1
|
Vo HVT, Kim N, Lee HJ. Vitamin Bs as Potent Anticancer Agents through MMP-2/9 Regulation. FRONT BIOSCI-LANDMRK 2025; 30:24072. [PMID: 39862072 DOI: 10.31083/fbl24072] [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: 04/26/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 01/27/2025]
Abstract
In recent years, the role of coenzymes, particularly those from the vitamin B group in modulating the activity of metalloenzymes has garnered significant attention in cancer treatment strategies. Metalloenzymes play pivotal roles in various cellular processes, including DNA repair, cell signaling, and metabolism, making them promising targets for cancer therapy. This review explores the complex interplay between coenzymes, specifically vitamin Bs, and metalloenzymes in cancer pathogenesis and treatment. Vitamins are an indispensable part of daily life, essential for optimal health and well-being. Beyond their recognized roles as essential nutrients, vitamins have increasingly garnered attention for their multifaceted functions within the machinery of cellular processes. In particular, vitamin Bs have emerged as a pivotal regulator within this intricate network, exerting profound effects on the functionality of metalloenzymes. Their ability to modulate metalloenzymes involved in crucial cellular pathways implicated in cancer progression presents a compelling avenue for therapeutic intervention. Key findings indicate that vitamin Bs can influence the activity and expression of metalloenzymes, thereby affecting processes such as DNA repair and cell signaling, which are critical in cancer development and progression. Understanding the mechanisms by which these coenzymes regulate metalloenzymes holds great promise for developing novel anticancer strategies. This review summarizes current knowledge on the interactions between vitamin Bs and metalloenzymes, highlighting their potential as anticancer agents and paving the way for innovative, cell-targeted cancer treatments.
Collapse
Affiliation(s)
- Ha Vy Thi Vo
- Department of Chemistry Education, Kongju National University, 32588 Gongju, Chungcheongnam-do, Republic of Korea
| | - Namdoo Kim
- Department of Chemistry, Kongju National University, 32588 Gongju, Chungcheongnam-do, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, 32588 Gongju, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
2
|
Nikfarjam Z, Rakhshi R, Zargari F, Aalikhani M, Hasan-Abad AM, Bazi Z. Repurposing raltegravir for reducing inflammation and treating cancer: a bioinformatics analysis. Sci Rep 2024; 14:30349. [PMID: 39639095 PMCID: PMC11621354 DOI: 10.1038/s41598-024-82065-8] [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: 09/24/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Inflammation is a defensive mechanism that safeguards the human body against detrimental stimuli. Within this intricate process, ADAM17, a zinc-dependent metalloprotease, emerges as an indispensable element, fostering the activation of diverse inflammatory and growth factors within the organism. Nonetheless, ADAM17 malfunctions can augment the rate of growth, inflammatory factors, and subsequent damage. Thus, in this study, we examined and repurposed drugs to suppress the activity of ADAM17. To this end, we employed bioinformatics techniques such as molecular docking, molecular dynamics, and pharmacokinetic studies. Five FDA-approved drugs including Raltegravir, Conivaptan, Paclitaxel, Saquinavir, and Venetoclax with the ability to impede the activity of the ADAM17 metalloenzyme were identified. Moreover, these drugs did not include strong zinc-binding functional groups when verified by the ACE functional group finder. However, further in silico analysis has indicated that Raltegravir demonstrates a commendable interaction with the active site amino acids and exhibits the most favorable pharmacokinetic properties compared to others. Considering the results of bioinformatics tools, it can be concluded that Raltegravir as an antiviral drug could be repurposed to prevent severe inflammatory response and tumorigenesis resulting from ADAM17 malfunction.
Collapse
Affiliation(s)
- Zahra Nikfarjam
- Department of Physical & Computational Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | - Reza Rakhshi
- Department of Medical Biotechnology, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farshid Zargari
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Chemistry, Faculty of Science, University of Sistan and Baluchestan (USB), Zahedan, Iran
| | - Mahdi Aalikhani
- Department of Medical Biotechnology, School of Paramedicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Bazi
- Department of Medical Biotechnology, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
3
|
Sankova MV, Nikolenko VN, Oganesyan MV, Sankov SV, Sinelnikov MY, Suslov AV, Trishina AS, Zharikova TS, Pontes-Silva A, Zharikov YO. Magnesium deficiency and its interaction with the musculoskeletal system, exercise, and connective tissue: an evidence synthesis. SPORT SCIENCES FOR HEALTH 2024; 20:715-726. [DOI: 10.1007/s11332-024-01179-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/06/2024] [Indexed: 09/13/2024]
|
4
|
Zhu S, He J, Yin L, Zhou J, Lian J, Ren Y, Zhang X, Yuan J, Wang G, Li X. Matrix metalloproteinases targeting in prostate cancer. Urol Oncol 2024; 42:275-287. [PMID: 38806387 DOI: 10.1016/j.urolonc.2024.05.002] [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: 02/06/2024] [Revised: 04/07/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Prostate cancer (PCa) is one of the most common tumors affecting men all over the world. PCa has brought a huge health burden to men around the world, especially for elderly men, but its pathogenesis is unclear. In prostate cancer, epigenetic inheritance plays an important role in the development, progression, and metastasis of the disease. An important role in cancer invasion and metastasis is played by matrix metalloproteinases (MMPs), zinc-dependent proteases that break down extracellular matrix. We review two important forms of epigenetic modification and the role of matrix metalloproteinases in tumor regulation, both of which may be of significant value as novel biomarkers for early diagnosis and prognosis monitoring. The author considers that both mechanisms have promising therapeutic applications for therapeutic agent research in prostate cancer, but that efforts should be made to mitigate or eliminate the side effects of drug therapy in order to maximize quality of life of patients. The understanding of epigenetic modification, MMPs, and their inhibitors in the functional regulation of prostate cancer is gradually advancing, it will provide a new technical means for the prevention of prostate cancer, early diagnosis, androgen-independent prostate cancer treatment, and drug research.
Collapse
Affiliation(s)
- Shuying Zhu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Jing He
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Liliang Yin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Jiawei Zhou
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Jiayi Lian
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yanli Ren
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xinling Zhang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Jinghua Yuan
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Gang Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Xiaoping Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China.
| |
Collapse
|
5
|
Jiang Z, Chen L, Huang L, Yu S, Lin J, Li M, Gao Y, Yang L. Bioactive Materials That Promote the Homing of Endogenous Mesenchymal Stem Cells to Improve Wound Healing. Int J Nanomedicine 2024; 19:7751-7773. [PMID: 39099796 PMCID: PMC11297574 DOI: 10.2147/ijn.s455469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024] Open
Abstract
Endogenous stem cell homing refers to the transport of endogenous mesenchymal stem cells (MSCs) to damaged tissue. The paradigm of using well-designed biomaterials to induce resident stem cells to home in to the injured site while coordinating their behavior and function to promote tissue regeneration is known as endogenous regenerative medicine (ERM). ERM is a promising new avenue in regenerative therapy research, and it involves the mobilizing of endogenous stem cells for homing as the principal means through which to achieve it. Comprehending how mesenchymal stem cells home in and grasp the influencing factors of mesenchymal stem cell homing is essential for the understanding and design of tissue engineering. This review summarizes the process of MSC homing, the factors influencing the homing process, analyses endogenous stem cell homing studies of interest in the field of skin tissue repair, explores the integration of endogenous homing promotion strategies with cellular therapies and details tissue engineering strategies that can be used to modulate endogenous homing of stem cells. In addition to providing more systematic theories and ideas for improved materials for endogenous tissue repair, this review provides new perspectives to explore the complex process of tissue remodeling to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Shengxiang Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jiabao Lin
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Mengyao Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
6
|
Lazar AM, Costea DO, Popp CG, Mastalier B. Skin Malignant Melanoma and Matrix Metalloproteinases: Promising Links to Efficient Therapies. Int J Mol Sci 2024; 25:7804. [PMID: 39063046 PMCID: PMC11277423 DOI: 10.3390/ijms25147804] [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: 06/08/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Skin malignant melanoma (MM) is one of the most frequent and aggressive neoplasia worldwide. Its associated high mortality rates are mostly due to its metastases, while diagnosis and treatment of MM in its early stages is of favorable prognostic. Even skin superficial MMs at incipient local stages can already present with lymph node invasion and distant metastases. Therefore, knowledge of the controllable risk factors and pathogenic mechanisms of MM development, spreading, and metastatic pattern, as well as early diagnosis, are essential to decrease the high mortality rates associated with cutaneous malignant melanoma. Genetic factors are incriminated, although lifetime-acquired genetic mutations appear to be even more frequently involved in the development of MM. Skin melanocytes divide only twice per year and have time to accumulate genetic mutations as a consequence of environmental aggressive factors, such as UV exposure. In the search for more promising therapies, matrix metalloproteinases have become of significant interest, such as MMP-1, MMP-2, MMP-9, and MMP-13, which have been linked to more aggressive forms of cancer and earlier metastases. Therefore, the development of specific synthetic inhibitors of MMP secretion or activity could represent a more promising and effective approach to the personalized treatment of MM patients.
Collapse
Affiliation(s)
- Angela Madalina Lazar
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- General Surgery Clinic, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Daniel Ovidiu Costea
- Second Surgery Clinic, Constanta District Clinical Emergency Hospital, 900591 Constanța, Romania
- Department of Surgery, University of Medicine and Pharmacy “Ovidius”, 900470 Constanta, Romania
| | | | - Bogdan Mastalier
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- General Surgery Clinic, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
7
|
Perarivalan I, Karunakaran J, Anbalagan N, Harishma S, Prasad V. Matrix metalloproteinase inhibitors in restorative dentistry. JOURNAL OF CONSERVATIVE DENTISTRY AND ENDODONTICS 2024; 27:566-571. [PMID: 38989495 PMCID: PMC11232771 DOI: 10.4103/jcde.jcde_199_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 07/12/2024]
Abstract
Matrix metalloproteinases (MMPs) have been identified as agents that disintegrate the collagen structures of dental hybrid layers, resulting in reduced restorative bond strength. Multiple MMP inhibitors (MMPIs) are known to counteract this degenerative mechanism, thereby preserving bond strength and promoting the longevity of resin-based restorations. Additionally, literature suggests that certain MMPI materials possess antimicrobial/anticariogenic properties, potentially reducing the risk of secondary caries development. Therefore, this review article aims to narrate on the integration of matrix metalloproteinase inhibitors into adhesive systems and their impact on bond strength.
Collapse
Affiliation(s)
- I Perarivalan
- Department of Conservative Dentistry and Endodontics, Chettinad Dental College and Research Institute, Kelambakkam, Tamil Nadu, India
| | - Janani Karunakaran
- Department of Conservative Dentistry and Endodontics, Chettinad Dental College and Research Institute, Kelambakkam, Tamil Nadu, India
| | - Nathashri Anbalagan
- Department of Conservative Dentistry and Endodontics, Chettinad Dental College and Research Institute, Kelambakkam, Tamil Nadu, India
| | - S Harishma
- Department of Conservative Dentistry and Endodontics, Chettinad Dental College and Research Institute, Kelambakkam, Tamil Nadu, India
| | - Vishnu Prasad
- Department of Conservative Dentistry and Endodontics, Chettinad Dental College and Research Institute, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
8
|
Sampaio Moura N, Schledwitz A, Alizadeh M, Patil SA, Raufman JP. Matrix metalloproteinases as biomarkers and therapeutic targets in colitis-associated cancer. Front Oncol 2024; 13:1325095. [PMID: 38288108 PMCID: PMC10824561 DOI: 10.3389/fonc.2023.1325095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Colorectal cancer (CRC) remains a major cause of morbidity and mortality. Therapeutic approaches for advanced CRC are limited and rarely provide long-term benefit. Enzymes comprising the 24-member matrix metalloproteinase (MMP) family of zinc- and calcium-dependent endopeptidases are key players in extracellular matrix degradation, a requirement for colon tumor expansion, invasion, and metastasis; hence, MMPs are an important research focus. Compared to sporadic CRC, less is known regarding the molecular mechanisms and the role of MMPs in the development and progression of colitis-associated cancer (CAC) - CRC on a background of chronic inflammatory bowel disease (IBD) - primarily ulcerative colitis and Crohn's disease. Hence, the potential of MMPs as biomarkers and therapeutic targets for CAC is uncertain. Our goal was to review data regarding the role of MMPs in the development and progression of CAC. We sought to identify promising prognostic and therapeutic opportunities and novel lines of investigation. A key observation is that since MMPs may be more active in early phases of CAC, using MMPs as biomarkers of advancing neoplasia and as potential therapeutic targets for adjuvant therapy in those with advanced stage primary CAC rather than overt metastases may yield more favorable outcomes.
Collapse
Affiliation(s)
- Natalia Sampaio Moura
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alyssa Schledwitz
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Madeline Alizadeh
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Seema A. Patil
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
- Medical Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, Baltimore, MD, United States
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Harwood SL, Enghild JJ. Engineering New Protease Inhibitors Using α 2-Macroglobulin. Methods Mol Biol 2024; 2747:279-294. [PMID: 38038947 DOI: 10.1007/978-1-0716-3589-6_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Protease inhibitors of the alpha-macroglobulin family (αM) have a unique mechanism that allows them to trap proteases that is dependent not on the protease's class, but rather on its cleavage specificity. Proteases trigger a conformational change in the αM protein by cleaving within a "bait region," resulting in the sequestering of the protease inside the αM molecule. This nonspecific inhibitory mechanism appears to have arisen early in the αM family, and the broad protease-trapping capacity that it allows may play a role in pathogen defense.Human α2-macroglobulin (A2M) is a tetrameric αM whose bait region is permissive to cleavage by most proteases, making it a broad-spectrum protease inhibitor. Recent work has demonstrated that the inhibitory capacity of A2M derives directly from its bait region sequence: modifying the bait region sequence to introduce or remove protease cleavage sites will modify A2M's inhibition of the relevant proteases accordingly. Thus, changing the amino acid sequence of the bait region presents an effective avenue for protein engineering of new protease inhibitors if the substrate specificity of the target protease is known. The design of new A2M-based protease inhibitors with tailored inhibitory capacities has potential applications in basic research and the clinic. In this chapter, we describe the general approach and considerations for the bait region engineering of A2M.
Collapse
Affiliation(s)
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
10
|
Balakina A, Gadomsky S, Kokovina T, Sashenkova T, Mishchenko D, Terentiev A. New Derivatives of N-Hydroxybutanamide: Preparation, MMP Inhibition, Cytotoxicity, and Antitumor Activity. Int J Mol Sci 2023; 24:16360. [PMID: 38003553 PMCID: PMC10671431 DOI: 10.3390/ijms242216360] [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: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Using a novel method of N-substituted succinimide ring opening, new N-hydroxybutanamide derivatives were synthesized. These compounds were evaluated for their ability to inhibit matrix metalloproteinases (MMPs) and their cytotoxicity. The iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide showed the inhibition of MMP-2, MMP-9, and MMP-14 with an IC50 of 1-1.5 μM. All the compounds exhibited low toxicity towards carcinoma cell lines HeLa and HepG2. The iodoaniline derivative was also slightly toxic to glioma cell lines A-172 and U-251 MG. Non-cancerous FetMSC and Vero cells were found to be the least sensitive to all the compounds. In vivo studies demonstrated that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide had low acute toxicity. In a mouse model of B16 melanoma, this compound showed both antitumor and antimetastatic effects, with a 61.5% inhibition of tumor growth and an 88.6% inhibition of metastasis. Our findings suggest that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide has potential as a lead structure for the development of new MMP inhibitors. Our new synthetic approach can be a cost-effective method for the synthesis of inhibitors of metalloenzymes with promising antitumor potential.
Collapse
Affiliation(s)
- Anastasia Balakina
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Svyatoslav Gadomsky
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Tatyana Kokovina
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
| | - Tatyana Sashenkova
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Denis Mishchenko
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
- Scientific and Educational Center in Chernogolovka, State University of Education, 141014 Mytishchi, Russia
| | - Alexei Terentiev
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
- Scientific and Educational Center in Chernogolovka, State University of Education, 141014 Mytishchi, Russia
| |
Collapse
|
11
|
Istyastono EP, Yuniarti N, Prasasty VD, Mungkasi S, Waskitha SSW, Yanuar MRS, Riswanto FDO. Caffeic Acid in Spent Coffee Grounds as a Dual Inhibitor for MMP-9 and DPP-4 Enzymes. Molecules 2023; 28:7182. [PMID: 37894660 PMCID: PMC10609219 DOI: 10.3390/molecules28207182] [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: 09/05/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Type 2 diabetes mellitus and diabetic foot ulcers remain serious worldwide health problems. Caffeic acid is one of the natural products that has been experimentally proven to have diverse pharmacological properties. This study aimed to assess the inhibitory activity of caffeic acid and ethanolic extract of spent coffee grounds targeting DPP-4 and MMP-9 enzymes and evaluate the molecular interactions through 50-ns molecular dynamics simulations. This study also introduced our new version of PyPLIF HIPPOS, PyPLIF HIPPOS 0.2.0, which allowed us to identify protein-ligand interaction fingerprints and interaction hotspots resulting from molecular dynamics simulations. Our findings revealed that caffeic acid inhibited the DPP-4 and MMP-9 activity with an IC50 of 158.19 ± 11.30 µM and 88.99 ± 3.35 µM while ethanolic extract of spent coffee grounds exhibited an IC50 of 227.87 ± 23.80 µg/100 µL and 81.24 ± 6.46 µg/100 µL, respectively. Molecular dynamics simulations showed that caffeic acid interacted in the plausible allosteric sites of DPP-4 and in the active site of MMP-9. PyPLIF HIPPOS 0.2.0 identified amino acid residues interacting more than 10% throughout the simulation, which were Lys463 and Trp62 in the plausible allosteric site of DPP-4 and His226 in the active site of MMP-9.
Collapse
Affiliation(s)
- Enade P. Istyastono
- Research Group of Computer-Aided Drug Design and Discovery of Bioactive Natural Products, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta 55282, Indonesia; (S.S.W.W.); (M.R.S.Y.); (F.D.O.R.)
| | - Nunung Yuniarti
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | - Vivitri D. Prasasty
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Sudi Mungkasi
- Department of Mathematics, Faculty of Science and Technology, Sanata Dharma University, Yogyakarta 55282, Indonesia;
| | - Stephanus S. W. Waskitha
- Research Group of Computer-Aided Drug Design and Discovery of Bioactive Natural Products, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta 55282, Indonesia; (S.S.W.W.); (M.R.S.Y.); (F.D.O.R.)
| | - Michael R. S. Yanuar
- Research Group of Computer-Aided Drug Design and Discovery of Bioactive Natural Products, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta 55282, Indonesia; (S.S.W.W.); (M.R.S.Y.); (F.D.O.R.)
| | - Florentinus D. O. Riswanto
- Research Group of Computer-Aided Drug Design and Discovery of Bioactive Natural Products, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta 55282, Indonesia; (S.S.W.W.); (M.R.S.Y.); (F.D.O.R.)
| |
Collapse
|
12
|
Jaguri A, Ahmad A. Breaching the Fortress of Tumor Microenvironment to Control Cancer Metastasis. Cancers (Basel) 2023; 15:4562. [PMID: 37760530 PMCID: PMC10526375 DOI: 10.3390/cancers15184562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
As the primary cause of death for >90% of cancers, metastasis is the fourth and final stage of cancer during which cells gain the ability to leave their primary site, invade surrounding tissues, and disseminate to distant organs [...].
Collapse
Affiliation(s)
- Aayami Jaguri
- Weill Cornell Medicine-Qatar, Doha 24811, Qatar;
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Bioengineering, Integral University, Lucknow 226026, UP, India
| |
Collapse
|
13
|
Shaik BB, Katari NK, Jonnalagadda SB. Internal stimuli-responsive nanocarriers for controlled anti-cancer drug release: a review. Ther Deliv 2023; 14:595-613. [PMID: 37877308 DOI: 10.4155/tde-2023-0041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023] Open
Abstract
Cancer disease is one of the most frequent life-threatening, with a high fatality rate worldwide. However, recent immunotherapy studies in various tumours have yielded unsatisfactory outcomes, with just a few individuals experiencing long-term responses. To overcome these issues, nowadays internal stimuli-responsive nanocarriers have been widely exploited to transport a wide range of active substances, including peptides, genes and medicines. These nanosystems could be chemically adjusted to produce target-based drug release at the target location, minimizing pathological and physiological difficulties while increasing therapeutic efficiency. This review highlights the various types of internal stimuli-responsive nanocarriers and applications in cancer diagnosis. This study can provide inspiration and impetus for exploiting more promising internal stimuli-responsive nanosystems for drug delivery.
Collapse
Affiliation(s)
- Baji Baba Shaik
- Department of Chemistry, School of Science, GITAM (Deemed to be) University, Hyderabad, Telangana, 502329, India
- School of Chemistry & Physics, Westville Campus, University of KwaZulu-Natal, P Bag X 54001, Durban, 4000, Kwa-Zulu Natal, South Africa
| | - Naresh Kumar Katari
- Department of Chemistry, School of Science, GITAM (Deemed to be) University, Hyderabad, Telangana, 502329, India
- School of Chemistry & Physics, Westville Campus, University of KwaZulu-Natal, P Bag X 54001, Durban, 4000, Kwa-Zulu Natal, South Africa
| | - Sreekanth B Jonnalagadda
- School of Chemistry & Physics, Westville Campus, University of KwaZulu-Natal, P Bag X 54001, Durban, 4000, Kwa-Zulu Natal, South Africa
| |
Collapse
|
14
|
Hu J, Liu F, Chen Y, Fu J, Ju H. Signal-On Mass Spectrometric Biosensing of Multiplex Matrix Metalloproteinases with a Phospholipid-Structured Mass-Encoded Microplate. Anal Chem 2023. [PMID: 37235973 DOI: 10.1021/acs.analchem.3c01039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The detection of matrix metalloproteinases (MMPs) is of great importance for diagnosis and staging of cancer. This work proposed a signal-on mass spectrometric biosensing strategy with a phospholipid-structured mass-encoded microplate for assessment of multiplex MMP activities. The designed substrate and internal standard peptides were subsequently labeled with the reagents of isobaric tags for relative and absolute quantification (iTRAQ), and DSPE-PEG(2000)maleimide was embedded on the surface of a 96-well glass bottom plate to fabricate the phospholipid-structured mass-encoded microplate, which offered a simulated environment of the extracellular space for enzyme reactions between MMPs and the substrates. The strategy achieved multiplex MMP activity assays by dropping the sample in the well for enzyme cleavages, followed by adding trypsin to release the coding regions for ultrahigh performance liquid chromatography-tandem mass spectrometric (UHPLC-MS/MS) analysis. The peak area ratios of released coding regions and their respective internal standard (IS) peptides exhibited satisfied linear ranges of 0.05-50, 0.1-250, and 0.1-100 ng mL-1 with the detection limits of 0.017, 0.046, and 0.032 ng mL-1 for MMP-2, MMP-7, and MMP-3, respectively. The proposed strategy demonstrated good practicability in inhibition analysis and detections of multiplex MMP activities in serum samples. It is of great potential for clinical applications and can be expanded for multiplex enzyme assays.
Collapse
Affiliation(s)
- Junjie Hu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- College of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining 272067, China
| | - Fei Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jia Fu
- College of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining 272067, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
15
|
Tang TT, Jiang L, Zhong Q, Ni ZJ, Thakur K, Khan MR, Wei ZJ. Saikosaponin D exerts cytotoxicity on human endometrial cancer ishikawa cells by inducing apoptosis and inhibiting metastasis through MAPK pathways. Food Chem Toxicol 2023; 177:113815. [PMID: 37209937 DOI: 10.1016/j.fct.2023.113815] [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: 04/11/2023] [Revised: 05/01/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Saikosaponin D (SSD) is one of plant secondary metabolic active substance with effective anti-tumor ability; however, the toxicity of Saikosaponin D on human endometrial cancer Ishikawa cells is still unclear. Our results revealed that SSD displayed cytotoxicity on the Ishikawa cell with an IC50 = 15.69 μM, but was non-toxic to the human normal cell line HEK293. SSD could upregulate p21 and Cyclin B to keep cells in the G2/M stage. In addition, it activated the death receptor and mitochondrion routes to induce apoptosis in Ishikawa cells. The transwell chamber and wound healing results showed that SSD inhibited the cell migration and invasion. In addition, we found that it was closely related to the MAPK cascade pathway, and it could mediate the three classical MAPK pathways to block cell metastasis. In conclusion, SSD could be potentially beneficial as a natural secondary metabolite in preventing and treating endometrial carcinoma.
Collapse
Affiliation(s)
- Tong-Tong Tang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China.
| | - Li Jiang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China.
| | - Qian Zhong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| | - Zhi-Jing Ni
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| | - Kiran Thakur
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| | - Mohammad Rizwan Khan
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Zhao-Jun Wei
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, People's Republic of China.
| |
Collapse
|
16
|
Kubik A, das Virgens IPA, Szabó A, Váradi M, Csizmarik A, Keszthelyi A, Majoros A, Fehérvári P, Hegyi P, Ács N, Nyirády P, Szarvas T. Comprehensive Analysis of the Prognostic Value of Circulating MMP-7 Levels in Urothelial Carcinoma: A Combined Cohort Analysis, Systematic Review, and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24097859. [PMID: 37175566 PMCID: PMC10178327 DOI: 10.3390/ijms24097859] [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: 03/03/2023] [Revised: 03/29/2023] [Accepted: 04/08/2023] [Indexed: 05/15/2023] Open
Abstract
Lymph node (LN) status is the most significant prognostic factor for invasive urothelial bladder cancer (UBC); however, the optimal extent of LN dissection (LND) is debated. We assessed circulating matrix metalloproteinase-7 (MMP-7) as a prognostic factor and decision-making marker for the extent of LND. Preoperative serum MMP-7 levels were determined in two independent UBC cohorts (n = 188; n = 68) and in one control cohort (n = 97) by using the ELISA method. A systematic review and meta-analysis on the prognostic role of circulating pretreatment MMP-7 levels were performed. Serum MMP-7 levels were higher in patients compared to controls (p < 0.001) with the highest levels in LN-positive cases. Half of LN-positive UBC patients had low MMP-7 levels, whereas the survival of LN-negative patients with high serum MMP-7 findings was poor. MMP-7 levels were independently associated with poor survival in both cohorts (p = 0.006, p < 0.001). Accordingly, our systematic review of six eligible publications revealed a 2.5-fold higher mortality risk in patients with high MMP-7 levels. In conclusion, preoperative MMP-7 level is a validated and independent prognostic factor in urothelial cancer. It cannot be used to decide between regional or extended LND but may be useful in identifying LN-negative high-risk patients with potentially undetected metastases.
Collapse
Affiliation(s)
- András Kubik
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
- Center for Translational Medicine, Semmelweis University, 1082 Budapest, Hungary
| | | | - Anett Szabó
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
- Center for Translational Medicine, Semmelweis University, 1082 Budapest, Hungary
| | - Melinda Váradi
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
| | - Anita Csizmarik
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
| | - Attila Keszthelyi
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
| | - Attila Majoros
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
| | - Péter Fehérvári
- Center for Translational Medicine, Semmelweis University, 1082 Budapest, Hungary
- Department of Biostatistics, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Péter Hegyi
- Center for Translational Medicine, Semmelweis University, 1082 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1083 Budapest, Hungary
| | - Nándor Ács
- Center for Translational Medicine, Semmelweis University, 1082 Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, 1088 Budapest, Hungary
| | - Péter Nyirády
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
| | - Tibor Szarvas
- Department of Urology, Semmelweis University, 1082 Budapest, Hungary
- Department of Urology, University of Duisburg-Essen and German Cancer Consortium (DKTK), 45147 Essen, Germany
| |
Collapse
|
17
|
Bahlmann LC, Xue C, Chin AA, Skirzynska A, Lu J, Thériault B, Uehling D, Yerofeyeva Y, Peters R, Liu K, Chen J, Martel AL, Yaffe M, Al-Awar R, Goswami RS, Ylanko J, Andrews DW, Kuruvilla J, Laister RC, Shoichet MS. Targeting tumour-associated macrophages in hodgkin lymphoma using engineered extracellular matrix-mimicking cryogels. Biomaterials 2023; 297:122121. [PMID: 37075613 DOI: 10.1016/j.biomaterials.2023.122121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023]
Abstract
Tumour-associated macrophages are linked with poor prognosis and resistance to therapy in Hodgkin lymphoma; however, there are no suitable preclinical models to identify macrophage-targeting therapeutics. We used primary human tumours to guide the development of a mimetic cryogel, wherein Hodgkin (but not Non-Hodgkin) lymphoma cells promoted primary human macrophage invasion. In an invasion inhibitor screen, we identified five drug hits that significantly reduced tumour-associated macrophage invasion: marimastat, batimastat, AS1517499, ruxolitinib, and PD-169316. Importantly, ruxolitinib has demonstrated recent success in Hodgkin lymphoma clinical trials. Both ruxolitinib and PD-169316 (a p38 mitogen-activated protein kinase (p38 MAPK) inhibitor) decreased the percent of M2-like macrophages; however, only PD-169316 enhanced the percentage of M1-like macrophages. We validated p38 MAPK as an anti-invasion drug target with five additional drugs using a high-content imaging platform. With our biomimetic cryogel, we modeled macrophage invasion in Hodgkin lymphoma and then used it for target discovery and drug screening, ultimately identifying potential future therapeutics.
Collapse
Affiliation(s)
- Laura C Bahlmann
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Chang Xue
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Allysia A Chin
- The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
| | - Arianna Skirzynska
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
| | - Joy Lu
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada
| | - Brigitte Thériault
- Drug Discovery Program, Ontario Institute of Cancer Research, 661 University Ave Suite 510, Toronto, Ontario, M5G 0A3, Canada
| | - David Uehling
- Drug Discovery Program, Ontario Institute of Cancer Research, 661 University Ave Suite 510, Toronto, Ontario, M5G 0A3, Canada
| | - Yulia Yerofeyeva
- Biomarker Imaging Research Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Rachel Peters
- Biomarker Imaging Research Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Kela Liu
- Biomarker Imaging Research Laboratory, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Jianan Chen
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada
| | - Anne L Martel
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada; Physical Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Martin Yaffe
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada; Physical Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - Rima Al-Awar
- Drug Discovery Program, Ontario Institute of Cancer Research, 661 University Ave Suite 510, Toronto, Ontario, M5G 0A3, Canada; Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Rashmi S Goswami
- Biological Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada; Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Jarkko Ylanko
- Biological Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada
| | - David W Andrews
- Department of Medical Biophysics, University of Toronto, 101 College St Suite 15-701, Toronto, Ontario, M5G 1L7, Canada; Biological Sciences, Odette Cancer Research Program, Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada; Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - John Kuruvilla
- Princess Margaret Cancer Centre, University Health Network, 610 University Ave, Toronto, Ontario, M5G 2C1, Canada
| | - Rob C Laister
- Princess Margaret Cancer Centre, University Health Network, 610 University Ave, Toronto, Ontario, M5G 2C1, Canada.
| | - Molly S Shoichet
- Institute of Biomedical Engineering, 164 College Street, Toronto, Ontario, M5S 3G9, Canada; The Donnelly Centre, University of Toronto, Toronto, 160 College St, Ontario, M5S 3E1, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada.
| |
Collapse
|
18
|
Bharadwaj S, Sahoo AK, Yadava U. Editorial: Advances in the therapeutic targeting of human matrix metalloproteinases in health and disease. Front Mol Biosci 2023; 10:1150474. [PMID: 36968281 PMCID: PMC10036910 DOI: 10.3389/fmolb.2023.1150474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Affiliation(s)
- Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, v.v.i., BIOCEV Research Center, Vestec, Czech Republic
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, Uttar Pradesh, India
- *Correspondence: Umesh Yadava, ; Amaresh Kumar Sahoo,
| | - Umesh Yadava
- Department of Physics, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur, India
- *Correspondence: Umesh Yadava, ; Amaresh Kumar Sahoo,
| |
Collapse
|
19
|
Chang M. Matrix metalloproteinase profiling and their roles in disease. RSC Adv 2023; 13:6304-6316. [PMID: 36825288 PMCID: PMC9942564 DOI: 10.1039/d2ra07005g] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Matrix metalloproteinases (MMPs) play roles in remodelling of the extracellular matrix that occurs during morphogenesis, repair, and angiogenesis. Dysregulation of extracellular matrix remodelling can lead to cell proliferation, invasion, and tissue fibrosis. Identification of a specific MMP(s) in a disease has been challenging due to the presence of 24 closely-related human MMPs, each existing in three forms, of which only one is active and capable of catalysis. This review focuses on methods for MMP profiling, with particular emphasis on the batimastat affinity resin that binds only to the active forms of MMPs and related ADAMs (a disintegrin and metalloproteinases), which are then identified by mass spectrometry. Use of the batimastat affinity resin has identified targets for intervention in several human diseases.
Collapse
Affiliation(s)
- Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame Notre Dame IN 46556 USA
| |
Collapse
|
20
|
Marcianò G, Vocca C, Rania V, Citraro R, De Sarro G, Gallelli L. Metalloproteases in Pain Generation and Persistence: A Possible Target? Biomolecules 2023; 13:biom13020268. [PMID: 36830637 PMCID: PMC9953417 DOI: 10.3390/biom13020268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/15/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a large family of zinc-dependent proteolytic enzymes associated with extracellular matrix protein turnover and tissue degradation. They participate to many different physiological reactions but are also hyperactivated in several diseases. Various literature studies have documented that MMPs play a role in the modulation of neuropathic and nociceptive pain. The heterogeneity of clinical and pre-clinical data is an important issue in this experimental context. Despite the presence of a good number of studies on MMP inhibitors, these drugs showed scarce efficacy and relevant side effects. In the present manuscript, we reviewed studies in the literature that define a possible role of MMPs in pain and the effects of their modulation.
Collapse
Affiliation(s)
- Gianmarco Marcianò
- Operative Unit of Pharmacology and Pharmacovigilance, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
| | - Cristina Vocca
- Operative Unit of Pharmacology and Pharmacovigilance, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
| | - Vincenzo Rania
- Operative Unit of Pharmacology and Pharmacovigilance, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
| | - Rita Citraro
- Operative Unit of Pharmacology and Pharmacovigilance, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University Magna Graecia, 88100 Catanzaro, Italy
| | - Giovambattista De Sarro
- Operative Unit of Pharmacology and Pharmacovigilance, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University Magna Graecia, 88100 Catanzaro, Italy
| | - Luca Gallelli
- Operative Unit of Pharmacology and Pharmacovigilance, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
- Research Center FAS@UMG, Department of Health Science, University Magna Graecia, 88100 Catanzaro, Italy
- Medifarmagen SRL, Department of Health Science, “Mater Domini” University Hospital, 88100 Catanzaro, Italy
- Correspondence: ; Tel.: +39-0961-712322
| |
Collapse
|
21
|
Regulation of MMP9 transcription by ETS1 in immortalized salivary gland epithelial cells of patients with salivary hypofunction and primary Sjögren's syndrome. Sci Rep 2022; 12:14552. [PMID: 36008454 PMCID: PMC9411565 DOI: 10.1038/s41598-022-18576-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/16/2022] [Indexed: 12/21/2022] Open
Abstract
Primary Sjögren’s syndrome (pSS) patients exhibit enhanced degradation of the salivary epithelium initially through MMP9 overexpression. We assessed the expression of MMP9 and an associated transcription factor, ETS1, in primary salivary gland epithelial cells (SGECs) and investigated potential regulatory mechanism(s) in immortalized SGECs. SGECs and iSGECs were derived from pSS and/or xerostomic “sicca” patients. siRNA knockdown of ETS1 in iSGECs was performed to determine MMP9 mRNA (qRT-PCR) and protein expression (ELISA). ETS1 binding to MMP9 promoter was assessed by luciferase activity and binding confirmed by mutagenesis and ChIP. Effects of ETS1 overexpression on progenitor and Epithelial-Mesenchymal transition (EMT) associated markers were determined by Western blot. Expression of ETS1 and its phosphorylated form in iSGECs was determined by immunofluorescence microscopy. ETS1 and MMP9 were overexpressed in SGECs of pSS and non-pSS sicca patients with salivary gland lymphocytic infiltration compared to non-pSS sicca patients without infiltration. ETS1 siRNA knockdown reduced both MMP9 mRNA and protein levels. ETS1 overexpression affected the expression of EMT and progenitor cell markers. Lastly, ETS1 bound the MMP9 promoter within the DNA region of −296 bp to −339 bp. ETS1 may impair salivary function through direct transcriptional control of the MMP9 promoter. ETS1 upregulation may also affect other factors involved in repair of the dysfunctional pSS salivary epithelium.
Collapse
|
22
|
Baidya SK, Banerjee S, Adhikari N, Jha T. Selective Inhibitors of Medium-Size S1' Pocket Matrix Metalloproteinases: A Stepping Stone of Future Drug Discovery. J Med Chem 2022; 65:10709-10754. [PMID: 35969157 DOI: 10.1021/acs.jmedchem.1c01855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Among various matrix metalloproteinases (MMPs), MMPs having medium-size S1' pockets are established as promising biomolecular targets for executing crucial roles in cancer, cardiovascular diseases, and neurodegenerative diseases. However, no such MMP inhibitors (MMPIs) are available to date as drug candidates despite a lot of continuous research work for more than three decades. Due to a high degree of structural resemblance among these MMPs, designing selective MMPIs is quite challenging. However, the variability and uniqueness of the S1' pockets of these MMPs make them promising targets for designing selective MMPIs. In this perspective, the overall structural aspects of medium-size S1' pocket MMPs including the unique binding patterns of enzyme-inhibitor interactions have been discussed in detail to acquire knowledge regarding selective inhibitor designing. This overall knowledge will surely be a curtain raiser for the designing of selective MMPIs as drug candidates in the future.
Collapse
Affiliation(s)
- Sandip Kumar Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
23
|
Chaves Filho AJM, Mottin M, Lós DB, Andrade CH, Macedo DS. The tetrapartite synapse in neuropsychiatric disorders: Matrix metalloproteinases (MMPs) as promising targets for treatment and rational drug design. Biochimie 2022; 201:79-99. [PMID: 35931337 DOI: 10.1016/j.biochi.2022.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/26/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Inflammation and an exacerbated immune response are widely accepted contributing mechanisms to the genesis and progression of major neuropsychiatric disorders. However, despite the impressive advances in understanding the neurobiology of these disorders, there is still no approved drug directly linked to the regulation of inflammation or brain immune responses. Importantly, matrix metalloproteinases (MMPs) comprise a group of structurally related endopeptidases primarily involved in remodeling extracellular matrix (ECM). In the central nervous system (CNS), these proteases control synaptic plasticity and strength, patency of the blood-brain barrier, and glia-neuron interactions through cleaved and non-cleaved mediators. Several pieces of evidence have pointed to a complex scenario of MMPs dysregulation triggered by neuroinflammation. Furthermore, major psychiatric disorders' affective symptoms and neurocognitive abnormalities are related to MMPs-mediated ECM changes and neuroglia activation. In the past decade, research efforts have been directed to broad-spectrum MMPs inhibitors with frustrating clinical results. However, in the light of recent advances in combinatorial chemistry and drug design technologies, specific and CNS-oriented MMPs modulators have been proposed as a new frontier of therapy for regulating ECM properties in the CNS. Therefore, here we aim to discuss the state of the art of MMPs and ECM abnormalities in major neuropsychiatric disorders, namely depression, bipolar disorder, and schizophrenia, the possible neuro-immune interactions involved in this complex scenario of MMPs dysregulation and propose these endopeptidases as promising targets for rational drug design.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | - Melina Mottin
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Deniele Bezerra Lós
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
24
|
Tassone P, Caruso C, White M, Tavares Dos Santos H, Galloway T, Dooley L, Zitsch R, Lester Layfield J, Baker O. The role of matrixmetalloproteinase-2 expression by fibroblasts in perineural invasion by oral cavity squamous cell carcinoma. Oral Oncol 2022; 132:106002. [PMID: 35779484 DOI: 10.1016/j.oraloncology.2022.106002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVES The mechanisms of perineural invasion (PNI) in oral cavity squamous cell carcinoma are only partially understood, and no studies have specifically investigated the role of perineural fibroblasts in PNI. Here, we identified fibroblasts within the microenvironment of perineural invasion and assessed their expression of matrix metalloproteinase-2 (MMP-2). MATERIALS AND METHODS Tumor specimens from 12 patients with oral cavity squamous cell carcinoma and pathologically-confirmed perineural invasion were stained by immunohistochemistry (IHC) for vimentin (positive control) and MMP-2. Scoring was quantified and compared at nerves involved with PNI and nerves uninvolved with PNI. RESULTS All 12 patients had perineural fibroblasts around involved and uninvolved nerves as marked by vimentin IHC staining. Perineural fibroblasts had detectable MMP-2 expression at areas of perineural invasion in all 12 patients, but no patients had MMP-2 expression by fibroblasts at nerves without PNI. CONCLUSION MMP-2 is expressed by fibroblasts within the microenvironment of perineural invasion, and MMP-2 expression by fibroblasts is a possible mechanism of perineural invasion by oral cavity squamous cell carcinoma. MMP-2 may be an anti-cancer target among oral cavity squamous cell carcinoma patients with PNI.
Collapse
Affiliation(s)
- Patrick Tassone
- Department of Otolaryngology - Head & Neck Surgery, University of Missouri, Columbia, MO, United States.
| | - Carla Caruso
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, United States
| | - Meghan White
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, United States
| | - Harim Tavares Dos Santos
- Department of Otolaryngology - Head & Neck Surgery, University of Missouri, Columbia, MO, United States; Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Tabitha Galloway
- Department of Otolaryngology - Head & Neck Surgery, University of Missouri, Columbia, MO, United States
| | - Laura Dooley
- Department of Otolaryngology - Head & Neck Surgery, University of Missouri, Columbia, MO, United States
| | - Robert Zitsch
- Department of Otolaryngology - Head & Neck Surgery, University of Missouri, Columbia, MO, United States
| | - J Lester Layfield
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, United States
| | - Olga Baker
- Department of Otolaryngology - Head & Neck Surgery, University of Missouri, Columbia, MO, United States; Department of Biochemistry, University of Missouri, Columbia, MO, United States; Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
25
|
Hu J, Liu F, Chen Y, Fu J, Shangguan G, Ju H. Mass-Encoded Suspension Array for Multiplex Detection of Matrix Metalloproteinase Activities. Anal Chem 2022; 94:6380-6386. [PMID: 35412800 DOI: 10.1021/acs.analchem.2c00854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This work designed a mass spectrometric biosensing strategy for the multiplex detection of matrix metalloproteinases (MMPs) with a mass-encoded suspension array. This array was fabricated as multiplex sensing probes by functionalizing magnetic beads with MMP-specific peptide-isobaric tags for relative and absolute quantification (iTRAQ) conjugates, which contained a hexahistidine tag for surface binding, a substrate region for MMP cleavage, and a coding region for the specific MMP. The integration of the multiplex coding ability of iTRAQ with ultrahigh performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) and the proteolysis method for peptide digestion endowed the biosensing method with high throughput and ultrahigh sensitivity. This strategy could be conveniently performed by mixing the sample and the suspension array for enzymatic reactions and then digesting the uncleaved peptides with trypsin to release the coding regions for UPLC-MS/MS analysis. With MMP-2 and MMP-7 as analytes, the relative changes of peak area ratios of coding regions showed good linear responses in the ranges of 0.2-100 and 0.5-400 ng mL-1, with detection limits of 0.064 and 0.17 ng mL-1, respectively. The analysis of MMP activity in serum samples and its change responding to inhibitors demonstrated the specificity, practicability, and expansibility of the proposed strategy. This work paves a new avenue for the activity assays of multiplex enzymes and promotes the development of mass spectrometric biosensing.
Collapse
Affiliation(s)
- Junjie Hu
- College of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining 272067, China.,State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Fei Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jia Fu
- College of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining 272067, China
| | - Guoqiang Shangguan
- College of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining 272067, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
26
|
Augoff K, Hryniewicz-Jankowska A, Tabola R, Stach K. MMP9: A Tough Target for Targeted Therapy for Cancer. Cancers (Basel) 2022; 14:cancers14071847. [PMID: 35406619 PMCID: PMC8998077 DOI: 10.3390/cancers14071847] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Having the capability to proteolyze diverse structural and signaling proteins, matrix metalloproteinase 9 (MMP9), one of the best-studied secretory endopeptidases, has been identified as a crucial mediator of processes closely associated with tumorigenesis, such as the extracellular matrix reorganization, epithelial to mesenchymal transition, cell migration, new blood vessel formation, and immune response. In this review, we present the current state of knowledge on MMP9 and its role in cancer growth in the context of cell adhesion/migration, cancer-related inflammation, and tumor microenvironment formation. We also summarize recent achievements in the development of selective MMP9 inhibitors and the limitations of using them as anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Augoff
- Department of Surgical Education, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Correspondence:
| | | | - Renata Tabola
- Department of Thoracic Surgery, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Kamilla Stach
- Department of Chemistry and Immunochemistry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
27
|
Chen S, Ning B, Song J, Yang Z, Zhou L, Chen Z, Mao L, Liu H, Wang Q, He S, Zhou Z. Enhanced pentose phosphate pathway activity promotes pancreatic ductal adenocarcinoma progression via activating YAP/MMP1 axis under chronic acidosis. Int J Biol Sci 2022; 18:2304-2316. [PMID: 35414794 PMCID: PMC8990471 DOI: 10.7150/ijbs.69526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Acidic microenvironment is a common physiological phenomenon in tumors, and is closely related to cancer development, but the effects of acidosis on pancreatic adenocarcinoma (PDAC) remains to be elucidated. Methods: Metabonomic assay and transcriptomic microarray were used to detect the changes of metabolites and gene expression profile respectively in acidosis-adapted PDAC cells. Wound healing, transwell and in vivo assay were applied to evaluate cell migration and invasion capacity. CCK8 and colony formation assays were performed to determine cell proliferation. Results: The acidosis-adapted PDAC cells had stronger metastasis and proliferation ability compared with the control cells. Metabonomic analysis showed that acidosis-adapted PDAC cells had both increased glucose and decreased glycolysis, implying a shift to pentose phosphate pathway. The metabolic shift further led to the inactivation of AMPK by elevating ATP. Transcriptomic analysis revealed that the differentially expressed genes in acidosis-adapted cells were enriched in extracellular matrix modification and Hippo signaling. Besides, MMP1 was the most upregulated gene in acidosis-adapted cells, mediated by the YAP/TAZ pathway, but could be reduced by AMPK activator. Conclusion: The present study showed that metabolic reprogramming promotes proliferation and metastasis of acidosis-adapted PDAC cells by inhibiting AMPK/Hippo signaling, thus upregulating MMP1.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Bo Ning
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Jinwen Song
- Department of Infectious Diseases, the Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Zihan Yang
- Department of Biomedical Science, City University of Hong Kong, Hong Kong SAR, China
| | - Li Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Zhiji Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Linhong Mao
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Hongtao Liu
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Qingliang Wang
- Department of Pathology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Song He
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| | - Zhihang Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, China
| |
Collapse
|
28
|
Bienstein M, Minond D, Schwaneberg U, Davari MD, Yildiz D. In Silico and Experimental ADAM17 Kinetic Modeling as Basis for Future Screening System for Modulators. Int J Mol Sci 2022; 23:1368. [PMID: 35163294 PMCID: PMC8835787 DOI: 10.3390/ijms23031368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/23/2022] [Indexed: 11/21/2022] Open
Abstract
Understanding the mechanisms of modulators' action on enzymes is crucial for optimizing and designing pharmaceutical substances. The acute inflammatory response, in particular, is regulated mainly by a disintegrin and metalloproteinase (ADAM) 17. ADAM17 processes several disease mediators such as TNFα and APP, releasing their soluble ectodomains (shedding). A malfunction of this process leads to a disturbed inflammatory response. Chemical protease inhibitors such as TAPI-1 were used in the past to inhibit ADAM17 proteolytic activity. However, due to ADAM17's broad expression and activity profile, the development of active-site-directed ADAM17 inhibitor was discontinued. New 'exosite' (secondary substrate binding site) inhibitors with substrate selectivity raised the hope of a substrate-selective modulation as a promising approach for inflammatory disease therapy. This work aimed to develop a high-throughput screen for potential ADAM17 modulators as therapeutic drugs. By combining experimental and in silico methods (structural modeling and docking), we modeled the kinetics of ADAM17 inhibitor. The results explain ADAM17 inhibition mechanisms and give a methodology for studying selective inhibition towards the design of pharmaceutical substances with higher selectivity.
Collapse
Affiliation(s)
- Marian Bienstein
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (M.B.); (U.S.)
| | - Dmitriy Minond
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
- Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
| | - Ulrich Schwaneberg
- Institute of Biotechnology, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (M.B.); (U.S.)
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, 52056 Aachen, Germany
| | - Mehdi D. Davari
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, 06120 Halle, Germany
| | - Daniela Yildiz
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Center for Human and Molecular Biology (ZHMB), University of Saarland, Kirrbergerstr., 66421 Homburg, Germany
| |
Collapse
|
29
|
Abdelrahman HA, Akawi N, Al-Shamsi AM, Ali A, Al-Jasmi F, John A, Hertecant J, Al-Gazali L, Ali BR. Bi-allelic null variant in matrix metalloproteinase-15, causes congenital cardiac defect, cholestasis jaundice, and failure to thrive. Clin Genet 2022; 101:403-410. [PMID: 34988996 DOI: 10.1111/cge.14107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 02/05/2023]
Abstract
Here, we delineate the phenotype of two siblings with a bi-allelic frameshift variant in MMP15 gene with congenital cardiac defects, cholestasis, and dysmorphism. Genome sequencing analysis revealed a recently reported homozygous frameshift variant (c.1058delC, p.Pro353Glnfs*102) in MMP15 gene that co-segregates with the phenotype in the family in a recessive mode of inheritance. Relative quantification of MMP15 mRNA showed evidence of degradation of the mutated transcript, presumably by nonsense mediated decay. Likewise, MMP15: p.Gly231Arg, a concurrently reported homozygous missense variant in another patient exhibiting a similar phenotype, was predicted to disrupt zinc ion binding to the MMP-15 enzyme catalytic domain, which is essential for substrate proteolysis, by structural modeling. Previous animal models and cellular findings suggested that MMP15 plays a crucial role in the formation of endocardial cushions. These findings confirm that MMP15 is an important gene in human development, particularly cardiac, and that its loss of function is likely to cause a severe disorder phenotype.
Collapse
Affiliation(s)
- Hanadi A Abdelrahman
- Department of Genetics and Genomics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Nadia Akawi
- Department of Genetics and Genomics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Aisha M Al-Shamsi
- Paediatrics Department, Tawam hospital, Al-Ain, United Arab Emirates
| | - Amanat Ali
- Department of Genetics and Genomics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Fatma Al-Jasmi
- Department of Genetics and Genomics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Department of Paediatrics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Anne John
- Department of Genetics and Genomics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Jozef Hertecant
- Paediatrics Department, Tawam hospital, Al-Ain, United Arab Emirates
| | - Lihadh Al-Gazali
- Department of Paediatrics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Heath Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Zayed Center for Health sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
30
|
Zaykov V, Chaqour B. The CCN2/CTGF interactome: an approach to understanding the versatility of CCN2/CTGF molecular activities. J Cell Commun Signal 2021; 15:567-580. [PMID: 34613590 DOI: 10.1007/s12079-021-00650-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023] Open
Abstract
Cellular communication network 2 (CCN2), also known as connective tissue growth factor (CTGF) regulates diverse cellular processes, some at odds with others, including adhesion, proliferation, apoptosis, and extracellular matrix (ECM) protein synthesis. Although a cause-and-effect relationship between CCN2/CTGF expression and local fibrotic reactions has initially been established, CCN2/CTGF manifests cell-, tissue-, and context-specific functions and differentially affects developmental and pathological processes ranging from progenitor cell fate decisions and angiogenesis to inflammation and tumorigenesis. CCN2/CTGF multimodular structure, binding to and activation or inhibition of multiple cell surface receptors, growth factors and ECM proteins, and susceptibility for proteolytic cleavage highlight the complexity to CCN2/CTGF biochemical attributes. CCN2/CTGF expression and dosage in the local environment affects a defined community of its interacting partners, and this results in sequestration of growth factors, interference with or potentiation of ligand-receptor binding, cellular internalization of CCN2/CTGF, inhibition or activation of proteases, and generation of CCN2/CTGF degradome products that add molecular diversity and expand the repertoire of functional modules in the cells and their microenvironment. Through these interactions, different intracellular signals and cellular responses are elicited culminating into physiological or pathological reactions. Thus, the CCN2/CTGF interactome is a defining factor of its tissue- and context-specific effects. Mapping of new CCN2/CTGF binding partners might shed light on yet unknown roles of CCN2/CTGF and provide a solid basis for tissue-specific targeting this molecule or its interacting partners in a therapeutic context.
Collapse
Affiliation(s)
- Viktor Zaykov
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Brahim Chaqour
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
31
|
Talib WH, Mahmod AI, Kamal A, Rashid HM, Alashqar AMD, Khater S, Jamal D, Waly M. Ketogenic Diet in Cancer Prevention and Therapy: Molecular Targets and Therapeutic Opportunities. Curr Issues Mol Biol 2021; 43:558-589. [PMID: 34287243 PMCID: PMC8928964 DOI: 10.3390/cimb43020042] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer is still one of the most significant global challenges facing public health, the world still lacks complementary approaches that would significantly enhance the efficacy of standard anticancer therapies. One of the essential strategies during cancer treatment is following a healthy diet program. The ketogenic diet (KD) has recently emerged as a metabolic therapy in cancer treatment, targeting cancer cell metabolism rather than a conventional dietary approach. The ketogenic diet (KD), a high-fat and very-low-carbohydrate with adequate amounts of protein, has shown antitumor effects by reducing energy supplies to cells. This low energy supply inhibits tumor growth, explaining the ketogenic diet's therapeutic mechanisms in cancer treatment. This review highlights the crucial mechanisms that explain the ketogenic diet's potential antitumor effects, which probably produces an unfavorable metabolic environment for cancer cells and can be used as a promising adjuvant in cancer therapy. Studies discussed in this review provide a solid background for researchers and physicians to design new combination therapies based on KD and conventional therapies.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Ayah Kamal
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Hasan M. Rashid
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Aya M. D. Alashqar
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Samar Khater
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Duaa Jamal
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931, Jordan; (A.I.M.); (A.K.); (H.M.R.); (A.M.D.A.); (S.K.); (D.J.)
| | - Mostafa Waly
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khoud 34-123, Oman;
| |
Collapse
|
32
|
Zak KM, Bostock MJ, Waligorska I, Thøgersen IB, Enghild JJ, Popowicz GM, Grudnik P, Potempa J, Ksiazek M. Latency, thermal stability, and identification of an inhibitory compound of mirolysin, a secretory protease of the human periodontopathogen Tannerella forsythia. J Enzyme Inhib Med Chem 2021; 36:1267-1281. [PMID: 34210221 PMCID: PMC8259862 DOI: 10.1080/14756366.2021.1937619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mirolysin is a secretory protease of Tannerella forsythia, a member of the dysbiotic oral microbiota responsible for periodontitis. In this study, we show that mirolysin latency is achieved by a “cysteine-switch” mechanism exerted by Cys23 in the N-terminal profragment. Mutation of Cys23 shortened the time needed for activation of the zymogen from several days to 5 min. The mutation also decreased the thermal stability and autoproteolysis resistance of promirolysin. Mature mirolysin is a thermophilic enzyme and shows optimal activity at 65 °C. Through NMR-based fragment screening, we identified a small molecule (compound (cpd) 9) that blocks promirolysin maturation and functions as a competitive inhibitor (Ki = 3.2 µM), binding to the S1′ subsite of the substrate-binding pocket. Cpd 9 shows superior specificity and does not interact with other T. forsythia proteases or Lys/Arg-specific proteases.
Collapse
Affiliation(s)
- Krzysztof M Zak
- Helmholtz Zentrum München, Institute of Structural Biology, Neuherberg, Germany.,Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mark J Bostock
- Helmholtz Zentrum München, Institute of Structural Biology, Neuherberg, Germany.,Biomolecular NMR and Center for Integrated Protein Science Munich at Department Chemie, Technical University of Munich, Garching, Germany
| | - Irena Waligorska
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Grzegorz M Popowicz
- Helmholtz Zentrum München, Institute of Structural Biology, Neuherberg, Germany.,Biomolecular NMR and Center for Integrated Protein Science Munich at Department Chemie, Technical University of Munich, Garching, Germany
| | - Przemyslaw Grudnik
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Krakow, Poland.,Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Miroslaw Ksiazek
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Krakow, Poland.,Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| |
Collapse
|
33
|
Harwood SL, Nielsen NS, Diep K, Jensen KT, Nielsen PK, Yamamoto K, Enghild JJ. Development of selective protease inhibitors via engineering of the bait region of human α 2-macroglobulin. J Biol Chem 2021; 297:100879. [PMID: 34139236 PMCID: PMC8267569 DOI: 10.1016/j.jbc.2021.100879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/02/2021] [Accepted: 06/13/2021] [Indexed: 01/15/2023] Open
Abstract
Human α2-macroglobulin (A2M) is an abundant protease inhibitor in plasma, which regulates many proteolytic processes and is involved in innate immunity. A2M’s unique protease-trapping mechanism of inhibition is initiated when a protease cleaves within the exposed and highly susceptible “bait region.” As the wild-type bait region is permissive to cleavage by most human proteases, A2M is accordingly a broad-spectrum protease inhibitor. In this study, we extensively modified the bait region in order to identify any potential functionally important elements in the bait region sequence and to engineer A2M proteins with restrictive bait regions, which more selectively inhibit a target protease. A2M in which the bait region was entirely replaced by glycine-serine repeats remained fully functional and was not cleaved by any tested protease. Therefore, this bait region was designated as the “tabula rasa” bait region and used as the starting point for further bait region engineering. Cleavage of the tabula rasa bait region by specific proteases was conveyed by the insertion of appropriate substrate sequences, e.g., basic residues for trypsin. Screening and optimization of tabula rasa bait regions incorporating matrix metalloprotease 2 (MMP2) substrate sequences produced an A2M that was specifically cleaved by MMPs and inhibited MMP2 cleavage activity as efficiently as wild-type A2M. We propose that this approach can be used to develop A2M-based protease inhibitors, which selectively inhibit target proteases, which might be applied toward the clinical inhibition of dysregulated proteolysis as occurs in arthritis and many types of cancer.
Collapse
Affiliation(s)
- Seandean Lykke Harwood
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark; Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Nadia Sukusu Nielsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Khang Diep
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | | | - Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
34
|
Pompili S, Latella G, Gaudio E, Sferra R, Vetuschi A. The Charming World of the Extracellular Matrix: A Dynamic and Protective Network of the Intestinal Wall. Front Med (Lausanne) 2021; 8:610189. [PMID: 33937276 PMCID: PMC8085262 DOI: 10.3389/fmed.2021.610189] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
The intestinal extracellular matrix (ECM) represents a complex network of proteins that not only forms a support structure for resident cells but also interacts closely with them by modulating their phenotypes and functions. More than 300 molecules have been identified, each of them with unique biochemical properties and exclusive biological functions. ECM components not only provide a scaffold for the tissue but also afford tensile strength and limit overstretch of the organ. The ECM holds water, ensures suitable hydration of the tissue, and participates in a selective barrier to the external environment. ECM-to-cells interaction is crucial for morphogenesis and cell differentiation, proliferation, and apoptosis. The ECM is a dynamic and multifunctional structure. The ECM is constantly renewed and remodeled by coordinated action among ECM-producing cells, degrading enzymes, and their specific inhibitors. During this process, several growth factors are released in the ECM, and they, in turn, modulate the deposition of new ECM. In this review, we describe the main components and functions of intestinal ECM and we discuss their role in maintaining the structure and function of the intestinal barrier. Achieving complete knowledge of the ECM world is an important goal to understand the mechanisms leading to the onset and the progression of several intestinal diseases related to alterations in ECM remodeling.
Collapse
Affiliation(s)
- Simona Pompili
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, Gastroenterology Unit, University of L'Aquila, L'Aquila, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberta Sferra
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonella Vetuschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
35
|
Polymerizable Matrix Metalloproteinases' Inhibitors with Potential Application for Dental Restorations. Biomedicines 2021; 9:biomedicines9040366. [PMID: 33807479 PMCID: PMC8065691 DOI: 10.3390/biomedicines9040366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
Collagen cleavage by matrix metalloproteinase (MMP) is considered a major cause of dental resins long term failure. Most MMP inhibitors display significant toxicity and are unsuitable for dental resins’ applications. Here we report a study of a new class of inhibitors that display the unique property of being co-polymerizable with other vinyl compounds present in commercial dental resins, limiting their release and potential toxicity. Computational affinity towards the active site of different MMP-1; -2; -8; -9 and -13 of several compounds showed interesting properties and were synthesized. These free compounds were tested concerning their toxicity upon contact with two different cell types, with no substantial decrease in cell viability at high concentrations. Even so, compound’s safety can be further improved upon copolymerization with commercial dental resins, limiting their release.
Collapse
|
36
|
Wojdas M, Dąbkowska K, Winsz-Szczotka K. Alterations of Extracellular Matrix Components in the Course of Juvenile Idiopathic Arthritis. Metabolites 2021; 11:132. [PMID: 33668781 PMCID: PMC7996267 DOI: 10.3390/metabo11030132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Juvenile idiopathic arthritis (JIA) is the most common group of chronic connective tissue diseases in children that is accompanied by joint structure and function disorders. Inflammation underlying the pathogenic changes in JIA, caused by hypersecretion of proinflammatory cytokines, leads to the destruction of articular cartilage. The degradation which progresses with the duration of JIA is not compensated by the extent of repair processes. These disorders are attributed in particular to changes in homeostasis of extracellular matrix (ECM) components, including proteoglycans, that forms articular cartilage. Changes in metabolism of matrix components, associated with the disturbance of their degradation and biosynthesis processes, are the basis of the progressive wear of joint structures observed in the course of JIA. Clinical evaluation and radiographic imaging are current methods to identify the destruction. The aim of this paper is to review enzymatic and non-enzymatic factors involved in catabolism of matrix components and molecules stimulating their biosynthesis. Therefore, we discuss the changes in these factors in body fluids of children with JIA and their potential diagnostic use in the assessment of disease activity. Understanding the changes in ECM components in the course of the child-hood arthritis may provide the introduction of both new diagnostic tools and new therapeutic strategies in children with JIA.
Collapse
Affiliation(s)
- Magdalena Wojdas
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, ul. Jedności 8, 41-200 Sosnowiec, Poland; (K.D.); (K.W.-S.)
| | | | | |
Collapse
|
37
|
Strategies to Target ADAM17 in Disease: From its Discovery to the iRhom Revolution. Molecules 2021; 26:molecules26040944. [PMID: 33579029 PMCID: PMC7916773 DOI: 10.3390/molecules26040944] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
For decades, disintegrin and metalloproteinase 17 (ADAM17) has been the object of deep investigation. Since its discovery as the tumor necrosis factor convertase, it has been considered a major drug target, especially in the context of inflammatory diseases and cancer. Nevertheless, the development of drugs targeting ADAM17 has been harder than expected. This has generally been due to its multifunctionality, with over 80 different transmembrane proteins other than tumor necrosis factor α (TNF) being released by ADAM17, and its structural similarity to other metalloproteinases. This review provides an overview of the different roles of ADAM17 in disease and the effects of its ablation in a number of in vivo models of pathological conditions. Furthermore, here, we comprehensively encompass the approaches that have been developed to accomplish ADAM17 selective inhibition, from the newest non-zinc-binding ADAM17 synthetic inhibitors to the exploitation of iRhom2 to specifically target ADAM17 in immune cells.
Collapse
|
38
|
Rudzińska M, Daglioglu C, Savvateeva LV, Kaci FN, Antoine R, Zamyatnin AA. Current Status and Perspectives of Protease Inhibitors and Their Combination with Nanosized Drug Delivery Systems for Targeted Cancer Therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:9-20. [PMID: 33442233 PMCID: PMC7797289 DOI: 10.2147/dddt.s285852] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022]
Abstract
In cancer treatments, many natural and synthetic products have been examined; among them, protease inhibitors are promising candidates for anti-cancer agents. Since dysregulated proteolytic activities can contribute to tumor development and metastasis, antagonization of proteases with tailored inhibitors is an encouraging approach. Although adverse effects of early designs of these inhibitors disappeared after the introduction of next-generation agents, most of the proposed inhibitors did not pass the early stages of clinical trials due to their nonspecific toxicity and lack of pharmacological effects. Therefore, new applications that modulate proteases more specifically and serve their programmed way of administration are highly appreciated. In this context, nanosized drug delivery systems have attracted much attention because preliminary studies have demonstrated that the therapeutic capacity of inhibitors has been improved significantly with encapsulated formulation as compared to their free forms. Here, we address this issue and discuss the current application and future clinical prospects of this potential combination towards targeted protease-based cancer therapy.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Cenk Daglioglu
- Biotechnology and Bioengineering Application and Research Center, Integrated Research Centers, Izmir Institute of Technology, Urla, Izmir 35430, Turkey
| | - Lyudmila V Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Fatma Necmiye Kaci
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Yakutiye, Erzurum 25050, Turkey
| | - Rodolphe Antoine
- CNRS, Institut Lumière Matière, Univ Lyon, Université Claude Bernard Lyon 1, Lyon F-69622, France
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia.,Department of Biotechnology, Sirius University of Science and Technology, Sochi 354340, Russia
| |
Collapse
|
39
|
Galo R, Marinho M, Silva Telles PD, Borsatto M. Shear bond strength of the adhesive/dentin interface after different etching protocols. J Conserv Dent 2021; 24:393-398. [PMID: 35282582 PMCID: PMC8896130 DOI: 10.4103/jcd.jcd_223_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/07/2021] [Accepted: 08/25/2021] [Indexed: 11/04/2022] Open
Abstract
Background: Aim: Materials and Methods: Results: Conclusion:
Collapse
|
40
|
Bi Q, Wang M, Zhao F, Wang M, Yin X, Ruan J, Wang D, Ji X. N-Butanol Fraction of Wenxia Formula Extract Inhibits the Growth and Invasion of Non-Small Cell Lung Cancer by Down-Regulating Sp1-Mediated MMP2 Expression. Front Pharmacol 2020; 11:594744. [PMID: 33329003 PMCID: PMC7734278 DOI: 10.3389/fphar.2020.594744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/21/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the most commonly diagnosed cancer and the leading cause of cancer death. It is necessary to develop effective anti-lung cancer therapeutics. Wenxia Formula (WXF), an empirical traditional Chinese herbal formula, has been reported to have significant antitumor activity. In this study, to further clarify the material basis of the anti-tumor effect of WXF, we investigated the cytotoxic effect of the N-butanol fraction of Wenxia Formula extract (NWXF) against two lung cancer and one normal human cell lines. The chemical profile of NWXF was characterized by UPLC/Q-TOF-MS analysis and a total of 201 compounds with mzCloud Best Match of greater than 70 were identified by using the online database mzCloud. To address the functional role of NWXF, we assessed cell proliferation, migration and invasion capabilities. Subcutaneous xenografts were constructed to determine the effect of NWXF in vivo. The results showed that NWXF effectively inhibited the proliferation and migration of non-small cell lung cancer (NSCLC) cells with little toxic effects on human bronchial epithelial cells. Meanwhile, orally administered NWXF exhibited prominent dose-dependent anti-tumor efficacy in vivo. Mechanistically, NWXF significantly downregulated MMP9 and Sp1-mediated MMP2 expression. In conclusion, NWXF might be a promising candidate for treatment of human lung cancer.
Collapse
Affiliation(s)
- QianYu Bi
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China.,College of Chinese Traditional Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - MengRan Wang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fang Zhao
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meng Wang
- College of Chinese Traditional Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Medicine, Jining NO.1 People's Hospital, Jining, China
| | - XiangJun Yin
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - JiaZhao Ruan
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - DeLong Wang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - XuMing Ji
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China.,Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China.,Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Hangzhou, China
| |
Collapse
|
41
|
Dual inhibitors of histone deacetylases and other cancer-related targets: A pharmacological perspective. Biochem Pharmacol 2020; 182:114224. [PMID: 32956642 DOI: 10.1016/j.bcp.2020.114224] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Epigenetic enzymes histone deacetylases (HDACs) are clinically validated anticancer drug targets which have been studied intensively in the past few decades. Although several drugs have been approved in this field, they are still limited to a subset of hematological malignancies (in particular T-cell lymphomas), with therapeutic potential not fully realized and the drug-resistance occurred after a certain period of use. To maximize the therapeutic potential of these classes of anticancer drugs, and to extend their application to solid tumors, numerous combination therapies containing an HDACi and an anticancer agent from other mechanisms are currently ongoing in clinical trials. Recently, dual targeting strategy comprising the HDACs component has emerged as an alternative approach for combination therapies. In this perspective, we intend to gather all HDACs-containing dual inhibitors related to cancer therapy published in literature since 2015, classify them into five categories based on targets' biological functions, and discuss the rationale why dual acting agents should work better than combinatorial therapies using two separate drugs. The article discusses the pharmacological aspects of these dual inhibitors, including in vitro biological activities, pharmacokinetic studies, in vivo efficacy studies, as well as available clinical trials. The review of the current status and advances should provide better analysis for future opportunities and challenges of this field.
Collapse
|
42
|
Serra R. Matrix Metalloproteinases in Health and Disease. Biomolecules 2020; 10:biom10081138. [PMID: 32752285 PMCID: PMC7465027 DOI: 10.3390/biom10081138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are members of an enzyme family and, under normal physiological conditions, are critical for maintaining tissue allostasis [...].
Collapse
Affiliation(s)
- Raffaele Serra
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, Catanzaro, 88100 Germaneto, Italy
| |
Collapse
|