the effects of AMF-26, BFA and golgicide A on cell homeostasis including Golgi structure, endoplasmic reticulum (ER) stress markers, secretion and viability, and found overall a significant correlation between these parameters. Furthermore, modulation of ADP-ribosylation factor expression has a profound impact on Golgi organization and survival in response to Golgi stress inducers.
Collapse
Affiliation(s)
- Tatiana I Ignashkova
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Heidelberg, Germany
| | - Mathieu Gendarme
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Heidelberg, Germany
| | - Katrin Peschk
- Medicinal Chemistry, Merck Biopharma, Merck KGaA, Darmstadt, Germany
| | | | - Ralph K Lindemann
- Translational Innovation Platform Oncology, Merck Biopharma, Merck KGaA, Darmstadt, Germany
| | - Jan H Reiling
- Metabolism and Signaling in Cancer, BioMed X Innovation Center, Heidelberg, Germany
| |
Collapse
|
48
|
Battistella C, Klok HA. Controlling and Monitoring Intracellular Delivery of Anticancer Polymer Nanomedicines. Macromol Biosci 2017; 17. [DOI: 10.1002/mabi.201700022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 03/03/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Claudia Battistella
- École Polytechnique Fédérale de Lausanne (EPFL); Institut des Matériaux et Institut des Sciences et Ingénierie Chimiques; Laboratoire des Polymères; Bâtiment MXD; Station 12 CH-1015 Lausanne Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL); Institut des Matériaux et Institut des Sciences et Ingénierie Chimiques; Laboratoire des Polymères; Bâtiment MXD; Station 12 CH-1015 Lausanne Switzerland
| |
Collapse
|
49
|
Misuth M, Joniova J, Horvath D, Dzurova L, Nichtova Z, Novotova M, Miskovsky P, Stroffekova K, Huntosova V. The flashlights on a distinct role of protein kinase C δ: Phosphorylation of regulatory and catalytic domain upon oxidative stress in glioma cells. Cell Signal 2017; 34:11-22. [PMID: 28237688 DOI: 10.1016/j.cellsig.2017.02.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/07/2017] [Accepted: 02/20/2017] [Indexed: 01/02/2023]
Abstract
Glioblastoma multiforme are considered to be aggressive high-grade tumors with poor prognosis for patient survival. Photodynamic therapy is one of the adjuvant therapies which has been used for glioblastoma multiforme during last decade. Hypericin, a photosensitizer, can be employed in this treatment. We have studied the effect of hypericin on PKCδ phosphorylation in U87 MG cells before and after light application. Hypericin increased PKCδ phosphorylation at tyrosine 155 in the regulatory domain and serine 645 in the catalytic domain. However, use of the light resulted in apoptosis, decreased phosphorylation of tyrosine 155 and enhanced serine 645. The PKCδ localization and phosphorylation of regulatory and catalytic domains were shown to play a distinct role in the anti-apoptotic response of glioma cells. We hypothesized that PKCδ phosphorylated at the regulatory domain is primarily present in the cytoplasm and in mitochondria before irradiation, and it may participate in Bcl-2 phosphorylation. After hypericin and light application, PKCδ phosphorylated at a regulatory domain which is in the nucleus. In contrast, PKCδ phosphorylated at the catalytic domain may be mostly active in the nucleus before irradiation, but active in the cytoplasm after the irradiation. In summary, light-induced oxidative stress significantly regulates PKCδ pro-survival and pro-apoptotic activity in glioma cells by its phosphorylation at serine 645 and tyrosine 155.
Collapse
Affiliation(s)
- Matus Misuth
- Department of Biophysics, Faculty of Sciences, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Jaroslava Joniova
- Department of Biophysics, Faculty of Sciences, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Denis Horvath
- Center for Interdisciplinary Biosciences, Faculty of Sciences, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Lenka Dzurova
- Department of Biophysics, Faculty of Sciences, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Zuzana Nichtova
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Science, Bratislava, Slovakia
| | - Marta Novotova
- Department of Muscle Cell Research, Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Science, Bratislava, Slovakia
| | - Pavol Miskovsky
- Center for Interdisciplinary Biosciences, Faculty of Sciences, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia; SAFTRA Photonics Ltd., Jesenna 5, 041 54, Kosice, Slovakia
| | - Katarina Stroffekova
- Department of Biophysics, Faculty of Sciences, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia; Center for Interdisciplinary Biosciences, Faculty of Sciences, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Veronika Huntosova
- Center for Interdisciplinary Biosciences, Faculty of Sciences, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia.
| |
Collapse
|
50
|
Lee SH, Yoo HJ, Rim DE, Cui Y, Lee A, Jung ES, Oh ST, Kim JG, Kwon OJ, Kim SY, Jeong SW. Nuclear Expression of GS28 Protein: A Novel Biomarker that Predicts Prognosis in Colorectal Cancers. Int J Med Sci 2017; 14. [PMID: 28638266 PMCID: PMC5479119 DOI: 10.7150/ijms.19368] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims: GS28 (Golgi SNARE protein, 28 kDa), a member of the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) protein family, plays a critical role in mammalian endoplasmic reticulum (ER)-Golgi or intra-Golgi vesicle transport. To date, few researches on the GS28 protein in human cancer tissues have been reported. In this study, we assessed the prognostic value of GS28 in patients with colorectal cancer (CRC). Methods and results: We screened for GS28 expression using immunohistochemistry in 230 surgical CRC specimens. The CRCs were right-sided and left-sided in 28.3% (65/230) and 71.3% (164/230) of patients, respectively. GS28 staining results were available in 214 cases. Among these, there were 26 nuclear predominant cases and 188 non-nuclear predominant cases. Stromal GS28 expression was noted in 152 cases of CRC. GS28 nuclear predominant immunoreactivity was significantly associated with advanced tumour stage (p = 0.045) and marginally associated with perineural invasion (p = 0.064). Decreased GS28 expression in the stromal cells was significantly associated with lymph node metastasis (N stage; p = 0.036). GS28 expression was not associated with epidermal growth factor receptor (EGFR) immunohistochemical positivity or KRAS mutation status. Investigation of the prognostic value of GS28 with Kaplan-Meier analysis revealed a correlation with overall survival (p = 0.004). Cases with GS28 nuclear predominant expression had significantly poorer overall survival than those with a non-nuclear predominant pattern. Conclusions: Taken together, these results indicate that GS28 nuclear predominant expression could serve as a prognostic marker for CRC and may help in identifying aggressive forms of CRC.
Collapse
Affiliation(s)
- Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Jae Yoo
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Do Eun Rim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yinji Cui
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Taek Oh
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jun Gi Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Oh-Joo Kwon
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Su Young Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong-Whan Jeong
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|