1
|
Law JD, Gao Y, Kovvali S, Thirugnanasambantham P, Wysocki VH, Ahmer BMM, Gopalan V. Identification of inhibitors of the Salmonella FraB deglycase, a drug target. FEBS Open Bio 2025; 15:773-792. [PMID: 39945759 PMCID: PMC12051030 DOI: 10.1002/2211-5463.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/23/2025] [Indexed: 05/06/2025] Open
Abstract
Nontyphoidal Salmonella is one of the most prevalent causes of human foodborne illnesses worldwide, with no narrow-spectrum antibiotics or vaccines available. Here, we seek to address this gap. During the host inflammatory response, Salmonella metabolizes fructose-asparagine as a nutrient using proteins encoded in the fra operon. Deletion of fraB leads to a build-up of 6-phosphofructose-aspartate, the substrate of FraB, and intoxicates Salmonella. Because fra genes are absent in mammals and most members of the human gut microbiome, FraB inhibitors are expected to have limited off-target effects and offer prospects as potential therapeutics. To identify such inhibitors, we conducted a high-throughput screening of small-molecule libraries using a FraB activity-based biochemical assay. We screened 131,165 compounds and identified 126 hits that could be obtained commercially for further characterization. When tested at 25 μm inhibitor in the presence of 1 mm 6-phosphofructose-aspartate, FraB activity was reduced ~ 30-100% by 65 compounds. Guided by preliminary cell-based data, we further characterized six compounds (one triazolidine, two thiadiazolidines, and three triazolothiadiazoles) and found them to exhibit IC50 values from ~ 3 to 100 μm and KI (inhibitor constant) values from ~ 1 to 29 μm. Native mass spectrometry revealed that all three triazolothiadiazoles were capable of binding FraB; we also obtained evidence that one of the triazolothiadiazoles binds FraB even in the presence of substrate. The recurrence of multiple pharmacophores bolsters prospects for farming more hits from compound libraries and for designing therapeutics against nontyphoidal Salmonella.
Collapse
Affiliation(s)
- Jamison D. Law
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOHUSA
| | - Yuan Gao
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOHUSA
- Native Mass Spectrometry Guided Structural Biology CenterThe Ohio State UniversityColumbusOHUSA
| | - Sravya Kovvali
- Department of MicrobiologyThe Ohio State UniversityColumbusOHUSA
| | | | - Vicki H. Wysocki
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOHUSA
- Native Mass Spectrometry Guided Structural Biology CenterThe Ohio State UniversityColumbusOHUSA
- Center for RNA BiologyThe Ohio State UniversityColumbusOHUSA
| | - Brian M. M. Ahmer
- Department of Microbial Infection and ImmunityThe Ohio State UniversityColumbusOHUSA
| | - Venkat Gopalan
- Department of Chemistry and BiochemistryThe Ohio State UniversityColumbusOHUSA
- Center for RNA BiologyThe Ohio State UniversityColumbusOHUSA
| |
Collapse
|
2
|
Diab L, Al Kattar S, Oueini N, Hawi J, Chrabieh A, Dosh L, Jurjus R, Leone A, Jurjus A. Syndecan-1: a key player in health and disease. Immunogenetics 2024; 77:9. [PMID: 39688651 DOI: 10.1007/s00251-024-01366-4] [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: 11/04/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024]
Abstract
Syndecan-1 (SDC-1) is a transmembrane protein localized on the basolateral surface of epithelial cells, encompassing a core protein with heparin sulfate and chondroitin sulfate glycosaminoglycan side chains. SDC-1 is involved in a panoply of cellular mechanisms including cell-to-cell adhesion, extracellular matrix interactions, cell cycle modulation, and lipid clearance. Alterations in the expression and function of SDC-1 are implicated in numerous disease entities, making it an attractive diagnostic and therapeutic target. However, despite its broad involvement in several disease processes, the underlying mechanism contributing to its diverse functions, pathogenesis, and therapeutic uses remains underexplored. Therefore, this review examines the role of SDC-1 in health and disease, focusing on liver pathologies, inflammatory diseases, infectious diseases, and cancer, and sheds light on SDC-1-based therapeutic approaches. Moreover, it delves into the mechanisms through which SDC-1 contributes to these diseases, emphasizing cell-type specific mechanisms. By comprehensively summarizing the significance of SDC-1, its association with several diseases, and its underlying mechanisms of action, the findings of this review could inform future research directions toward the development of targeted therapies and early diagnosis for a multitude of disease entities.
Collapse
Affiliation(s)
- Lara Diab
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Sahar Al Kattar
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Naim Oueini
- Department of Agriculture and Food Engineering, School of Engineering, Holy Spirit University, Kaslik, Jounieh, Lebanon
| | - Jihad Hawi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Antoine Chrabieh
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Laura Dosh
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Rosalyn Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Angelo Leone
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
3
|
Gowda SV, Kim NY, Harsha KB, Gowda D, Suresh RN, Deivasigamani A, Mohan CD, Hui KM, Sethi G, Ahn KS, Rangappa KS. A new 1,2,3-triazole-indirubin hybrid suppresses tumor growth and pulmonary metastasis by mitigating the HGF/c-MET axis in hepatocellular carcinoma. J Adv Res 2024:S2090-1232(24)00377-1. [PMID: 39216686 DOI: 10.1016/j.jare.2024.08.033] [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: 05/01/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a fatal cancer that is often diagnosed at the advanced stages which limits the available therapeutic options. The interaction of HGF with c-MET (a receptor tyrosine kinase) results in the activation of c-MET which subsequently triggers the PI3K/Akt/mTOR axis. Overexpression of c-MET in HCC tissues has been demonstrated to contribute to tumor progression and metastasis. OBJECTIVES We aimed to synthesize triazole-indirubin conjugates, examine their growth suppressor efficacy in cell-based assays, and investigate the antitumor as well as antimetastatic activity of lead cytotoxic agent in the orthotopic mice model. METHODS A series of triazole-indirubin hybrids were synthesized and cytotoxicity, apoptogenic, and antimigratory effect of the lead compound (CRI9) was evaluated using MTT assay, cell cycle analysis, annexin-V/PI assay, TUNEL assay, and wound healing assay. The effect of CRI9 on the operation of the HGF/c-MET/PI3K/Akt/mTOR axis was examined using western blotting and transfection experiments. Acute toxicity, antitumor, and antimetastatic activity of CRI9 were examined in NCr nude mice. The expression of c-MET/PI3K/Akt/mTOR, CD31, and Ki-67 was examined using immunohistochemistry and western blotting. RESULTS Among the new compounds, CRI9 consistently displayed potent cytotoxicity against HGF-induced HCC cells. CRI9 induced apoptosis as evidenced by increased sub G1 cells, annexin-V+/PI+ cells, TUNEL+ cells, and cleavage of procaspase-3 and PARP. CRI9 inhibited HGF-induced phosphorylation of c-METY1234/1235 and subsequently suppressed the PI3K/Akt/mTOR axis. Also, depletion of c-MET or inhibition of c-MET by CRI9 resulted in suppression of the PI3K/Akt/mTOR axis. CRI9 showed no toxic effects in NCr nude mice and displayed a potent antitumor and antimetastatic effect in the orthotopic HCC mice model. CRI9 also reduced the levels of phospho-c-MET, CD31, and Ki-67 and suppressed the activation of the PI3K/Akt/mTOR axis in tumor tissues. CONCLUSION CRI9 has been identified as a new inhibitor of the c-MET/PI3K/Akt/mTOR axis in HCC preclinical models.
Collapse
Affiliation(s)
- Shalini V Gowda
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Kachigere B Harsha
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Darshini Gowda
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Rajaghatta N Suresh
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Chakrabhavi Dhananjaya Mohan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | | |
Collapse
|
4
|
Yu TY, Zhang G, Chai XX, Ren L, Yin DC, Zhang CY. Recent progress on the effect of extracellular matrix on occurrence and progression of breast cancer. Life Sci 2023; 332:122084. [PMID: 37716504 DOI: 10.1016/j.lfs.2023.122084] [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: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer (BC) metastasis is an enormous challenge targeting BC therapy. The extracellular matrix (ECM), the principal component of the BC metastasis niche, is the pivotal driver of breast tumor development, whose biochemical and biophysical characteristics have attracted widespread attention. Here, we review the biological effects of ECM constituents and the influence of ECM stiffness on BC metastasis and drug resistance. We provide an overview of the relative signal transduction mechanisms, existing metastasis models, and targeted drug strategies centered around ECM stiffness. It will shed light on exploring more underlying targets and developing specific drugs aimed at ECM utilizing biomimetic platforms, which are promising for breast cancer treatment.
Collapse
Affiliation(s)
- Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo 315103, Zhejiang, PR China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| |
Collapse
|
5
|
Yuan J, Yang L, Li Z, Zhang H, Wang Q, Huang J, Wang B, Mohan CD, Sethi G, Wang G. The role of the tumor microenvironment in endocrine therapy resistance in hormone receptor-positive breast cancer. Front Endocrinol (Lausanne) 2023; 14:1261283. [PMID: 37900137 PMCID: PMC10611521 DOI: 10.3389/fendo.2023.1261283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Endocrine therapy is the prominent strategy for the treatment of hormone-positive breast cancers. The emergence of resistance to endocrine therapy is a major health concern among hormone-positive breast cancer patients. Resistance to endocrine therapy demands the design of newer therapeutic strategies. The understanding of underlying molecular mechanisms of endocrine resistance, components of the tumor microenvironment (TME), and interaction of resistant breast cancer cells with the cellular/acellular components of the intratumoral environment are essential to formulate new therapeutic strategies for the treatment of endocrine therapy-resistant breast cancers. In the first half of the article, we have discussed the general mechanisms (including mutations in estrogen receptor gene, reregulated activation of signaling pathways, epigenetic changes, and cell cycle alteration) responsible for endocrine therapy resistance in hormone-positive breast cancers. In the latter half, we have emphasized the precise role of cellular (cancer-associated fibroblasts, immune cells, and cancer stem cells) and acellular components (collagen, fibronectin, and laminin) of TME in the development of endocrine resistance in hormone-positive breast cancers. In sum, the article provides an overview of the relationship between endocrine resistance and TME in hormone-positive breast cancers.
Collapse
Affiliation(s)
- Jie Yuan
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Yang
- Department of Clinical Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Zhi Li
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Hua Zhang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Qun Wang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Jun Huang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Bei Wang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Chakrabhavi Dhananjaya Mohan
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore Karnataka, India
- FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Geng Wang
- Department of Endocrine and Vascular Surgery, Taihe Hospital, Hubei University of Medicine, Hubei, China
| |
Collapse
|
6
|
Zhang Y, Cui L. Discovery and development of small-molecule heparanase inhibitors. Bioorg Med Chem 2023; 90:117335. [PMID: 37257254 PMCID: PMC10884955 DOI: 10.1016/j.bmc.2023.117335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023]
Abstract
Heparanase-1 (HPSE) is a promising yet challenging therapeutic target. It is the only known enzyme that is responsible for cleavage of heparan sulfate (HS) side chains from heparan sulfate proteoglycans (HSPGs), and is the key enzyme involved in the remodeling and degradation of the extracellular matrix (ECM). Overexpression of HPSE is found in various types of diseases, including cancers, inflammations, diabetes, and viral infections. Inhibiting HPSE can restore ECM functions and integrity, making the development of HPSE inhibitors a highly sought-after topic. So far, all HPSE inhibitors that have entered clinical trials belong to the category of HS mimetics, and no small-molecule or drug-like HPSE inhibitors have made similar progress. None of the HS mimetics have been approved as drugs, with some clinical trials discontinued due to poor bioavailability, side effects, and unfavorable pharmacokinetics characteristics. Small-molecule HPSE inhibitors are, therefore, particularly appealing due to their drug-like characteristics. Advances in the chemical spaces and drug design technologies, including the increasing use of in vitro and in silico screening methods, have provided new opportunities in drug discovery. This article aims to review the discovery and development of small-molecule HPSE inhibitors via screening strategies to shed light on the future endeavors in the development of novel HPSE inhibitors.
Collapse
Affiliation(s)
- Yuzhao Zhang
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
7
|
De Novo Design of Imidazopyridine-Tethered Pyrazolines That Target Phosphorylation of STAT3 in Human Breast Cancer Cells. Bioengineering (Basel) 2023; 10:bioengineering10020159. [PMID: 36829653 PMCID: PMC9952374 DOI: 10.3390/bioengineering10020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/02/2023] [Accepted: 01/08/2023] [Indexed: 01/26/2023] Open
Abstract
In breast cancer (BC), STAT3 is hyperactivated. This study explored the design of imidazopyridine-tethered pyrazolines as a de novo drug strategy for inhibiting STAT3 phosphorylation in human BC cells. This involved the synthesis and characterization of two series of compounds namely, 1-(3-(2,6-dimethylimidazo [1,2-a]pyridin-3-yl)-5-(3-nitrophenyl)-4,5-dihydro-1H-pyrazol-1-yl)-2-(4-(substituted)piperazin-1-yl)ethanone and N-substituted-3-(2,6-dimethylimidazo[1,2-a]pyridin-3-yl)-5-(3-nitrophenyl)-4,5-dihydro-1H-pyrazoline-1-carbothioamides. Compound 3f with 2,3-dichlorophenyl substitution was recognized among the tested series as a lead structure that inhibited the viability of MCF-7 cells with an IC50 value of 9.2 μM. A dose- and time-dependent inhibition of STAT3 phosphorylation at Tyr705 and Ser727 was observed in MCF-7 and T47D cells when compound 3f was added in vitro. Calculations using density functional theory showed that the title compounds HOMOs and LUMOs are situated on imidazopyridine-pyrazoline and nitrophenyl rings, respectively. Hence, compound 3f effectively inhibited STAT3 phosphorylation in MCF-7 and T47D cells, indicating that these structures may be an alternative synthon to target STAT3 signaling in BC.
Collapse
|
8
|
Budziak-Wieczorek I, Ślusarczyk L, Myśliwa-Kurdziel B, Kurdziel M, Srebro-Hooper M, Korona-Glowniak I, Gagoś M, Gładyszewski G, Stepulak A, Kluczyk D, Matwijczuk A. Spectroscopic characterization and assessment of microbiological potential of 1,3,4-thiadiazole derivative showing ESIPT dual fluorescence enhanced by aggregation effects. Sci Rep 2022; 12:22140. [PMID: 36550169 PMCID: PMC9780306 DOI: 10.1038/s41598-022-26690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
In the presented study, advanced experimental techniques, including electronic absorption and fluorescence spectroscopies [with Resonance Light Scattering (RLS)], measurements of fluorescence lifetimes in the frequency domain, calculations of dipole moment fluctuations, quantum yields, and radiative and non-radiative transfer constants, were used to characterize a selected analogue from the group of 1,3,4-thiadiazole, namely: 4-[5-(naphthalen-1-ylmethyl)-1,3,4-thiadiazol-2-yl]benzene-1,3-diol (NTBD), intrinsically capable to demonstrate enol → keto excited-states intramolecular proton transfer (ESIPT) effects. The results of spectroscopic analyses conducted in solvent media as well as selected mixtures were complemented by considering biological properties of the derivative in question, particularly in terms of its potential microbiological activity. The compound demonstrated a dual fluorescence effect in non-polar solvents, e.g. chloroform and DMSO/H2O mixtures, while in polar solvents only a single emission maximum was detected. In the studied systems, ESIPT effects were indeed observed, as was the associated phenomenon of dual fluorescence, and, as demonstrated for the DMSO: H2O mixtures, the same could be relatively easily induced by aggregation effects related to aggregation-induced emission (AIE). Subsequently conducted quantum-chemical (TD-)DFT calculations supported further possibility of ESIPT effects. The following article provides a comprehensive description of the spectroscopic and biological properties of the analyzed 1,3,4-thiadiazole derivatives, highlighting its potential applicability as a very good fluorescence probes as well as a compound capable of high microbiological activity.
Collapse
Affiliation(s)
- Iwona Budziak-Wieczorek
- grid.411201.70000 0000 8816 7059Department of Chemistry, Faculty of Food Science and Biotechnology, University of Life Sciences in Lublin, Akademicka 15, 20-950 Lublin, Poland
| | - Lidia Ślusarczyk
- grid.411201.70000 0000 8816 7059Department of Biophysics, Faculty of Environmental Biology, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| | - Beata Myśliwa-Kurdziel
- grid.5522.00000 0001 2162 9631Department of Plant Physiology and Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Martyna Kurdziel
- grid.5522.00000 0001 2162 9631Department of Plant Physiology and Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Monika Srebro-Hooper
- grid.5522.00000 0001 2162 9631Department of Theoretical Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Izabela Korona-Glowniak
- grid.411484.c0000 0001 1033 7158Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Mariusz Gagoś
- grid.29328.320000 0004 1937 1303Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland ,grid.411484.c0000 0001 1033 7158Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Grzegorz Gładyszewski
- grid.41056.360000 0000 8769 4682Department of Applied Physics, Lublin University of Technology, Nadbystrzycka 38, 20-618 Lublin, Poland
| | - Andrzej Stepulak
- grid.411484.c0000 0001 1033 7158Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Dariusz Kluczyk
- grid.29328.320000 0004 1937 1303Department of Plant Physiology and Biophysics, Institute of Biological Sciences, Maria Curie-Sklodowska University, 20-033 Lublin, Poland
| | - Arkadiusz Matwijczuk
- grid.411201.70000 0000 8816 7059Department of Biophysics, Faculty of Environmental Biology, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| |
Collapse
|
9
|
Massironi N, Colombo M, Cosentino C, Fiandra L, Mauri M, Kayal Y, Testa F, Torri G, Urso E, Vismara E, Vlodavsky I. Heparin-Superparamagnetic Iron Oxide Nanoparticles for Theranostic Applications. Molecules 2022; 27:molecules27207116. [PMID: 36296711 PMCID: PMC9611043 DOI: 10.3390/molecules27207116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022] Open
Abstract
In this study, superparamagnetic iron oxide nanoparticles (SPIONs) were engineered with an organic coating composed of low molecular weight heparin (LMWH) and bovine serum albumin (BSA), providing heparin-based nanoparticle systems (LMWH@SPIONs). The purpose was to merge the properties of the heparin skeleton and an inorganic core to build up a targeted theranostic nanosystem, which was eventually enhanced by loading a chemotherapeutic agent. Iron oxide cores were prepared via the co-precipitation of iron salts in an alkaline environment and oleic acid (OA) capping. Dopamine (DA) was covalently linked to BSA and LMWH by amide linkages via carbodiimide coupling. The following ligand exchange reaction between the DA-BSA/DA-LMWH and OA was conducted in a biphasic system composed of water and hexane, affording LMWH@SPIONs stabilized in water by polystyrene sulfonate (PSS). Their size and morphology were investigated via dynamic light scattering (DLS) and transmission electron microscopy (TEM), respectively. The LMWH@SPIONs’ cytotoxicity was tested, showing marginal or no toxicity for samples prepared with PSS at concentrations of 50 µg/mL. Their inhibitory activity on the heparanase enzyme was measured, showing an effective inhibition at concentrations comparable to G4000 (N-desulfo-N-acetyl heparin, a non-anticoagulant and antiheparanase heparin derivative; Roneparstat). The LMWH@SPION encapsulation of paclitaxel (PTX) enhanced the antitumor effect of this chemotherapeutic on breast cancer cells, likely due to an improved internalization of the nanoformulated drug with respect to the free molecule. Lastly, time-domain NMR (TD-NMR) experiments were conducted on LMWH@SPIONs obtaining relaxivity values within the same order of magnitude as currently used commercial contrast agents.
Collapse
Affiliation(s)
- Nicolò Massironi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, 20133 Milan, Italy
| | - Miriam Colombo
- Department of Biotechnology and Biosciences, University of Milano Bicocca, 20126 Milan, Italy
| | - Cesare Cosentino
- Istituto di Ricerche Chimiche e Biochimiche “Giuliana Ronzoni”, 20133 Milan, Italy
| | - Luisa Fiandra
- Department of Biotechnology and Biosciences, University of Milano Bicocca, 20126 Milan, Italy
| | - Michele Mauri
- Department of Materials Science, University of Milano Bicocca, 20125 Milan, Italy
| | - Yasmina Kayal
- Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa 2611001, Israel
| | - Filippo Testa
- Department of Biotechnology and Biosciences, University of Milano Bicocca, 20126 Milan, Italy
| | - Giangiacomo Torri
- Istituto di Ricerche Chimiche e Biochimiche “Giuliana Ronzoni”, 20133 Milan, Italy
- Correspondence: (G.T.); (E.V.); Tel.: +39-02-7064-1624 (G.T.); +39-02-2399-3088 (E.V.)
| | - Elena Urso
- Istituto di Ricerche Chimiche e Biochimiche “Giuliana Ronzoni”, 20133 Milan, Italy
| | - Elena Vismara
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, 20133 Milan, Italy
- Correspondence: (G.T.); (E.V.); Tel.: +39-02-7064-1624 (G.T.); +39-02-2399-3088 (E.V.)
| | - Israel Vlodavsky
- Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa 2611001, Israel
| |
Collapse
|
10
|
Guo S, Wu X, Lei T, Zhong R, Wang Y, Zhang L, Zhao Q, Huang Y, Shi Y, Wu L. The Role and Therapeutic Value of Syndecan-1 in Cancer Metastasis and Drug Resistance. Front Cell Dev Biol 2022; 9:784983. [PMID: 35118073 PMCID: PMC8804279 DOI: 10.3389/fcell.2021.784983] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/03/2021] [Indexed: 12/17/2022] Open
Abstract
Metastasis and relapse are major causes of cancer-related fatalities. The elucidation of relevant pathomechanisms and adoption of appropriate countermeasures are thus crucial for the development of clinical strategies that inhibit malignancy progression as well as metastasis. An integral component of the extracellular matrix, the type 1 transmembrane glycoprotein syndecan-1 (SDC-1) binds cytokines and growth factors involved in tumor microenvironment modulation. Alterations in its localization have been implicated in both cancer metastasis and drug resistance. In this review, available data regarding the structural characteristics, shedding process, and nuclear translocation of SDC-1 are detailed with the aim of highlighting strategies directly targeting SDC-1 as well as SDC-1-mediated carcinogenesis.
Collapse
Affiliation(s)
- Sen Guo
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - XinYi Wu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Lei
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Zhong
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - YiRan Wang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - QingYi Zhao
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Yin Shi
- Department of Acupuncture and Moxibustion, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Outpatient Department, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- *Correspondence: Yin Shi, ; Luyi Wu,
| | - Luyi Wu
- Key Laboratory of Acupuncture and Immunological Effects, Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
- *Correspondence: Yin Shi, ; Luyi Wu,
| |
Collapse
|
11
|
The HPSE Gene Insulator-A Novel Regulatory Element That Affects Heparanase Expression, Stem Cell Mobilization, and the Risk of Acute Graft versus Host Disease. Cells 2021; 10:cells10102523. [PMID: 34685503 PMCID: PMC8534152 DOI: 10.3390/cells10102523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 01/30/2023] Open
Abstract
The HPSE gene encodes heparanase (HPSE), a key player in cancer, inflammation, and autoimmunity. We have previously identified a strong HPSE gene enhancer involved in self-regulation of heparanase by negative feedback exerted in a functional rs4693608 single-nucleotide polymorphism (SNP) dependent manner. In the present study, we analyzed the HPSE gene insulator region, located in intron 9 and containing rs4426765, rs28649799, and rs4364254 SNPs. Our results indicate that this region exhibits HPSE regulatory activity. SNP substitutions lead to modulation of a unique DNA-protein complex that affects insulator activity. Analysis of interactions between enhancer and insulator SNPs revealed that rs4693608 has a major effect on HPSE expression and the risk of post-transplantation acute graft versus host disease (GVHD). The C alleles of insulator SNPs rs4364254 and rs4426765 modify the activity of the HPSE enhancer, resulting in altered HPSE expression and increased risk of acute GVHD. Moreover, rs4426765 correlated with HPSE expression in activated mononuclear cells, as well as with CD3 levels and lymphocyte counts following G-CSF mobilization. rs4363084 and rs28649799 were found to be associated with CD34+ levels. Our study provides new insight into the mechanism of HPSE gene regulation and its impact on normal and pathological processes in the hematopoietic system.
Collapse
|