1
|
Citarella A, Petrella S, Moi D, Dimasi A, Braga T, Ruberto L, Pieraccini S, Sironi M, Micale N, Schirmeister T, Damia G, Fasano V, Silvani A, Giannini C, Passarella D. Synthesis of α-fluorocinnamate derivatives as novel cathepsin S inhibitors with in vitro antiproliferative activity against pancreatic cancer cells. Bioorg Med Chem 2024; 115:117987. [PMID: 39509759 DOI: 10.1016/j.bmc.2024.117987] [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: 06/24/2024] [Revised: 10/14/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
Cathepsins, key members of the papain-like family of cysteine proteases, are crucial for proteolysis processes within human cells, including osteolysis, immunomodulation and apoptosis. Recent research has highlighted the significant role of cathepsins, particularly the L, S, K, and B subtypes, in pancreatic cancer. This has driven the development of novel cathepsin inhibitors as potential treatments to inhibit tumor progression, migration and invasion. Targeting cathepsin S (CatS) has shown promise in reducing tumor progression and enhancing the efficacy of chemotherapeutic agents in preclinical models. Building on our previous work where we employed ethyl p-aminocinnamate ester derivatives for covalent inhibition of cysteine proteases, herein we have designed and synthesized three new derivatives basing on an isosteric replacement (H-F) at the level of cinnamate moiety. These derivatives emerged as potent covalent inhibitors of CatS (1.8-2.6 µM) with 2F showing also weak inhibition activity against CatL (20 %) and CatB (29 %). In vitro assays of 2F against pancreatic cancer cell lines BXPC3 and CAPAN1 revealed significant antiproliferative activity, with IC50 = 5.79 µM and 20.75 µM, respectively. These findings underscore the potential of α-fluorocinnamate-based cysteine protease inhibitors as promising candidates for further development in targeting CatS and CatL with the aim to reduce pancreatic cancer cell proliferation.
Collapse
Affiliation(s)
- Andrea Citarella
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy.
| | - Serena Petrella
- Laboratory of Gynecological Preclinical Oncology, Experimental Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Davide Moi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alessandro Dimasi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Tommaso Braga
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Lorenzo Ruberto
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Stefano Pieraccini
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Maurizio Sironi
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, I-98166 Messina, Italy
| | - Tanja Schirmeister
- Department of Medicinal Chemistry, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Giovanna Damia
- Laboratory of Gynecological Preclinical Oncology, Experimental Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy
| | - Valerio Fasano
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alessandra Silvani
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Clelia Giannini
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Daniele Passarella
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| |
Collapse
|
2
|
Park JY, Park KM. Recent discovery of natural substances with cathepsin L-inhibitory activity for cancer metastasis suppression. Eur J Med Chem 2024; 277:116754. [PMID: 39128327 DOI: 10.1016/j.ejmech.2024.116754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Cathepsin L (CTSL), a cysteine cathepsin protease of the papain superfamily, plays a crucial role in cancer progression and metastasis. Dysregulation of CTSL is frequently observed in tumor malignancies, leading to the degradation of extracellular matrix and facilitating epithelial-mesenchymal transition (EMT), a key process in malignant cancer metastasis. This review mainly provides a comprehensive information about recent findings on natural inhibitors targeting CTSL and their anticancer effects, which have emerged as potent anticancer therapeutic agents or metastasis-suppressive adjuvants. Specifically, inhibitors are categorized into small-molecule and macromolecule inhibitors, with a particular emphasis on cathepsin propeptide-type macromolecules. Additionally, the article explores the molecular mechanisms of CTSL involvement in cancer metastasis, highlighting its regulation at transcriptional, translational, post-translational, and epigenetic levels. This work underscores the importance of understanding natural CTSL inhibitors and provides researchers with practical insights to advance the relevant fields and discover novel CTSL-targeting inhibitors from natural sources.
Collapse
Affiliation(s)
- Jun-Young Park
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyung-Min Park
- Department of Food Science and Biotechnology, Wonkwang University, Iksan, 54538, Republic of Korea.
| |
Collapse
|
3
|
Liu Y, Liu Q, Shang H, Li J, Chai H, Wang K, Guo Z, Luo T, Liu S, Liu Y, Wang X, Zhang H, Wu C, Song SJ, Yang J. Potential application of natural compounds in ischaemic stroke: Focusing on the mechanisms underlying "lysosomocentric" dysfunction of the autophagy-lysosomal pathway. Pharmacol Ther 2024; 263:108721. [PMID: 39284368 DOI: 10.1016/j.pharmthera.2024.108721] [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: 03/27/2024] [Revised: 07/06/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Ischaemic stroke (IS) is the second leading cause of death and a major cause of disability worldwide. Currently, the clinical management of IS still depends on restoring blood flow via pharmacological thrombolysis or mechanical thrombectomy, with accompanying disadvantages of narrow therapeutic time window and risk of haemorrhagic transformation. Thus, novel pathophysiological mechanisms and targeted therapeutic candidates are urgently needed. The autophagy-lysosomal pathway (ALP), as a dynamic cellular lysosome-based degradative process, has been comprehensively studied in recent decades, including its upstream regulatory mechanisms and its role in mediating neuronal fate after IS. Importantly, increasing evidence has shown that IS can lead to lysosomal dysfunction, such as lysosomal membrane permeabilization, impaired lysosomal acidity, lysosomal storage disorder, and dysfunctional lysosomal ion homeostasis, which are involved in the IS-mediated defects in ALP function. There is tightly regulated crosstalk between transcription factor EB (TFEB), mammalian target of rapamycin (mTOR) and lysosomal function, but their relationship remains to be systematically summarized. Notably, a growing body of evidence emphasizes the benefits of naturally derived compounds in the treatment of IS via modulation of ALP function. However, little is known about the roles of natural compounds as modulators of lysosomes in the treatment of IS. Therefore, in this context, we provide an overview of the current understanding of the mechanisms underlying IS-mediated ALP dysfunction, from a lysosomal perspective. We also provide an update on the effect of natural compounds on IS, according to their chemical structural types, in different experimental stroke models, cerebral regions and cell types, with a primary focus on lysosomes and autophagy initiation. This review aims to highlight the therapeutic potential of natural compounds that target lysosomal and ALP function for IS treatment.
Collapse
Affiliation(s)
- Yueyang Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hanxiao Shang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jichong Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - He Chai
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kaixuan Wang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenkun Guo
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Tianyu Luo
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shiqi Liu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yan Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuemei Wang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hangyi Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Chunfu Wu
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province; Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province; Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Jingyu Yang
- Key Laboratory of Efficacy Evaluation of New Drug Candidate, Liaoning Province; Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
4
|
Tedeschini T, Campara B, Grigoletto A, Zanotto I, Cannella L, Gabbia D, Matsuno Y, Suzuki A, Yoshioka H, Armirotti A, De Martin S, Pasut G. Optimization of a pendant-shaped PEGylated linker for antibody-drug conjugates. J Control Release 2024; 375:74-89. [PMID: 39216599 DOI: 10.1016/j.jconrel.2024.08.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
In this work, we conceived and developed antibody-drug conjugates (ADCs) that could efficiently release the drug after enzymatic cleavage of the linker moiety by tumoral proteases. The antibody-drug linkers we used are the result of a rational optimization of a previously reported PEGylated linker, PUREBRIGHT® MA-P12-PS, which showed excellent drug loading capacities but lacked an inbuilt drug discharge mechanism, thus limiting the potency of the resulting ADCs. To address this limitation, we chose to incorporate a protease-sensitive trigger into the linker to favor the release of a "PEGless" drug inside the tumor cells and, therefore, obtain potent ADCs. Currently, most marketed ADCs are based on the Val-Cit dipeptide followed by a self-immolative spacer for releasing the drug in its unmodified form. Here, we selected two untraditional peptide sequences, a Phe-Gly dipeptide and a Val-Ala-Gly tripeptide and placed one or the other in between the drug on one side (N-terminus) and the rest of the linker, including the PEG moiety, on the other side (C-terminus), without a self-immolative group. We found that both linkers responded to cathepsin B, a reference lysosomal enzyme, and liberated a PEG-free drug catabolite, as desired. We then used the two linkers to generate ADCs based on trastuzumab (a HER2-targeting antibody) and DM1 (a microtubule-targeted cytotoxic agent) with an average drug-to-antibody ratio (DAR) of 4 or 8. The ADCs showed restored cytotoxicity in vitro, which was proportional to the DM1 loading and generally higher for the ADCs bearing Val-Ala-Gly in their structure. In an ovarian cancer mouse model, the DAR 8 ADC based on Val-Ala-Gly behaved better than Kadcyla® (an approved ADC of DAR 3.5 used as control throughout this study), leading to a higher tumor volume reduction and more prolonged median survival. Taken together, our results depict a successful linker optimization process and encourage the application of the Val-Ala-Gly tripeptide as an alternative to other existing protease-sensitive triggers for ADCs.
Collapse
Affiliation(s)
- T Tedeschini
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy.
| | - B Campara
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - A Grigoletto
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - I Zanotto
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - L Cannella
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - D Gabbia
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - Y Matsuno
- NOF CORPORATION, Life Science Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - A Suzuki
- NOF CORPORATION, Life Science Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - H Yoshioka
- NOF CORPORATION, Life Science Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - A Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - S De Martin
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - G Pasut
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy.
| |
Collapse
|
5
|
Rot AE, Hrovatin M, Bokalj B, Lavrih E, Turk B. Cysteine cathepsins: From diagnosis to targeted therapy of cancer. Biochimie 2024; 226:10-28. [PMID: 39245316 DOI: 10.1016/j.biochi.2024.09.001] [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: 03/17/2024] [Revised: 08/23/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Cysteine cathepsins are a fascinating group of proteolytic enzymes that play diverse and crucial roles in numerous biological processes, both in health and disease. Understanding these proteases is essential for uncovering novel insights into the underlying mechanisms of a wide range of disorders, such as cancer. Cysteine cathepsins influence cancer biology by participating in processes such as extracellular matrix degradation, angiogenesis, immune evasion, and apoptosis. In this comprehensive review, we explore foundational research that illuminates the diverse and intricate roles of cysteine cathepsins as diagnostic markers and therapeutic targets for cancer. This review aims to provide valuable insights into the clinical relevance of cysteine cathepsins and explore their capacity to advance personalised and targeted medical interventions in oncology.
Collapse
Affiliation(s)
- Ana Ercegovič Rot
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Matija Hrovatin
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Bor Bokalj
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Ernestina Lavrih
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
6
|
Cappellesso F, Mazzone M, Virga F. Acid affairs in anti-tumour immunity. Cancer Cell Int 2024; 24:354. [PMID: 39465367 PMCID: PMC11514911 DOI: 10.1186/s12935-024-03520-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolic rewiring of cancer cells is one of the hallmarks of cancer. As a consequence, the metabolic landscape of the tumour microenvironment (TME) differs compared to correspondent healthy tissues. Indeed, due to the accumulation of acid metabolites, such as lactate, the pH of the TME is generally acidic with a pH drop that can be as low as 5.6. Disruptions in the acid-base balance and elevated lactate levels can drive malignant progression not only through cell-intrinsic mechanisms but also by impacting the immune response. Generally, acidity and lactate dampen the anti-tumour response of both innate and adaptive immune cells favouring tumour progression and reducing the response to immunotherapy. In this review, we summarize the current knowledge on the functional, metabolic and epigenetic effects of acidity and lactate on the cells of the immune system. In particular, we focus on the role of monocarboxylate transporters (MCTs) and other solute carrier transporters (SLCs) that, by mediating the exchange of lactate (among other metabolites) and bicarbonate, participate in pH regulation and lactate transport in the cancer context. Finally, we discuss advanced approaches to target pH or lactate in the TME to enhance the anti-tumour immune response.
Collapse
Affiliation(s)
- Federica Cappellesso
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, Inflammation Research Center, VIB, Brussels, Belgium.
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Federico Virga
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029, Spain.
| |
Collapse
|
7
|
Lepori I, Roncetti M, Vitiello M, Barresi E, De Paolo R, Tentori PM, Baldanzi C, Santi M, Evangelista M, Signore G, Tedeschi L, Gravekamp C, Cardarelli F, Taliani S, Da Settimo F, Siegrist MS, Poliseno L. Enhancing the Anticancer Activity of Attenuated Listeria monocytogenes by Cell Wall Functionalization with "Clickable" Doxorubicin. ACS Chem Biol 2024; 19:2131-2140. [PMID: 39317359 PMCID: PMC11494506 DOI: 10.1021/acschembio.4c00250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 09/26/2024]
Abstract
Among bacteria used as anticancer vaccines, attenuated Listeria monocytogenes (Lmat) stands out, because it spreads from one infected cancer cell to the next, induces a strong adaptive immune response, and is suitable for repeated injection cycles. Here, we use click chemistry to functionalize the Lmat cell wall and turn the bacterium into an "intelligent carrier" of the chemotherapeutic drug doxorubicin. Doxorubicin-loaded Lmat retains most of its biological properties and, compared to the control fluorophore-functionalized bacteria, shows enhanced cytotoxicity against melanoma cells both in vitro and in a xenograft model in zebrafish. Our results show that drugs can be covalently loaded on the Lmat cell wall and pave the way to the development of new two-in-one therapeutic approaches combining immunotherapy with chemotherapy.
Collapse
Affiliation(s)
- Irene Lepori
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| | - Marta Roncetti
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
- University of Siena, Siena 53100, Italy
| | - Marianna Vitiello
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
- CISUP-Center for Instrument Sharing, University of Pisa, Pisa 56126, Italy
| | - Raffaella De Paolo
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| | - Paolo Maria Tentori
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Pisa 56126, Italy
| | - Caterina Baldanzi
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| | - Melissa Santi
- NEST-Scuola Normale Superiore, Istituto Nanoscienze, CNR (CNR-NANO), Pisa 56126, Italy
| | - Monica Evangelista
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
| | - Giovanni Signore
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Pisa 56017, Italy
| | - Lorena Tedeschi
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
| | - Claudia Gravekamp
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York 10461, United States
| | - Francesco Cardarelli
- NEST-Scuola Normale Superiore, Istituto Nanoscienze, CNR (CNR-NANO), Pisa 56126, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
- CISUP-Center for Instrument Sharing, University of Pisa, Pisa 56126, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Pisa 56126, Italy
- CISUP-Center for Instrument Sharing, University of Pisa, Pisa 56126, Italy
| | - M Sloan Siegrist
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003-9316, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003-9316, United States
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| |
Collapse
|
8
|
Garrute FV, Pacheco ABF, Lu GJ, Machado JC. A Bioengineered Cathepsin B-sensitive Gas Vesicle Nanosystem That Responds With Increased Gray-level Intensity of Ultrasound Biomicroscopic Images. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00360-0. [PMID: 39394005 DOI: 10.1016/j.ultrasmedbio.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVE This work aimed to promote the interaction of a modified gas vesicle (GV) with cathepsin B (CTSB) protease and analysed their backscattered signal by ultrasound (US). METHODS We modified the sequence of the gene coding for GvpC to contain a CTSB cleavage and expressed the protein in an Escherichia coli recombinant system. The protein was purified and added to GVs preparations in which the original GvpC was removed (ΔGV), constituting the modified GV (GV*). Western blot testing was used to compare GVs with GvpC and engineered GvpC at starting (T0) and after 24 h (T24) reacting with CTSB. A 21 MHz US B-mode and non-linear contrast mode (5% total power) imaged US phantoms having samples of GVwt, ΔGV (stripped GV), GV* and CTSB + GV*. Also, a 21 MHz US B-mode imaged US phantoms having a tumour cell line extracellular fraction (TCEF) and the TCEF + GV* sample. A 100% total US power was applied to collapse the GV structure. RESULTS On Western blotting, we detected a decrease in engineered GvpC levels 24 h after the incubation of GV* with CTSB, compared with the concentration at T0, suggesting that CTSB cleaved the engineered GvpC. Regions-of-interest over image of phantom cross-sections were determined and the B-mode image mean grey-level intensity resulted in a significant (p < 0.05) increase comparing CTSB + GV* with PBS (control), GVwt, ΔGV and GV*. Non-linear mode image grey-level intensity from CTSB + GV* increased by 11.79, 7.86 and 14.75 dB from samples containing GVwt, ΔGV and GV*, respectively. GV preparations incubated with TCEF and the TCEF + GV* sample showed an increase of 81% in signal compared with TCEF + GVwt. CONCLUSION The increased US backscattered signal intensity suggests GVs as a potential biosensor for protease activity, possibly aiding the detection of protease-rich tissue regions.
Collapse
Affiliation(s)
- Felipe Vianna Garrute
- Biomedical Engineering Program, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Beatriz F Pacheco
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - George J Lu
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - João Carlos Machado
- Biomedical Engineering Program, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Post-Graduate Program in Surgical Sciences, Department of Surgery, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
9
|
Linders DGJ, Bijlstra OD, Fallert LC, Dekker-Ensink NG, March TL, Pool M, Walker E, Straight B, Basilion JP, Bogyo M, Burggraaf J, Hilling DE, Vahrmeijer AL, Kuppen PJK, Crobach ASLP. Immunohistochemical Evaluation of Cathepsin B, L, and S Expression in Breast Cancer Patients. Mol Imaging Biol 2024:10.1007/s11307-024-01955-5. [PMID: 39331316 DOI: 10.1007/s11307-024-01955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/12/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE Cysteine cathepsins are proteases that play a role in normal cellular physiology and neoplastic transformation. Elevated expression and enzymatic activity of cathepsins in breast cancer (BCa) indicates their potential as a target for tumor imaging. In particular cathepsin B (CTSB), L (CTSL), and S (CTSS) are used as targets for near-infrared (NIR) fluorescence imaging (FI), a technique that allows real-time intraoperative tumor visualization and resection margin assessment. Therefore, this immunohistochemical study explores CTSB, CTSL, and CTSS expression levels in a large breast cancer patient cohort, to investigate in which BCa patients the use of cathepsin-targeted NIR FI may have added value. PROCEDURES Protein expression was analyzed in tumor tissue microarrays (TMA) of BCa patients using immunohistochemistry and quantified as a total immunostaining score (TIS), ranging from 0-12. In total, the tissues of 557 BCa patients were included in the TMA. RESULTS CTSB, CTSL, and CTSS were successfully scored in respectively 340, 373 and 252 tumors. All tumors showed CTSB, CTSL, and/or CTSS expression to some extent (TIS > 0). CTSB, CTSL, and CTSS expression was scored as high (TIS > 6) in respectively 28%, 80%, and 18% of tumors. In 89% of the tumors scored for all three cathepsins, the expression level of one or more of these proteases was scored as high (TIS > 6). Tumors showed significantly higher cathepsin expression levels with advancing Bloom-Richardson grade (p < 0.05). Cathepsin expression was highest in estrogen receptor (ER)-negative, human epidermal growth factor receptor 2(HER2)-positive and triple-negative (TN) tumors. There was no significant difference in cathepsin expression between tumors that were treated with neoadjuvant systemic therapy and tumors that were not. CONCLUSIONS The expression of at least one of the cysteine cathepsins B, L and S in all breast tumor tissues tested suggests that cathepsin-activatable imaging agents with broad reactivity for these three proteases will likely be effective in the vast majority of breast cancer patients, regardless of molecular subtype and treatment status. Patients with high grade ER-negative, HER2-positive, or TN tumors might show higher imaging signals.
Collapse
Affiliation(s)
- Daan G J Linders
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
| | - Okker D Bijlstra
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Laura C Fallert
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - N Geeske Dekker-Ensink
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Taryn L March
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Martin Pool
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Ethan Walker
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | | | - James P Basilion
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Akrotome Imaging Inc, Charlotte, NC, 28205, USA
- Department of Radiology, Case School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacobus Burggraaf
- Centre for Human Drug Research, 2333 AL, Leiden, The Netherlands
- Leiden Academic Center for Drug Research, Leiden University, 2300 RA, Leiden, The Netherlands
| | - Denise E Hilling
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - A Stijn L P Crobach
- Department of Pathology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
10
|
Liu X, Lu X, Li X, Zhang M, Xue L, Yang S, Hu R, Cui Y, Jiang X, Sun G. Highly selective fluorescent probe for cysteine via a tandem reaction and its bioimaging application in HeLa cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 326:125223. [PMID: 39348738 DOI: 10.1016/j.saa.2024.125223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/24/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
Cysteine, as a vital endogenous small molecule mercaptan, plays a crucial role in various physiological processes. The high sensitivity and selectivity of fluorescent probes provide a method to monitor cysteine, which is helpful to understand the mechanism of cysteine in physiological processes more comprehensively. However, the detection of cysteine can be interfered by other small molecule biothiols. Therefore, the design of fluorescent probe based on the structural characteristics and reactivity of cysteine has become research focus currently. Given the biological compatibilities, biological targets, the metabolic pathway of 3-hydoxythalidomide, and its unique fluorescent properties, herein, we have designed a chemodosimeter, 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl acrylate, for the detection of cysteine based on a tandem reaction of thiol-ene click chemistry and aminolysis involving 3-hydroxythalidomide as a parent compound. Experimental data exhibited that the probe showed unique selectivity and sensitivity for cysteine over other amino acid and biothiols. In addition, the fluorescent intensity at 511 nm increased linearly as a function of cysteine concentration in the range of 0-6 × 10-7 M (regression factor, R2 = 0.999), with a limit of detection of 6.1 nM. The sensing mechanism was confirmed through 1H NMR titration and density functional theory calculations. Additionally, the probe was also successfully utilized for the detection of cysteine in sewage and for bioimaging in HeLa cells.
Collapse
Affiliation(s)
- Xiaolei Liu
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China; School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China.
| | - Xiangxiang Lu
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Xiang Li
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Miaomiao Zhang
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Liang Xue
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Shuaijun Yang
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Riming Hu
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Yu Cui
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Xuchuan Jiang
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China.
| | - Guoxin Sun
- Institute for Smart Materials and Engineering, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| |
Collapse
|
11
|
Jurczak P, Fayad N, Benard M, Czaplewska P, Hildebrandt N. Monomer-Dimer Equilibrium of Human Cystatin C During Internalization Into Cancer Cells. Chembiochem 2024; 25:e202400226. [PMID: 38761032 DOI: 10.1002/cbic.202400226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/26/2024] [Accepted: 05/17/2024] [Indexed: 05/20/2024]
Abstract
Human cystatin C (hCC) is a physiologically important protein that serves as intra- and extracellular cysteine proteinase inhibitor in homeostasis. However, in pathological states it dimerizes and further oligomerizes accumulating into a toxic amyloid. HCC forms an active monomer in the extracellular space and becomes an inactive dimer when internalized in cellular organelles. However, hCC cell penetration and its oligomeric state during this process are not well understood. To determine if and how the oligomeric state influences hCC transmembrane migration, we investigated the internalization of the hCC wild type protein as well as three different mutants, which exclusively exist in the monomeric or multimeric state into HeLa cells via confocal fluorescence microscopy. Our results showed that the preferred pathway was endocytosis and that the oligomeric state did not significantly influence the internalization because both monomeric and dimeric hCC migrated into HeLa cells. Considering the differences of the active monomeric and the passive dimeric states of hCC, our findings contribute to a better understanding of the intra and extra cellular functions of hCC and their interaction with cysteine proteases.
Collapse
Affiliation(s)
- Przemyslaw Jurczak
- Laboratory of Mass Spectrometry, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, Gdańsk, 80-307, Poland
- Laboratoire COBRA (UMR6014 & FR3038), Université de Rouen Normandie, CNRS, INSA, Normandie Université, Rouen, 76000, France
| | - Nour Fayad
- Laboratoire COBRA (UMR6014 & FR3038), Université de Rouen Normandie, CNRS, INSA, Normandie Université, Rouen, 76000, France
| | - Magalie Benard
- PRIMACEN, Univ Rouen Normandie, INSERM, CNRS, HeRacLeS US51 UAR2026, Rouen, 76000, France
| | - Paulina Czaplewska
- Laboratory of Mass Spectrometry, Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, Gdańsk, 80-307, Poland
| | - Niko Hildebrandt
- Department of Chemistry, Seoul National University, Seoul, 08826, South Korea
- Department of Engineering Physics, McMaster University, 1280 Main Street West, Hamilton, L8S4 L7, Canada
| |
Collapse
|
12
|
Trojani MC, Santucci-Darmanin S, Breuil V, Carle GF, Pierrefite-Carle V. Lysosomal exocytosis: From cell protection to protumoral functions. Cancer Lett 2024; 597:217024. [PMID: 38871244 DOI: 10.1016/j.canlet.2024.217024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024]
Abstract
Lysosomes are single membrane bounded group of acidic organelles that can be involved in a process called lysosomal exocytosis which leads to the extracellular release of their content. Lysosomal exocytosis is required for plasma membrane repair or remodeling events such as bone resorption, antigen presentation or mitosis, and for protection against toxic agents such as heavy metals. Recently, it has been showed that to fulfill this protective role, lysosomal exocytosis needs some autophagic proteins, in an autophagy-independent manner. In addition to these crucial physiological roles, lysosomal exocytosis plays a major protumoral role in various cancers. This effect is exerted through tumor microenvironment modifications, including extracellular matrix remodeling, acidosis, oncogenic and profibrogenic signals. This review provides a comprehensive overview of the different elements released in the microenvironment during lysosomal exocytosis, i.e. proteases, exosomes, and protons, and their effects in the context of tumor development and treatment.
Collapse
Affiliation(s)
- Marie-Charlotte Trojani
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; Service de Rhumatologie, CHU de Nice, Nice, France
| | - Sabine Santucci-Darmanin
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; CNRS, Paris, France
| | - Véronique Breuil
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; Service de Rhumatologie, CHU de Nice, Nice, France
| | - Georges F Carle
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; CNRS, Paris, France
| | - Valérie Pierrefite-Carle
- UMR E-4320 TIRO-MATOs CEA/DRF/Institut Joliot, Université Côte d'Azur, Faculté de Médecine Nice, France; INSERM, Paris, France.
| |
Collapse
|
13
|
Chen Y, Zhang L, Fang L, Chen C, Zhang D, Peng T. Modular Development of Enzyme-Activatable Proteolysis Targeting Chimeras for Selective Protein Degradation and Cancer Targeting. JACS AU 2024; 4:2564-2577. [PMID: 39055140 PMCID: PMC11267540 DOI: 10.1021/jacsau.4c00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 07/27/2024]
Abstract
As an emerging therapeutic modality, proteolysis targeting chimeras (PROTACs) indiscriminately degrade proteins in both healthy and diseased cells, posing a risk of on-target off-site toxicity in normal tissues. Herein, we present the modular development of enzyme-activatable PROTACs, which utilize enzyme-recognition moieties to block protein degradation activities and can be specifically activated by elevated enzymes in cancer cells to enable cell-selective protein degradation and cancer targeting. We identified the methylene alkoxy carbamate (MAC) unit as an optimal self-immolative linker, possessing high stability and release efficiency for conjugating enzyme-recognition moieties with PROTACs. Leveraging the MAC linker, we developed a series of enzyme-activatable PROTACs, harnessing distinct enzymes for cancer-cell-selective protein degradation. Significantly, we introduced the first dual-enzyme-activatable PROTAC that requires the presence of two cancer-associated enzymes for activation, demonstrating highly selective protein degradation in cancer cells over nonmalignant cells, potent in vivo antitumor efficacy, and no off-tumor toxicity to normal tissues. The broad applicability of enzyme-activatable PROTACs was further demonstrated by caging other PROTACs via the MAC linker to target different proteins and E3 ligases. Our work underscores the substantial potential of enzyme-activatable PROTACs in overcoming the off-site toxicity associated with conventional PROTACs and offers new opportunities for targeted cancer treatment.
Collapse
Affiliation(s)
- Yanchi Chen
- State
Key Laboratory of Chemical Oncogenomics, School of Chemical Biology
and Biotechnology, Peking University Shenzhen
Graduate School, Shenzhen 518055, China
- National
Key Laboratory of Non-Food Biomass Energy Technology, National Engineering
Research Center for Non-Food Biorefinery, Institute of Grand Health, Guangxi Academy of Sciences, 98 Daling Road, Nanning 530007, China
| | - Lina Zhang
- State
Key Laboratory of Chemical Oncogenomics, School of Chemical Biology
and Biotechnology, Peking University Shenzhen
Graduate School, Shenzhen 518055, China
| | - Lincheng Fang
- State
Key Laboratory of Chemical Oncogenomics, School of Chemical Biology
and Biotechnology, Peking University Shenzhen
Graduate School, Shenzhen 518055, China
| | - Chengjie Chen
- State
Key Laboratory of Chemical Oncogenomics, School of Chemical Biology
and Biotechnology, Peking University Shenzhen
Graduate School, Shenzhen 518055, China
| | - Dong Zhang
- State
Key Laboratory of Chemical Oncogenomics, School of Chemical Biology
and Biotechnology, Peking University Shenzhen
Graduate School, Shenzhen 518055, China
| | - Tao Peng
- State
Key Laboratory of Chemical Oncogenomics, School of Chemical Biology
and Biotechnology, Peking University Shenzhen
Graduate School, Shenzhen 518055, China
- Institute
of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
14
|
Pitkänen M, Matilainen O. Milk Fat Globule Membrane-Containing Protein Powder Promotes Fitness in Caenorhabditis elegans. Nutrients 2024; 16:2290. [PMID: 39064733 PMCID: PMC11280102 DOI: 10.3390/nu16142290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Milk-derived peptides and milk fat globule membrane (MFGM) have gained interest as health-promoting food ingredients. However, the mechanisms by which these nutraceuticals modulate the function of biological systems often remain unclear. We utilized Caenorhabditis elegans to elucidate how MFGM-containing protein powder (MProPow), previously used in a clinical trial, affect the physiology of this model organism. Our results demonstrate that MProPow does not affect lifespan but promotes the fitness of the animals. Surprisingly, gene expression analysis revealed that MProPow decreases the expression of genes functioning on innate immunity, which also translates into reduced survival on pathogenic bacteria. One of the innate immunity-associated genes showing reduced expression upon MProPow supplementation is cpr-3, the homolog of human cathepsin B. Interestingly, knockdown of cpr-3 enhances fitness, but not in MProPow-treated animals, suggesting that MProPow contributes to fitness by downregulating the expression of this gene. In summary, this research highlights the value of C. elegans in testing the biological activity of food supplements and nutraceuticals. Furthermore, this study should encourage investigations into whether milk-derived peptides and MFGM mediate their beneficial effects through the modulation of cathepsin B expression in humans.
Collapse
Affiliation(s)
| | - Olli Matilainen
- The Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00790 Helsinki, Finland;
| |
Collapse
|
15
|
Lu Y, Huang Y, Jin J, Yu J, Lu W, Zhu S. Design, synthesis, and biological evaluation of cathepsin B cleavage albumin-binding SN38 prodrug in breast cancer. Bioorg Chem 2024; 147:107370. [PMID: 38621338 DOI: 10.1016/j.bioorg.2024.107370] [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: 12/08/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024]
Abstract
Here, we introduce a novel and effective approach utilizing a cathepsin B cleavage albumin-binding SN38 prodrug specifically designed for the treatment of metastatic breast cancer. Termed Mal-va-mac-SN38, our prodrug exhibits a unique ability to rapidly and covalently bind with endogenous albumin, resulting in the formation of HSA-va-mac-SN38. This prodrug demonstrates exceptional stability in human plasma. Importantly, HSA-va-mac-SN38 showcases an impressive enhancement in cellular uptake by 4T1 breast cancer cells, primarily facilitated through caveolin-mediated endocytosis. Intriguingly, the release of the active SN38, is triggered by the enzymatic activity of cathepsin B within the lysosomal environment. In vivo studies employing a lung metastasis 4T1 breast cancer model underscore the potency of HSA-va-mac-SN38. Histological immunohistochemical analyses further illuminate the multifaceted impact of our prodrug, showcasing elevated levels of apoptosis, downregulated expression of matrix metalloproteinases, and inhibition of angiogenesis, all critical factors contributing to the anti-metastatic effect observed. Biodistribution studies elucidate the capacity of Mal-va-mac-SN38 to augment tumor accumulation through covalent binding to serum albumin, presenting a potential avenue for targeted therapeutic interventions. Collectively, our findings propose a promising therapeutic avenue for metastatic breast cancer, through the utilization of a cathepsin B-cleavable albumin-binding prodrug.
Collapse
Affiliation(s)
- Yingxin Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Ying Huang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Jiyu Jin
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Jiahui Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China.
| | - Shulei Zhu
- Innovation Center for AI and Drug Discovery, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China.
| |
Collapse
|
16
|
Denison M, Garcia SP, Ullrich A, Podgorski I, Gibson H, Turro C, Kodanko JJ. Ruthenium-Cathepsin Inhibitor Conjugates for Green Light-Activated Photodynamic Therapy and Photochemotherapy. Inorg Chem 2024; 63:7973-7983. [PMID: 38616353 PMCID: PMC11066580 DOI: 10.1021/acs.inorgchem.4c01008] [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] [Indexed: 04/16/2024]
Abstract
Dysregulated cathepsin activity is linked to various human diseases including metabolic disorders, autoimmune conditions, and cancer. Given the overexpression of cathepsin in the tumor microenvironment, cathepsin inhibitors are promising pharmacological agents and drug delivery vehicles for cancer treatment. In this study, we describe the synthesis and photochemical and biological assessment of a dual-action agent based on ruthenium that is conjugated with a cathepsin inhibitor, designed for both photodynamic therapy (PDT) and photochemotherapy (PCT). The ruthenium-cathepsin inhibitor conjugate was synthesized through an oxime click reaction, combining a pan-cathepsin inhibitor based on E64d with the Ru(II) PCT/PDT fragment [Ru(dqpy)(dppn)], where dqpy = 2,6-di(quinoline-2-yl)pyridine and dppn = benzo[i]dipyrido[3,2-a:2',3'-c]phenazine. Photochemical investigations validated the conjugate's ability to release a triazole-containing cathepsin inhibitor for PCT and to generate singlet oxygen for PDT upon exposure to green light. Inhibition studies demonstrated the conjugate's potent and irreversible inactivation of purified and intracellular cysteine cathepsins. Two Ru(II) PCT/PDT agents based on the [Ru(dqpy)(dppn)] moiety were evaluated for photoinduced cytotoxicity in 4T1 murine triple-negative breast cancer cells, L929 fibroblasts, and M0, M1, and M2 macrophages. The cathepsin inhibitor conjugate displayed notable selectivity for inducing cell death under irradiation compared to dark conditions, mitigating toxicity in the dark observed with the triazole control complex [Ru(dqpy)(dppn)(MeTz)]2+ (MeTz = 1-methyl-1H-1,2,4-triazole). Notably, our lead complex is among a limited number of dual PCT/PDT agents activated with green light.
Collapse
Affiliation(s)
- Madeline Denison
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, Michigan 48202, United States
| | - Santana P Garcia
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Alexander Ullrich
- Department of Oncology, Wayne State University, Detroit, Michigan 48201, United States
| | - Izabela Podgorski
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan 48201, United States
- Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Heather Gibson
- Department of Oncology, Wayne State University, Detroit, Michigan 48201, United States
- Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| | - Claudia Turro
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jeremy J Kodanko
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, Michigan 48202, United States
- Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| |
Collapse
|
17
|
Coene J, Wilms S, Verhelst SHL. Photopharmacology of Protease Inhibitors: Current Status and Perspectives. Chemistry 2024; 30:e202303999. [PMID: 38224181 DOI: 10.1002/chem.202303999] [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: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/16/2024]
Abstract
Proteases are involved in many essential biological processes. Dysregulation of their activity underlies a wide variety of human diseases. Photopharmacology, as applied on various classes of proteins, has the potential to assist protease research by enabling spatiotemporal control of protease activity. Moreover, it may be used to decrease side-effects of protease-targeting drugs. In this review, we discuss the current status of the chemical design of photoactivatable proteases inhibitors and their biological application. Additionally, we give insight into future possibilities for further development of this field of research.
Collapse
Affiliation(s)
- Jonathan Coene
- Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49, box 901b, 3000, Leuven, Belgium
| | - Simon Wilms
- Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49, box 901b, 3000, Leuven, Belgium
| | - Steven H L Verhelst
- Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49, box 901b, 3000, Leuven, Belgium
| |
Collapse
|
18
|
Park JH, Sung Y, Jo S, Lee SH, Ryu JH, Sun IC, Ahn CH. Applications of Cu 2+-Loaded Silica Nanoparticles to Photothermal Therapy and Tumor-Specific Fluorescence Imaging. J Funct Biomater 2024; 15:81. [PMID: 38667538 PMCID: PMC11051373 DOI: 10.3390/jfb15040081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Copper-based nanomaterials have been employed as therapeutic agents for cancer therapy and diagnosis. Nevertheless, persistent challenges, such as cellular toxicity, non-uniform sizes, and low photothermal efficiency, often constrain their applications. In this study, we present Cu2+-loaded silica nanoparticles fabricated through the chelation of Cu2+ ions by silanol groups. The integration of Cu2+ ions into uniformly sized silica nanoparticles imparts a photothermal therapy effect. Additionally, the amine functionalization of the silica coating facilitates the chemical conjugation of tumor-specific fluorescence probes. These probes are strategically designed to remain in an 'off' state through the Förster resonance energy transfer mechanism until exposed to cysteine enzymes in cancer cells, inducing the recovery of their fluorescence. Consequently, our Cu2+-loaded silica nanoparticles demonstrate an efficient photothermal therapy effect and selectively enable cancer imaging.
Collapse
Affiliation(s)
- Ji-Ho Park
- NanoBio Materials Laboratory, Department of Materials Science and Engineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea; (J.-H.P.)
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; (Y.S.)
| | - Yejin Sung
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; (Y.S.)
| | - SeongHoon Jo
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seung Ho Lee
- NanoBio Materials Laboratory, Department of Materials Science and Engineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea; (J.-H.P.)
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; (Y.S.)
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; (Y.S.)
| | - In-Cheol Sun
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; (Y.S.)
| | - Cheol-Hee Ahn
- NanoBio Materials Laboratory, Department of Materials Science and Engineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea; (J.-H.P.)
| |
Collapse
|
19
|
Sookai S, Akerman MP, Munro OQ. Chiral Au(III) chelates exhibit unique NCI-60 cytotoxicity profiles and interactions with human serum albumin. Dalton Trans 2024; 53:5089-5104. [PMID: 38375922 DOI: 10.1039/d3dt04024k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Au(III) bis(pyrrolide-imine) chelates are emerging as a class of versatile, efficacious metallodrug candidates. Here, we synthesised two enantiopure chiral ligands H2L1 and H2L2 (tetradentate cyclohexane-1,2-diamine-bridged bis(pyrrole-imine) derivatives). Metallation of the ligands with Au(III) afforded the chiral cationic complexes AuL1 and AuL2. The in vitro cytotoxicities of AuL1 and AuL2 determined in the NCI-60 single-dose drug screen were 56.5% and 89.1%, respectively. AuL1 was subsequently selected for a five-dose NCI-60 screen, attaining GI50, IC50, and LC50 values of 4.7, 9.3 and 39.8 μM, respectively. Hierarchical cluster analysis of the NCI-60 data indicated that the profile for AuL1 was similar to that of vinblastine sulfate, a microtubule-targeting vinca alkaloid. Reactions of AuL1 with glutathione (GSH) in vitro confirmed its susceptibility to reduction, Au(III) → Au(I), by intracellular thiols. Because human serum albumin (HSA) is responsible for transporting clinically deployed and investigational drugs, we studied the uptake of AuL1 and AuL2 by HSA to delineate how chirality impacts their protein-binding affinity. Steady-state fluorescence quenching data acquired on the native protein and data from site-specific probes showed that the compounds bind at sites close enough to Trp-214 (subdomain IIA) of HSA to quench the fluorophore. The bimolecular quenching rate constants, Kq, were ca. 102 times higher than the maximum diffusion-controlled collision constant of a biomolecule in water (1010 M-1 s-1), confirming that static fluorescence quenching was the dominant mechanism. The Stern-Volmer constants, KSV, were ∼104 M-1 at 37 °C, while the affinity constants, Ka (37 °C), measured ∼2.1 × 104 M-1 (AuL1) and ∼1.2 × 104 M-1 (AuL2) for enthalpy-driven ligand uptake targeting Sudlow's site I. Although far- and near-UV CD spectroscopy indicated that both complexes minimally perturb the secondary and tertiary structure of HSA, substantial shifts in the CD spectra were recorded for both protein-bound ligands. This study highlights the role of chirality in determining the cytotoxicity profiles and protein binding behaviour of enantiomeric Au(III) chelates.
Collapse
Affiliation(s)
- Sheldon Sookai
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, PO WITS 2050, Johannesburg, South Africa.
| | - Matthew P Akerman
- School of Chemistry and Physics, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, PO WITS 2050, Johannesburg, South Africa.
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| |
Collapse
|
20
|
Kim MJ, Kim S, Reinheckel T, Krainc D. Inhibition of cysteine protease cathepsin L increases the level and activity of lysosomal glucocerebrosidase. JCI Insight 2024; 9:e169594. [PMID: 38329128 PMCID: PMC10967467 DOI: 10.1172/jci.insight.169594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/13/2023] [Indexed: 02/09/2024] Open
Abstract
The glucocerebrosidase (GCase) encoded by the GBA1 gene hydrolyzes glucosylceramide (GluCer) to ceramide and glucose in lysosomes. Homozygous or compound heterozygous GBA1 mutations cause the lysosomal storage disease Gaucher disease (GD) due to severe loss of GCase activity. Loss-of-function variants in the GBA1 gene are also the most common genetic risk factor for Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Restoring lysosomal GCase activity represents an important therapeutic approach for GBA1-associated diseases. We hypothesized that increasing the stability of lysosomal GCase protein could correct deficient GCase activity in these conditions. However, it remains unknown how GCase stability is regulated in the lysosome. We found that cathepsin L, a lysosomal cysteine protease, cleaves GCase and regulates its stability. In support of these data, GCase protein was elevated in the brain of cathepsin L-KO mice. Chemical inhibition of cathepsin L increased both GCase levels and activity in fibroblasts from patients with GD. Importantly, inhibition of cathepsin L in dopaminergic neurons from a patient GBA1-PD led to increased GCase levels and activity as well as reduced phosphorylated α-synuclein. These results suggest that targeting cathepsin L-mediated GCase degradation represents a potential therapeutic strategy for GCase deficiency in PD and related disorders that exhibit decreased GCase activity.
Collapse
Affiliation(s)
- Myung Jong Kim
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Soojin Kim
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Medical Faculty and BIOSS Centre for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
21
|
Yang N, Sun M, Wang H, Hu D, Zhang A, Khan S, Chen Z, Chen D, Xie S. Progress of stimulus responsive nanosystems for targeting treatment of bacterial infectious diseases. Adv Colloid Interface Sci 2024; 324:103078. [PMID: 38215562 DOI: 10.1016/j.cis.2024.103078] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
In recent decades, due to insufficient concentration at the lesion site, low bioavailability and increasingly serious resistance, antibiotics have become less and less dominant in the treatment of bacterial infectious diseases. It promotes the development of efficient drug delivery systems, and is expected to achieve high absorption, targeted drug release and satisfactory therapy effects. A variety of endogenous stimulation-responsive nanosystems have been constructed by using special infection microenvironments (pH, enzymes, temperature, etc.). In this review, we firstly provide an extensive review of the current research progress in antibiotic treatment dilemmas and drug delivery systems. Then, the mechanism of microenvironment characteristics of bacterial infected lesions was elucidated to provide a strong theoretical basis for bacteria-targeting nanosystems design. In particular, the discussion focuses on the design principles of single-stimulus and dual-stimulus responsive nanosystems, as well as the use of endogenous stimulus-responsive nanosystems to deliver antimicrobial agents to target locations for combating bacterial infectious diseases. Finally, the challenges and prospects of endogenous stimulus-responsive nanosystems were summarized.
Collapse
Affiliation(s)
- Niuniu Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Mengyuan Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Huixin Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Danlei Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Aoxue Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Suliman Khan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Zhen Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
22
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
23
|
Zhu L, Zeng Q, Wang J, Deng F, Jin S. Cathepsin V drives lung cancer progression by shaping the immunosuppressive environment and adhesion molecules cleavage. Aging (Albany NY) 2023; 15:13961-13979. [PMID: 38078882 PMCID: PMC10756122 DOI: 10.18632/aging.205278] [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: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/21/2023]
Abstract
Cathepsin V (CTSV) is a cysteine cathepsin protease that plays a crucial role in extracellular matrix degradation. CTSV is correlated with poor prognosis in various cancers, but the underlying mechanism remains elusive. Here, we observed that CSTV is upregulated in lung cancer and is a poor prognosis factor for lung cancer. CTSV acts as a driver in the metastasis of lung cancer both in vitro and in vivo. CTSV promotes lung cancer metastasis by downregulating adhesion molecules, including fibronectin, E-cadherin, and N-cadherin. Our data revealed that CTSV functions by mediating the fragmentation of fibronectin, E-cadherin, and N-cadherin in cleavage, remodeling the extracellular matrix (ECM). The rationally designed antibody targeting CTSV blocks its cleaving ability towards fibronectin, E-cadherin, and N-cadherin, suppressing migration and invasion. Furthermore, we found that CTSV expression is negatively correlated with immune cell infiltration and immune scores and inhibits T cell activity. Targeting CTSV with specific antibodies effectively suppressed lung cancer metastasis in a mouse model. Our study demonstrates the critical role of CTSV in the immunity and metastasis of lung cancer, suggesting that the CTSV-targeting approach is a promising strategy for lung cancer.
Collapse
Affiliation(s)
- Lifei Zhu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Dermatology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qi Zeng
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Jinxiang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Precision Medicine Center, Sun Yat-Sen University, Shenzhen 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shi Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| |
Collapse
|
24
|
Sharma S, Chandra K, Naik A, Sharma A, Sharma R, Thakur A, Grewal AS, Dhingra AK, Banerjee A, Liou JP, Guru SK, Nepali K. Flavone-based dual PARP-Tubulin inhibitor manifesting efficacy against endometrial cancer. J Enzyme Inhib Med Chem 2023; 38:2276665. [PMID: 37919954 PMCID: PMC10627047 DOI: 10.1080/14756366.2023.2276665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/24/2023] [Indexed: 11/04/2023] Open
Abstract
Structural tailoring of the flavone framework (position 7) via organopalladium-catalyzed C-C bond formation was attempted in this study. The impact of substituents with varied electronic effects (phenyl ring, position 2 of the benzopyran scaffold) on the antitumor properties was also assessed. Resultantly, the efforts yielded a furyl arm bearing benzopyran possessing a 4-fluoro phenyl ring (position 2) (14) that manifested a magnificent antitumor profile against the Ishikawa cell lines mediated through dual inhibition of PARP and tubulin [(IC50 (PARP1) = 74 nM, IC50 (PARP2) = 109 nM) and tubulin (IC50 = 1.4 µM)]. Further investigations confirmed the ability of 14 to induce apoptosis as well as autophagy and cause cell cycle arrest at the G2/M phase. Overall, the outcome of the study culminated in a tractable dual PARP-tubulin inhibitor endowed with an impressive activity profile against endometrial cancer.
Collapse
Affiliation(s)
- Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kavya Chandra
- Department of Biological Sciences, BITS Pilani KK Birla Goa campus, Goa, India
| | - Aliva Naik
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | | | | | - Arnab Banerjee
- Department of Biological Sciences, BITS Pilani KK Birla Goa campus, Goa, India
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
Balamkundu S, Liu CF. Lysosomal-Cleavable Peptide Linkers in Antibody-Drug Conjugates. Biomedicines 2023; 11:3080. [PMID: 38002080 PMCID: PMC10669454 DOI: 10.3390/biomedicines11113080] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Antibody-drug Conjugates (ADCs) are a powerful therapeutic modality for cancer treatment. ADCs are multi-functional biologics in which a disease-targeting antibody is conjugated to an effector payload molecule via a linker. The success of currently used ADCs has been largely attributed to the development of linker systems, which allow for the targeted release of cytocidal payload drugs inside cancer cells. Many lysosomal proteases are over expressed in human cancers. They can effectively cleave a variety of peptide sequences, which can be exploited for the design of ADC linker systems. As a well-established linker, valine-citrulline-p-aminobenzyl carbamate (ValCitPABC) is used in many ADCs that are already approved or under preclinical and clinical development. Although ValCitPABC and related linkers are readily cleaved by cathepsins in the lysosome while remaining reasonably stable in human plasma, many studies have shown that they are susceptible to carboxylesterase 1C (Ces1C) in mouse and rat plasma, which hinders the preclinical evaluation of ADCs. Furthermore, neutropenia and thrombocytopenia, two of the most commonly observed dose-limiting adverse effects of ADCs, are believed to result from the premature hydrolysis of ValCitPABC by human neutrophil elastase. In addition to ValCitPABC, the GGFG tetrapeptidyl-aminomethoxy linker is also cathepsin-cleavable and is used in the highly successful ADC drug, DS8201a. In addition to cathepsin-cleavable linkers, there is also growing interest in legumain-sensitive linkers for ADC development. Increasing plasma stability while maintaining lysosomal cleavability of ADC linkers is an objective of intensive current research. This review reports recent advances in the design and structure-activity relationship studies of various peptide/peptidomimetic linkers in this field.
Collapse
Affiliation(s)
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
26
|
Stoka V, Vasiljeva O, Nakanishi H, Turk V. The Role of Cysteine Protease Cathepsins B, H, C, and X/Z in Neurodegenerative Diseases and Cancer. Int J Mol Sci 2023; 24:15613. [PMID: 37958596 PMCID: PMC10650516 DOI: 10.3390/ijms242115613] [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: 07/22/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Papain-like cysteine proteases are composed of 11 human cysteine cathepsins, originally located in the lysosomes. They exhibit broad specificity and act as endopeptidases and/or exopeptidases. Among them, only cathepsins B, H, C, and X/Z exhibit exopeptidase activity. Recently, cysteine cathepsins have been found to be present outside the lysosomes and often participate in various pathological processes. Hence, they have been considered key signalling molecules. Their potentially hazardous proteolytic activities are tightly regulated. This review aims to discuss recent advances in understanding the structural aspects of these four cathepsins, mechanisms of their zymogen activation, regulation of their activities, and functional aspects of these enzymes in neurodegeneration and cancer. Neurodegenerative effects have been evaluated, particularly in Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and neuropsychiatric disorders. Cysteine cathepsins also participate in tumour progression and metastasis through the overexpression and secretion of proteases, which trigger extracellular matrix degradation. To our knowledge, this is the first review to provide an in-depth analysis regarding the roles of cysteine cathepsins B, H, C, and X in neurodegenerative diseases and cancer. Further advances in understanding the functions of cysteine cathepsins in these conditions will result in the development of novel, targeted therapeutic strategies.
Collapse
Affiliation(s)
- Veronika Stoka
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, SI-1000 Ljubljana, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- CytomX Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women’s University, Hiroshima 731-0153, Japan;
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia;
- Jožef Stefan International Postgraduate School, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
27
|
Ildiz ES, Gvozdenovic A, Kovacs WJ, Aceto N. Travelling under pressure - hypoxia and shear stress in the metastatic journey. Clin Exp Metastasis 2023; 40:375-394. [PMID: 37490147 PMCID: PMC10495280 DOI: 10.1007/s10585-023-10224-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023]
Abstract
Cancer cell invasion, intravasation and survival in the bloodstream are early steps of the metastatic process, pivotal to enabling the spread of cancer to distant tissues. Circulating tumor cells (CTCs) represent a highly selected subpopulation of cancer cells that tamed these critical steps, and a better understanding of their biology and driving molecular principles may facilitate the development of novel tools to prevent metastasis. Here, we describe key research advances in this field, aiming at describing early metastasis-related processes such as collective invasion, shedding, and survival of CTCs in the bloodstream, paying particular attention to microenvironmental factors like hypoxia and mechanical stress, considered as important influencers of the metastatic journey.
Collapse
Affiliation(s)
- Ece Su Ildiz
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Werner J Kovacs
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland.
| |
Collapse
|
28
|
Gutierrez-Ruiz OL, Johnson KM, Krueger EW, Nooren RE, Cruz-Reyes N, Heppelmann CJ, Hogenson TL, Fernandez-Zapico ME, McNiven MA, Razidlo GL. Ectopic expression of DOCK8 regulates lysosome-mediated pancreatic tumor cell invasion. Cell Rep 2023; 42:113042. [PMID: 37651233 PMCID: PMC10591794 DOI: 10.1016/j.celrep.2023.113042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 06/22/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023] Open
Abstract
Amplified lysosome activity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) orchestrated by oncogenic KRAS that mediates tumor growth and metastasis, though the mechanisms underlying this phenomenon remain unclear. Using comparative proteomics, we found that oncogenic KRAS significantly enriches levels of the guanine nucleotide exchange factor (GEF) dedicator of cytokinesis 8 (DOCK8) on lysosomes. Surprisingly, DOCK8 is aberrantly expressed in a subset of PDAC, where it promotes cell invasion in vitro and in vivo. DOCK8 associates with lysosomes and regulates lysosomal morphology and motility, with loss of DOCK8 leading to increased lysosome size. DOCK8 promotes actin polymerization at the surface of lysosomes while also increasing the proteolytic activity of the lysosomal protease cathepsin B. Critically, depletion of DOCK8 significantly reduces cathepsin-dependent extracellular matrix degradation and impairs the invasive capacity of PDAC cells. These findings implicate ectopic expression of DOCK8 as a key driver of KRAS-driven lysosomal regulation and invasion in pancreatic cancer cells.
Collapse
Affiliation(s)
- Omar L Gutierrez-Ruiz
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Katherine M Johnson
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Eugene W Krueger
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Roseanne E Nooren
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicole Cruz-Reyes
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Tara L Hogenson
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark A McNiven
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Gina L Razidlo
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
29
|
Schmidt CJ, Kim DK, Pendarvis GK, Abasht B, McCarthy FM. Proteomic insight into human directed selection of the domesticated chicken Gallus gallus. PLoS One 2023; 18:e0289648. [PMID: 37549140 PMCID: PMC10406324 DOI: 10.1371/journal.pone.0289648] [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: 04/19/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023] Open
Abstract
Chicken domestication began at least 3,500 years ago for purposes of divination, cockfighting, and food. Prior to industrial scale chicken production, domestication selected larger birds with increased egg production. In the mid-20th century companies began intensive selection with the broiler (meat) industry focusing on improved feed conversion, rapid growth, and breast muscle yield. Here we present proteomic analysis comparing the modern broiler line, Ross 708, with the UIUC legacy line which is not selected for growth traits. Breast muscle proteome analysis identifies cellular processes that have responded to human directed artificial selection. Mass spectrometry was used to identify protein level differences in the breast muscle of 6-day old chicks from Modern and Legacy lines. Our results indicate elevated levels of stress proteins, ribosomal proteins and proteins that participate in the innate immune pathway in the Modern chickens. Furthermore, the comparative analyses indicated expression differences for proteins involved in multiple biochemical pathways. In particular, the Modern line had elevated levels of proteins affecting the pentose phosphate pathway, TCA cycle and fatty acid oxidation while proteins involved in the first phase of glycolysis were reduced compared to the Legacy line. These analyses provide hypotheses linking the morphometric changes driven by human directed selection to biochemical pathways. These results also have implications for the poultry industry, specifically Wooden Breast disease which is linked to rapid breast muscle growth.
Collapse
Affiliation(s)
- Carl J. Schmidt
- Department of Animal and Food Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Dong Kyun Kim
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - G Ken Pendarvis
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States of America
| | - Behnam Abasht
- Department of Animal and Food Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Fiona M. McCarthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
30
|
Razi S, Haghparast A, Chodari Khameneh S, Ebrahimi Sadrabadi A, Aziziyan F, Bakhtiyari M, Nabi-Afjadi M, Tarhriz V, Jalili A, Zalpoor H. The role of tumor microenvironment on cancer stem cell fate in solid tumors. Cell Commun Signal 2023; 21:143. [PMID: 37328876 PMCID: PMC10273768 DOI: 10.1186/s12964-023-01129-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/15/2023] [Indexed: 06/18/2023] Open
Abstract
In the last few decades, the role of cancer stem cells in initiating tumors, metastasis, invasion, and resistance to therapies has been recognized as a potential target for tumor therapy. Understanding the mechanisms by which CSCs contribute to cancer progression can help to provide novel therapeutic approaches against solid tumors. In this line, the effects of mechanical forces on CSCs such as epithelial-mesenchymal transition, cellular plasticity, etc., the metabolism pathways of CSCs, players of the tumor microenvironment, and their influence on the regulating of CSCs can lead to cancer progression. This review focused on some of these mechanisms of CSCs, paving the way for a better understanding of their regulatory mechanisms and developing platforms for targeted therapies. While progress has been made in research, more studies will be required in the future to explore more aspects of how CSCs contribute to cancer progression. Video Abstract.
Collapse
Affiliation(s)
- Sara Razi
- Vira Pioneers of Modern Science (VIPOMS), Tehran, Iran
| | | | | | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran
- Cytotech and Bioinformatics Research Group, Tehran, Iran
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vahideh Tarhriz
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, P.O. Box 5163639888, Tabriz, Iran.
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran.
- Parvaz Research Ideas Supporter Institute, Tehran, Iran.
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
31
|
Frolova AS, Chepikova OE, Deviataikina AS, Solonkina AD, Zamyatnin AA. New Perspectives on the Role of Nuclear Proteases in Cell Death Pathways. BIOLOGY 2023; 12:797. [PMID: 37372081 DOI: 10.3390/biology12060797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
Multiple factors can trigger cell death via various pathways, and nuclear proteases have emerged as essential regulators of these processes. While certain nuclear proteases have been extensively studied and their mechanisms of action are well understood, others remain poorly characterized. Regulation of nuclear protease activity is a promising therapeutic strategy that could selectively induce favorable cell death pathways in specific tissues or organs. Thus, by understanding the roles of newly discovered or predicted nuclear proteases in cell death processes, we can identify new pharmacological targets for improving therapeutic outcomes. In this article, we delved into the role of nuclear proteases in several types of cell death and explore potential avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Anastasia S Frolova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Olga E Chepikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anna S Deviataikina
- Institute of Biodesign and Complex Systems Modeling, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Alena D Solonkina
- Institute of Biodesign and Complex Systems Modeling, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
32
|
Pérez-Díez I, Andreu Z, Hidalgo MR, Perpiñá-Clérigues C, Fantín L, Fernandez-Serra A, de la Iglesia-Vaya M, Lopez-Guerrero JA, García-García F. A Comprehensive Transcriptional Signature in Pancreatic Ductal Adenocarcinoma Reveals New Insights into the Immune and Desmoplastic Microenvironments. Cancers (Basel) 2023; 15:2887. [PMID: 37296850 PMCID: PMC10252129 DOI: 10.3390/cancers15112887] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) prognoses and treatment responses remain devastatingly poor due partly to the highly heterogeneous, aggressive, and immunosuppressive nature of this tumor type. The intricate relationship between the stroma, inflammation, and immunity remains vaguely understood in the PDAC microenvironment. Here, we performed a meta-analysis of stroma-, and immune-related gene expression in the PDAC microenvironment to improve disease prognosis and therapeutic development. We selected 21 PDAC studies from the Gene Expression Omnibus and ArrayExpress databases, including 922 samples (320 controls and 602 cases). Differential gene enrichment analysis identified 1153 significant dysregulated genes in PDAC patients that contribute to a desmoplastic stroma and an immunosuppressive environment (the hallmarks of PDAC tumors). The results highlighted two gene signatures related to the immune and stromal environments that cluster PDAC patients into high- and low-risk groups, impacting patients' stratification and therapeutic decision making. Moreover, HCP5, SLFN13, IRF9, IFIT2, and IFI35 immune genes are related to the prognosis of PDAC patients for the first time.
Collapse
Affiliation(s)
- Irene Pérez-Díez
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, 46012 Valencia, Spain
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
| | - Zoraida Andreu
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
| | - Marta R. Hidalgo
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
| | - Carla Perpiñá-Clérigues
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
| | - Lucía Fantín
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain
| | - Antonio Fernandez-Serra
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain
| | - María de la Iglesia-Vaya
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, 46012 Valencia, Spain
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
| | - José A. Lopez-Guerrero
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain
- Department of Pathology, Medical School, Catholic University of Valencia, 46001 Valencia, Spain
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain
- IVO-CIPF Joint Research Unit of Cancer, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain; (Z.A.)
| |
Collapse
|
33
|
Hey G, Rao R, Carter A, Reddy A, Valle D, Patel A, Patel D, Lucke-Wold B, Pomeranz Krummel D, Sengupta S. Ligand-Gated Ion Channels: Prognostic and Therapeutic Implications for Gliomas. J Pers Med 2023; 13:jpm13050853. [PMID: 37241023 DOI: 10.3390/jpm13050853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Gliomas are common primary brain malignancies that remain difficult to treat due to their overall aggressiveness and heterogeneity. Although a variety of therapeutic strategies have been employed for the treatment of gliomas, there is increasing evidence that suggests ligand-gated ion channels (LGICs) can serve as a valuable biomarker and diagnostic tool in the pathogenesis of gliomas. Various LGICs, including P2X, SYT16, and PANX2, have the potential to become altered in the pathogenesis of glioma, which can disrupt the homeostatic activity of neurons, microglia, and astrocytes, further exacerbating the symptoms and progression of glioma. Consequently, LGICs, including purinoceptors, glutamate-gated receptors, and Cys-loop receptors, have been targeted in clinical trials for their potential therapeutic benefit in the diagnosis and treatment of gliomas. In this review, we discuss the role of LGICs in the pathogenesis of glioma, including genetic factors and the effect of altered LGIC activity on the biological functioning of neuronal cells. Additionally, we discuss current and emerging investigations regarding the use of LGICs as a clinical target and potential therapeutic for gliomas.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rohan Rao
- College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ashley Carter
- Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Akshay Reddy
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Daisy Valle
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Anjali Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Drashti Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 23608, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Soma Sengupta
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
34
|
Pukkanasut P, Whitt J, Guenter R, Lynch SE, Gallegos C, Rosendo-Pineda MJ, Gomora JC, Chen H, Lin D, Sorace A, Jaskula-Sztul R, Velu SE. Voltage-Gated Sodium Channel Na V1.7 Inhibitors with Potent Anticancer Activities in Medullary Thyroid Cancer Cells. Cancers (Basel) 2023; 15:2806. [PMID: 37345144 PMCID: PMC10216335 DOI: 10.3390/cancers15102806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Our results from quantitative RT-PCR, Western blotting, immunohistochemistry, and the tissue microarray of medullary thyroid cancer (MTC) cell lines and patient specimens confirm that VGSC subtype NaV1.7 is uniquely expressed in aggressive MTC and not expressed in normal thyroid cells and tissues. We establish the druggability of NaV1.7 in MTC by identifying a novel inhibitor (SV188) and investigate its mode of binding and ability to inhibit INa current in NaV1.7. The whole-cell patch-clamp studies of the SV188 in the NaV1.7 channels expressed in HEK-293 cells show that SV188 inhibited the INa current in NaV1.7 with an IC50 value of 3.6 µM by a voltage- and use-dependent blockade mechanism, and the maximum inhibitory effect is observed when the channel is open. SV188 inhibited the viability of MTC cell lines, MZ-CRC-1 and TT, with IC50 values of 8.47 μM and 9.32 μM, respectively, and significantly inhibited the invasion of MZ-CRC-1 cells by 35% and 52% at 3 μM and 6 μM, respectively. In contrast, SV188 had no effect on the invasion of TT cells derived from primary tumor, which have lower basal expression of NaV1.7. In addition, SV188 at 3 μM significantly inhibited the migration of MZ-CRC-1 and TT cells by 27% and 57%, respectively.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Jason Whitt
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
| | - Rachael Guenter
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
| | - Shannon E. Lynch
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
| | - Carlos Gallegos
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Margarita Jacaranda Rosendo-Pineda
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.J.R.-P.); (J.C.G.)
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.J.R.-P.); (J.C.G.)
| | - Herbert Chen
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Diana Lin
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Anna Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
35
|
Aganovic A. pH-dependent endocytosis mechanisms for influenza A and SARS-coronavirus. Front Microbiol 2023; 14:1190463. [PMID: 37234537 PMCID: PMC10206014 DOI: 10.3389/fmicb.2023.1190463] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic and the influenza epidemics have revived the interest in understanding how these highly contagious enveloped viruses respond to alterations in the physicochemical properties of their microenvironment. By understanding the mechanisms and conditions by which viruses exploit the pH environment of the host cell during endocytosis, we can gain a better understanding of how they respond to pH-regulated anti-viral therapies but also pH-induced changes in extracellular environments. This review provides a detailed explanation of the pH-dependent viral structural changes preceding and initiating viral disassembly during endocytosis for influenza A (IAV) and SARS coronaviruses. Drawing upon extensive literature from the last few decades and latest research, I analyze and compare the circumstances in which IAV and SARS-coronavirus can undertake endocytotic pathways that are pH-dependent. While there are similarities in the pH-regulated patterns leading to fusion, the mechanisms and pH activation differ. In terms of fusion activity, the measured activation pH values for IAV, across all subtypes and species, vary between approximately 5.0 to 6.0, while SARS-coronavirus necessitates a lower pH of 6.0 or less. The main difference between the pH-dependent endocytic pathways is that the SARS-coronavirus, unlike IAV, require the presence of specific pH-sensitive enzymes (cathepsin L) during endosomal transport. Conversely, the conformational changes in the IAV virus under acidic conditions in endosomes occur due to the specific envelope glycoprotein residues and envelope protein ion channels (viroporins) getting protonated by H+ ions. Despite extensive research over several decades, comprehending the pH-triggered conformational alterations of viruses still poses a significant challenge. The precise mechanisms of protonation mechanisms of certain during endosomal transport for both viruses remain incompletely understood. In absence of evidence, further research is needed.
Collapse
Affiliation(s)
- Amar Aganovic
- Faculty of Engineering Science and Technology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
36
|
Li S, Pritchard DM, Yu LG. Galectin-3 promotes secretion of proteases that decrease epithelium integrity in human colon cancer cells. Cell Death Dis 2023; 14:268. [PMID: 37055381 PMCID: PMC10102123 DOI: 10.1038/s41419-023-05789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023]
Abstract
Galectin-3 is a galactoside-binding protein that is commonly overexpressed in many epithelial cancers. It is increasingly recognized as a multi-functional, multi-mode promoter in cancer development, progression, and metastasis. This study reports that galectin-3 secretion by human colon cancer cells induces cancer cell secretion, in an autocrine/paracrine manner, of a number of proteases including cathepsin-B, MMP-1 and MMP-13. The secretion of these proteases causes disruption of epithelial monolayer integrity, increases its permeability and promotes tumour cell invasion. This effect of galectin-3 is shown to be mediated through induction of cellular PYK2-GSK3α/β signalling and can be prevented by the presence of galectin-3 binding inhibitors. This study thus reveals an important mechanism in galectin-3-mediated promotion of cancer progression and metastasis. It provides further evidence to the increased realization of galectin-3 as a potential therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Shun Li
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - David Mark Pritchard
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lu-Gang Yu
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
37
|
Du W, Gong XL, Tian Y, Zhu X, Peng Y, Wang YW. Coumarin-Based Fluorescence Probe for Differentiated Detection of Biothiols and Its Bioimaging in Cells. BIOSENSORS 2023; 13:bios13040447. [PMID: 37185522 PMCID: PMC10136212 DOI: 10.3390/bios13040447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
In this work, a coumarin derivative, SWJT-14, was synthesized as a fluorescence probe to distinguish cysteine (Cys), homocysteine (Hcy) and glutathione (GSH) in aqueous solutions. The detection limit of Cys, Hcy and GSH for the probe was 0.02 μM, 0.42 μM and 0.92 μM, respectively, which was lower than biothiols in cells. The probe reacted with biothiols to generate different products with different conjugated structures. Additionally, it could distinguish Cys, Hcy and GSH using fluorescence and UV-Vis spectra. The detection mechanism was confirmed by MS. SWJT-14 was successfully used in cellular experiments and detected both endogenous and exogenous biothiols.
Collapse
Affiliation(s)
- Wei Du
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiu-Lin Gong
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Yang Tian
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Xi Zhu
- Department of Neurology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Yu Peng
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Ya-Wen Wang
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
38
|
ElAbd H, Bacher P, Tholey A, Lenz TL, Franke A. Challenges and opportunities in analyzing and modeling peptide presentation by HLA-II proteins. Front Immunol 2023; 14:1107266. [PMID: 37063883 PMCID: PMC10090296 DOI: 10.3389/fimmu.2023.1107266] [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: 11/24/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
The human leukocyte antigen (HLA) proteins are an indispensable component of adaptive immunity because of their role in presenting self and foreign peptides to T cells. Further, many complex diseases are associated with genetic variation in the HLA region, implying an important role for specific HLA-presented peptides in the etiology of these diseases. Identifying the specific set of peptides presented by an individual’s HLA proteins in vivo, as a whole being referred to as the immunopeptidome, has therefore gathered increasing attention for different reasons. For example, identifying neoepitopes for cancer immunotherapy, vaccine development against infectious pathogens, or elucidating the role of HLA in autoimmunity. Despite the tremendous progress made during the last decade in these areas, several questions remain unanswered. In this perspective, we highlight five remaining key challenges in the analysis of peptide presentation and T cell immunogenicity and discuss potential solutions to these problems. We believe that addressing these questions would not only improve our understanding of disease etiology but will also have a direct translational impact in terms of engineering better vaccines and in developing more potent immunotherapies.
Collapse
Affiliation(s)
- Hesham ElAbd
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Petra Bacher
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
- Institute of Immunology, University of Kiel, Kiel, Germany
| | - Andreas Tholey
- Proteomics & Bioanalytics, Institute for Experimental Medicine, University of Kiel, Kiel, Germany
| | - Tobias L. Lenz
- Research Unit for Evolutionary Immunogenomics, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
- *Correspondence: Andre Franke,
| |
Collapse
|
39
|
Behring L, Ruiz-Gómez G, Trapp C, Morales M, Wodtke R, Köckerling M, Kopka K, Pisabarro MT, Pietzsch J, Löser R. Dipeptide-Derived Alkynes as Potent and Selective Irreversible Inhibitors of Cysteine Cathepsins. J Med Chem 2023; 66:3818-3851. [PMID: 36867428 PMCID: PMC10041539 DOI: 10.1021/acs.jmedchem.2c01360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
The potential of designing irreversible alkyne-based inhibitors of cysteine cathepsins by isoelectronic replacement in reversibly acting potent peptide nitriles was explored. The synthesis of the dipeptide alkynes was developed with special emphasis on stereochemically homogeneous products obtained in the Gilbert-Seyferth homologation for C≡C bond formation. Twenty-three dipeptide alkynes and 12 analogous nitriles were synthesized and investigated for their inhibition of cathepsins B, L, S, and K. Numerous combinations of residues at positions P1 and P2 as well as terminal acyl groups allowed for the derivation of extensive structure-activity relationships, which were rationalized by computational covalent docking for selected examples. The determined inactivation constants of the alkynes at the target enzymes span a range of >3 orders of magnitude (3-10 133 M-1 s-1). Notably, the selectivity profiles of alkynes do not necessarily reflect those of the nitriles. Inhibitory activity at the cellular level was demonstrated for selected compounds.
Collapse
Affiliation(s)
- Lydia Behring
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| | - Gloria Ruiz-Gómez
- BIOTEC, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Christian Trapp
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Maryann Morales
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Robert Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Martin Köckerling
- Institute of Chemistry, University of Rostock, Albert-Einstein-Straße 3a, 18059 Rostock, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| | - M Teresa Pisabarro
- BIOTEC, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Mommsenstraße 4, 01069 Dresden, Germany
| |
Collapse
|
40
|
Canè S, Barouni RM, Fabbi M, Cuozzo J, Fracasso G, Adamo A, Ugel S, Trovato R, De Sanctis F, Giacca M, Lawlor R, Scarpa A, Rusev B, Lionetto G, Paiella S, Salvia R, Bassi C, Mandruzzato S, Ferrini S, Bronte V. Neutralization of NET-associated human ARG1 enhances cancer immunotherapy. Sci Transl Med 2023; 15:eabq6221. [PMID: 36921034 DOI: 10.1126/scitranslmed.abq6221] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Myeloid cells can restrain antitumor immunity by metabolic pathways, such as the degradation of l-arginine, whose concentrations are regulated by the arginase 1 (ARG1) enzyme. Results from preclinical studies indicate the important role of arginine metabolism in pancreatic ductal adenocarcinoma (PDAC) progression, suggesting a potential for clinical application; however, divergent evolution in ARG1 expression and function in rodents and humans has restricted clinical translation. To overcome this dichotomy, here, we show that neutrophil extracellular traps (NETs), released by spontaneously activated neutrophils isolated from patients with PDAC, create a microdomain where cathepsin S (CTSS) cleaves human (h)ARG1 into different molecular forms endowed with enhanced enzymatic activity at physiological pH. NET-associated hARG1 suppresses T lymphocytes whose proliferation is restored by either adding a hARG1-specific monoclonal antibody (mAb) or preventing CTSS-mediated cleavage, whereas small-molecule inhibitors are not effective. We show that ARG1 blockade, combined with immune checkpoint inhibitors, can restore CD8+ T cell function in ex vivo PDAC tumors. Furthermore, anti-hARG1 mAbs increase the frequency of adoptively transferred tumor-specific CD8+ T cells in tumor and enhance the effectiveness of immune checkpoint therapy in humanized mice. Thus, this study shows that extracellular ARG1, released by activated myeloid cells, localizes in NETs, where it interacts with CTSS that in turn cleaves ARG1, producing major molecular forms endowed with different enzymatic activity at physiological pH. Once exocytosed, ARG1 activity can be targeted by mAbs, which bear potential for clinical application for the treatment of PDAC and require further exploration.
Collapse
Affiliation(s)
- Stefania Canè
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | - Roza Maria Barouni
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | - Marina Fabbi
- Ospedale Policlinico San Martino, IRCCS, Genova16132, Italy
| | - John Cuozzo
- ZebiAI Therapeutics Inc., Waltham, MA 02467, USA
| | - Giulio Fracasso
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | - Annalisa Adamo
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | - Stefano Ugel
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | - Rosalinda Trovato
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | - Francesco De Sanctis
- Department of Medicine, Section of Immunology, University of Verona, Verona 37129, Italy
| | | | - Rita Lawlor
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona 37134, Italy
| | - Aldo Scarpa
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona 37134, Italy.,Department of Diagnostic and Public Health, University of Verona, Verona 37134, Italy
| | - Borislav Rusev
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona 37134, Italy.,Department of Diagnostic and Public Health, University of Verona, Verona 37134, Italy
| | - Gabriella Lionetto
- General and Pancreatic Surgery, Pancreas Institute, University of Verona, Verona 37134, Italy
| | - Salvatore Paiella
- General and Pancreatic Surgery, Pancreas Institute, University of Verona, Verona 37134, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery, Pancreas Institute, University of Verona, Verona 37134, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery, Pancreas Institute, University of Verona, Verona 37134, Italy
| | - Susanna Mandruzzato
- Dipartimento di Scienze Chirurgiche Oncologiche e Gastroenterologiche, University of Padova, Padova 35122, Italy.,Istituto Oncologico Veneto IRCCS, Padova 35128, Italy
| | | | | |
Collapse
|
41
|
Atmaca U, Saglamtas R, Sert Y, Çelik M, Gülçin İ. Metal‐Free Synthesis via Intramolecular Cyclization, Enzyme Inhibition Properties and Molecular Docking of Novel Isoindolinones. ChemistrySelect 2023. [DOI: 10.1002/slct.202204578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Affiliation(s)
- Ufuk Atmaca
- Oltu Vocational Collage Atatürk University 25400- Oltu-Erzurum Turkey
- Department of Chemistry Faculty of Science Atatürk University 25240- Erzurum Turkey
| | - Ruya Saglamtas
- Department of Medical Services and Technology Vocational School of Health Services Agri Ibrahim Cecen University 04100- Agri Turkey
| | - Yusuf Sert
- Sorgun Vocational School & Department of Physics Yozgat Bozok University 47800- Yozgat Turkey
| | - Murat Çelik
- Department of Chemistry Faculty of Science Atatürk University 25240- Erzurum Turkey
| | - İlhami Gülçin
- Department of Chemistry Faculty of Science Atatürk University 25240- Erzurum Turkey
| |
Collapse
|
42
|
Sumithaa C, Ganeshpandian M. Half-Sandwich Ruthenium Arene Complexes Bearing Clinically Approved Drugs as Ligands: The Importance of Metal-Drug Synergism in Metallodrug Design. Mol Pharm 2023; 20:1453-1479. [PMID: 36802711 DOI: 10.1021/acs.molpharmaceut.2c01027] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
A novel strategy in metallodrug discovery today is incorporating clinically approved drugs into metal complexes as coordinating ligands. Using this strategy, various drugs have been repurposed to prepare organometallic complexes to overcome the resistance of drugs and to design promising alternatives to currently available metal-based drugs. Notably, the combination of organoruthenium moiety and clinical drug in a single molecule has been shown, in some instances, to enhance pharmacological activity and reduce toxicity in comparison to the parent drug. Thus, for the past two decades, there has been increasing interest in exploiting metal-drug synergism to develop multifunctional organoruthenium drug candidates. Herein, we summarized the recent reports of rationally designed half-sandwich Ru(arene) complexes containing different FDA-approved drugs. This review also focuses on the mode of coordination of drugs, ligand-exchange kinetics, mechanism of action, and structure-activity relationship of organoruthenated complexes containing drugs. We hope this discussion may serve to shed light on future developments in ruthenium-based metallopharmaceuticals.
Collapse
Affiliation(s)
- Chezhiyan Sumithaa
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, India
| | - Mani Ganeshpandian
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, India
| |
Collapse
|
43
|
Bonatto V, Lameiro RF, Rocho FR, Lameira J, Leitão A, Montanari CA. Nitriles: an attractive approach to the development of covalent inhibitors. RSC Med Chem 2023; 14:201-217. [PMID: 36846367 PMCID: PMC9945868 DOI: 10.1039/d2md00204c] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Nitriles have broad applications in medicinal chemistry, with more than 60 small molecule drugs on the market containing the cyano functional group. In addition to the well-known noncovalent interactions that nitriles can perform with macromolecular targets, they are also known to improve drug candidates' pharmacokinetic profiles. Moreover, the cyano group can be used as an electrophilic warhead to covalently bind an inhibitor to a target of interest, forming a covalent adduct, a strategy that can present benefits over noncovalent inhibitors. This approach has gained much notoriety in recent years, mainly with diabetes and COVID-19-approved drugs. Nevertheless, the application of nitriles in covalent ligands is not restricted to it being the reactive center, as it can also be employed to convert irreversible inhibitors into reversible ones, a promising strategy for kinase inhibition and protein degradation. In this review, we introduce and discuss the roles of the cyano group in covalent inhibitors, how to tune its reactivity and the possibility of achieving selectivity only by replacing the warhead. Finally, we provide an overview of nitrile-based covalent compounds in approved drugs and inhibitors recently described in the literature.
Collapse
Affiliation(s)
- Vinícius Bonatto
- Medicinal and Biological Chemistry Group, São Carlos Institute of Chemistry, University of São Paulo Avenue Trabalhador Sancarlense, 400 13566-590 São Carlos/SP Brazil
| | - Rafael F Lameiro
- Medicinal and Biological Chemistry Group, São Carlos Institute of Chemistry, University of São Paulo Avenue Trabalhador Sancarlense, 400 13566-590 São Carlos/SP Brazil
| | - Fernanda R Rocho
- Medicinal and Biological Chemistry Group, São Carlos Institute of Chemistry, University of São Paulo Avenue Trabalhador Sancarlense, 400 13566-590 São Carlos/SP Brazil
| | - Jerônimo Lameira
- Medicinal and Biological Chemistry Group, São Carlos Institute of Chemistry, University of São Paulo Avenue Trabalhador Sancarlense, 400 13566-590 São Carlos/SP Brazil
- Institute of Biological Science, Federal University of Pará Rua Augusto Correa S/N Belém PA Brazil
| | - Andrei Leitão
- Medicinal and Biological Chemistry Group, São Carlos Institute of Chemistry, University of São Paulo Avenue Trabalhador Sancarlense, 400 13566-590 São Carlos/SP Brazil
| | - Carlos A Montanari
- Medicinal and Biological Chemistry Group, São Carlos Institute of Chemistry, University of São Paulo Avenue Trabalhador Sancarlense, 400 13566-590 São Carlos/SP Brazil
| |
Collapse
|
44
|
Xing Y, Yang M, Chen X. Fabrication of P and N Co-Doped Carbon Dots for Fe 3+ Detection in Serum and Lysosomal Tracking in Living Cells. BIOSENSORS 2023; 13:230. [PMID: 36831996 PMCID: PMC9954533 DOI: 10.3390/bios13020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Doping with heteroatoms allows the retention of the general characteristics of carbon dots while allowing their physicochemical and photochemical properties to be effectively modulated. In this work, we report the preparation of ultrastable P and N co-doped carbon dots (PNCDs) that can be used for the highly selective detection of Fe3+ and the tracking of lysosomes in living cells. Fluorescent PNCDs were facilely prepared via a hydrothermal treatment of ethylenediamine and phytic acid, and they exhibited a high quantum yield of 22.0%. The strong coordination interaction between the phosphorus groups of PNCDs and Fe3+ rendered them efficient probes for use in selective Fe3+ detection, with a detection limit of 0.39 μM, and we demonstrated their practicability by accurately detecting the Fe3+ contents in bio-samples. At the same time, PNCDs exhibited high lysosomal location specificity in different cell lines due to surface lipophilic amino groups, and real-time tracking of the lysosome morphology in HeLa cells was achieved. The present work suggests that the fabrication of heteroatom-doped CDs might be an effective strategy to provide promising tools for cytology, such as organelle tracking.
Collapse
Affiliation(s)
- Yanzhi Xing
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Mei Yang
- School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Xuwei Chen
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| |
Collapse
|
45
|
Linders DGJ, Bijlstra OD, Fallert LC, Hilling DE, Walker E, Straight B, March TL, Valentijn ARPM, Pool M, Burggraaf J, Basilion JP, Vahrmeijer AL, Kuppen PJK. Cysteine Cathepsins in Breast Cancer: Promising Targets for Fluorescence-Guided Surgery. Mol Imaging Biol 2023; 25:58-73. [PMID: 36002710 PMCID: PMC9971096 DOI: 10.1007/s11307-022-01768-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022]
Abstract
The majority of breast cancer patients is treated with breast-conserving surgery (BCS) combined with adjuvant radiation therapy. Up to 40% of patients has a tumor-positive resection margin after BCS, which necessitates re-resection or additional boost radiation. Cathepsin-targeted near-infrared fluorescence imaging during BCS could be used to detect residual cancer in the surgical cavity and guide additional resection, thereby preventing tumor-positive resection margins and associated mutilating treatments. The cysteine cathepsins are a family of proteases that play a major role in normal cellular physiology and neoplastic transformation. In breast cancer, the increased enzymatic activity and aberrant localization of many of the cysteine cathepsins drive tumor progression, proliferation, invasion, and metastasis. The upregulation of cysteine cathepsins in breast cancer cells indicates their potential as a target for intraoperative fluorescence imaging. This review provides a summary of the current knowledge on the role and expression of the most important cysteine cathepsins in breast cancer to better understand their potential as a target for fluorescence-guided surgery (FGS). In addition, it gives an overview of the cathepsin-targeted fluorescent probes that have been investigated preclinically and in breast cancer patients. The current review underscores that cysteine cathepsins are highly suitable molecular targets for FGS because of favorable expression and activity patterns in virtually all breast cancer subtypes. This is confirmed by cathepsin-targeted fluorescent probes that have been shown to facilitate in vivo breast cancer visualization and tumor resection in mouse models and breast cancer patients. These findings indicate that cathepsin-targeted FGS has potential to improve treatment outcomes in breast cancer patients.
Collapse
Affiliation(s)
- Daan G. J. Linders
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Okker D. Bijlstra
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Laura C. Fallert
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Denise E. Hilling
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Ethan Walker
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH 44106 USA
| | | | - Taryn L. March
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - A. Rob P. M. Valentijn
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Martin Pool
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jacobus Burggraaf
- Centre for Human Drug Research, 2333 CL Leiden, The Netherlands
- Leiden Academic Center for Drug Research, 2333 AL Leiden, The Netherlands
| | - James P. Basilion
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH 44106 USA
- Department of Radiology, Case School of Medicine, Case Western Reserve University, Cleveland, OH 44106 USA
| | | | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
46
|
Lysosomes as a Target of Anticancer Therapy. Int J Mol Sci 2023; 24:ijms24032176. [PMID: 36768500 PMCID: PMC9916765 DOI: 10.3390/ijms24032176] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Lysosomes are organelles containing acidic hydrolases that are responsible for lysosomal degradation and the maintenance of cellular homeostasis. They play an important role in autophagy, as well as in various cell death pathways, such as lysosomal and apoptotic death. Various agents, including drugs, can induce lysosomal membrane permeability, resulting in the translocation of acidic hydrolases into the cytoplasm, which promotes lysosomal-mediated death. This type of death may be of great importance in anti-cancer therapy, as both cancer cells with disturbed pathways leading to apoptosis and drug-resistant cells can undergo it. Important compounds that damage the lysosomal membrane include lysosomotropic compounds, antihistamines, immunosuppressants, DNA-damaging drugs, chemotherapeutics, photosensitizers and various plant compounds. An interesting approach in the treatment of cancer and the search for ways to overcome the chemoresistance of cancer cells may also be combining lysosomotropic compounds with targeted modulators of autophagy to induce cell death. These compounds may be an alternative in oncological treatment, and lysosomes may become a promising therapeutic target for many diseases, including cancer. Understanding the functional relationships between autophagy and apoptosis and the possibilities of their regulation, both in relation to normal and cancer cells, can be used to develop new and more effective anticancer therapies.
Collapse
|
47
|
Role and Application of Biocatalysts in Cancer Drug Discovery. Catalysts 2023. [DOI: 10.3390/catal13020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A biocatalyst is an enzyme that speeds up or slows down the rate at which a chemical reaction occurs and speeds up certain processes by 108 times. It is used as an anticancer agent because it targets drug activation inside the tumor microenvironment while limiting damage to healthy cells. Biocatalysts have been used for the synthesis of different heterocyclic compounds and is also used in the nano drug delivery systems. The use of nano-biocatalysts for tumor-targeted delivery not only aids in tumor invasion, angiogenesis, and mutagenesis, but also provides information on the expression and activity of many markers related to the microenvironment. Iosmapinol, moclobemide, cinepazide, lysine dioxygenase, epothilone, 1-homophenylalanine, and many more are only some of the anticancer medicines that have been synthesised using biocatalysts. In this review, we have highlighted the application of biocatalysts in cancer therapies as well as the use of biocatalysts in the synthesis of drugs and drug-delivery systems in the tumor microenvironment.
Collapse
|
48
|
Peyton SR, Platt MO, Cukierman E. Challenges and Opportunities Modeling the Dynamic Tumor Matrisome. BME FRONTIERS 2023; 4:0006. [PMID: 37849664 PMCID: PMC10521682 DOI: 10.34133/bmef.0006] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/28/2022] [Indexed: 10/19/2023] Open
Abstract
We need novel strategies to target the complexity of cancer and, particularly, of metastatic disease. As an example of this complexity, certain tissues are particularly hospitable environments for metastases, whereas others do not contain fertile microenvironments to support cancer cell growth. Continuing evidence that the extracellular matrix (ECM) of tissues is one of a host of factors necessary to support cancer cell growth at both primary and secondary tissue sites is emerging. Research on cancer metastasis has largely been focused on the molecular adaptations of tumor cells in various cytokine and growth factor environments on 2-dimensional tissue culture polystyrene plates. Intravital imaging, conversely, has transformed our ability to watch, in real time, tumor cell invasion, intravasation, extravasation, and growth. Because the interstitial ECM that supports all cells in the tumor microenvironment changes over time scales outside the possible window of typical intravital imaging, bioengineers are continuously developing both simple and sophisticated in vitro controlled environments to study tumor (and other) cell interactions with this matrix. In this perspective, we focus on the cellular unit responsible for upholding the pathologic homeostasis of tumor-bearing organs, cancer-associated fibroblasts (CAFs), and their self-generated ECM. The latter, together with tumoral and other cell secreted factors, constitute the "tumor matrisome". We share the challenges and opportunities for modeling this dynamic CAF/ECM unit, the tools and techniques available, and how the tumor matrisome is remodeled (e.g., via ECM proteases). We posit that increasing information on tumor matrisome dynamics may lead the field to alternative strategies for personalized medicine outside genomics.
Collapse
Affiliation(s)
- Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Manu O. Platt
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| |
Collapse
|
49
|
Huang H, Zhang Y, Xu X, Liu Y, Zhao J, Ma L, Lei J, Ge W, Li N, Ma E, Li Y, Yuan L. Design and synthesis of dual cathepsin L and S inhibitors and antimetastatic activity evaluation in pancreatic cancer cells. Bioorg Med Chem Lett 2023; 80:129087. [PMID: 36427655 DOI: 10.1016/j.bmcl.2022.129087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 10/27/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022]
Abstract
Currently, the migration and invasion of cancer cells remain the main factors of poor prognosis in the majority of cancer patients. Developing an effective antimetastatic agent is crucial for cancer therapy. Our recent research revealed that Cat L and S are expressed concurrently in metastatic pancreatic cancer cells. Asperphenamate analog ASPER-29, which exhibits dual Cat L and S inhibitory potency, showed a definite antimetastatic effect on pancreatic cancer BxPC-3 and PANC-1 cells. To further improve the antimetastatic ability of asperphenamate-type molecules, 24 derivatives were designed and synthesized by a scaffold-hopping strategy. The cathepsin inhibitory activity assay results showed that most of the derivatives exhibited dual inhibitory effects on Cat L and S. Among all derivatives, Compound B1a showed the strongest inhibitory activity, with IC50 values of 4.10 ± 0.14 μM and 1.79 ± 0.11 μM, which were 1.5-fold and 2.8-fold more potent than those of positive drugs against Cat L and S, respectively. Further wound-healing and transwell chamber assays demonstrated that B1a presented significant antimetastatic ability in vitro.
Collapse
Affiliation(s)
- Haoqiang Huang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Yi Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Xiaohong Xu
- Center for Drug Evaluation, NMPA, Beijing 100022, PR China
| | - Yongzheng Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Juanping Zhao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Lili Ma
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Jie Lei
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Wentao Ge
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Enlong Ma
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yanchun Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Lei Yuan
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China.
| |
Collapse
|
50
|
Barreca M, Lang N, Tarantelli C, Spriano F, Barraja P, Bertoni F. Antibody-drug conjugates for lymphoma patients: preclinical and clinical evidences. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:763-794. [PMID: 36654819 PMCID: PMC9834635 DOI: 10.37349/etat.2022.00112] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/08/2022] [Indexed: 12/28/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a recent, revolutionary approach for malignancies treatment, designed to provide superior efficacy and specific targeting of tumor cells, compared to systemic cytotoxic chemotherapy. Their structure combines highly potent anti-cancer drugs (payloads or warheads) and monoclonal antibodies (Abs), specific for a tumor-associated antigen, via a chemical linker. Because the sensitive targeting capabilities of monoclonal Abs allow the direct delivery of cytotoxic payloads to tumor cells, these agents leave healthy cells unharmed, reducing toxicity. Different ADCs have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of a wide range of malignant conditions, both as monotherapy and in combination with chemotherapy, including for lymphoma patients. Over 100 ADCs are under preclinical and clinical investigation worldwide. This paper it provides an overview of approved and promising ADCs in clinical development for the treatment of lymphoma. Each component of the ADC design, their mechanism of action, and the highlights of their clinical development progress are discussed.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Noémie Lang
- Division of Oncology, Department of Oncology, Faculty of Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
| | - Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| |
Collapse
|