1
|
Kim HD, Choi H, Park JY, Kim CH. Distinct structural basis and catalytic classification of matrix metalloproteinases and their endogenous tissue inhibitors with glycosylation issue in cellular and tissue regulation. Arch Biochem Biophys 2025; 769:110436. [PMID: 40280381 DOI: 10.1016/j.abb.2025.110436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Matrix metalloproteinase (MMP) enzymes cleave proteins on the extracellular matrix (ECM) region. MMPs are categorized as Zn2+-binding endo-proteinases. MMPs are stringently regulated in cancers, inflammatory cells and tissues. There are 29 types of MMPs as initially expressed in inactive zymogens (proMMPs) and activated by proteolysis in vertebrates including human. MMPs consist of three highly conserved parts of pro-MMP in precursor, catalytic and hemopexin domains. The MMPs are composed of systemic complexes with their endogenously expressed inhibitors of the tissue inhibitors of metalloproteinases (TIMPs). Therefore, TIMPs intrinsically control such activated MMPs, indicating the existence of self-modulation capacity. N-linked glycosylation (N-glycosylation) saves biological information than known phosphorylation, ubiquitination and acetylation. The MMPs are roughly present as membrane-merged and secreted glycoproteins. MMPs N-glycans regulate cellular behaviors, immune tolerance, and developing angiogenesis. Aberrant N-glycosylation of MMPs may cause the pathogenic properties. N-glycosylation shapes phenotypes of MMPs-producing cells during early MMPs involved in human. Additionally, issues of MMPs and TIMPs glycosylation have been described to view the importance of the glycans in their interaction with owns and other targets. Most of MMPs and 4 TIMPs are not well studied for their glycosylation and its functional roles.
Collapse
Affiliation(s)
- Hee-Do Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea
| | - Hyunju Choi
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea
| | - Jun-Young Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea; Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Suwon, Gyunggi-Do, 16419, Republic of Korea; Samsung Advanced Institute of Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
2
|
Mantry S, Das PK, Sankaraiah J, Panda S, Silakabattini K, Reddy Devireddy AK, Barik CS, Khalid M. Advancement on heparin-based hydrogel/scaffolds in biomedical and tissue engineering applications: Delivery carrier and pre-clinical implications. Int J Pharm 2025:125733. [PMID: 40398669 DOI: 10.1016/j.ijpharm.2025.125733] [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: 02/06/2025] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025]
Abstract
The advancement of biomaterials utilization in biomedical and tissue regenerative applications has emerged progressively. Hydrogels are three-dimensional, hydrophilic polymeric networks that replicate the natural extracellular matrix (ECM), establishing a hydrated porous milieu that emulates biological functions such as proliferation and differentiation of cellular components. The application of biological macromolecules, particularly Heparin-based hydrogel, has garnered considerable interest owing to various intrinsic biological and mechanical properties. This comprehensive review paper is designed to elucidate the derivation of heparin and its purification method for biomedical uses. The article briefly outlines the diverse physiochemical and biological properties of heparin derivative-based hydrogels/scaffolds and emphasizes their significance as vehicles for growth factors, genes, and cells in complex biomedical and tissue engineering applications. This publication also summarizes the potential concerns associated with heparin-based derivatives, efforts to address these issues, and current clinical perspectives. This represents the inaugural instance of an extensive summarization of heparin-based hydrogels in biomedical applications, emphasizing pre-clinical and clinical investigations, which will further assist the scientific community in addressing the challenges associated with heparin-based hydrogels in biomedical contexts.
Collapse
Affiliation(s)
- Shubhrajit Mantry
- Department of Pharmacy, Sarala Birla University, Birla Knowledge City, P.O.- Mahilong, Purulia Road, Ranchi 835103 Jharkhand, India.
| | - Prabhat Kumar Das
- Department of Pharmacology, GRY Institute of Pharmacy, Borawan, Khargone, MP, India
| | - Jonna Sankaraiah
- Department of Process Development, Medytox Inc., 102, Osongsaengmyeong 4-ro, Osong-eup, Heugdeok-gu, Cheongju-si, Chungcheongbuk-do, South Korea
| | - Satyajit Panda
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur, Cuttack, Odisha 754202, India.
| | - Kotaiah Silakabattini
- Department of Pharmacognosy, Chebrolu Hanumaiah Institute of Pharmaceutical Sciences, Chandramoulipuram, Chowdavaram, Guntur 522019 Andhra Pradesh, India
| | - Ashok Kumar Reddy Devireddy
- Department of Pharmacology, A M Reddy Memorial College of Pharmacy, Petlurivaripalem, Narasaraopet, Palnadu (Dt), A.P 522601, India
| | - Chandra Sekhar Barik
- Department of Pharmacology, School of Pharmacy, DRIEMS University, Kotasahi, Kairapari, Tangi, Cuttack, Odisha 754022, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| |
Collapse
|
3
|
Zhao L, Du GL, Ruze A, Qi HZ, Zhang CS, Li QL, Deng AX, Zhao BH, Hu S, Gai MT, Gao XM. Novel function of macrophage migration inhibitory factor in regulating post-infarct inflammation and the therapeutic significance. J Adv Res 2025:S2090-1232(25)00348-0. [PMID: 40383291 DOI: 10.1016/j.jare.2025.05.030] [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: 02/04/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
INTRODUCTION Recent studies indicate that macrophage migration inhibitory factor (MIF) has a dual role in myocardial infarction (MI), with different cellular sources of MIF influencing inflammation and healing differentially. OBJECTIVES To investigate the role and underlying mechanism of MIF in MI and interventional efficacy targeting MIF. METHODS Wild-type (WT), global MIF gene knockout (KO) and chimeric mice were subjected to coronary artery occlusion. The inflammatory responses and healing processes following MI were studied in both in vivo and in vitro settings. Furthermore, the therapeutic potential of pharmacological MIF inhibition to improve the prognosis of MI was explored. RESULTS Globally, MIF enhanced systemic and local inflammatory responses, as well as splenic monocyte mobilization, in mice with MI. MIF promoted monocyte migration through CCR2 and CXCR4 in peripheral blood mononuclear cells (PBMCs) and the infarcted myocardium. Additionally, MIF augmented angiotensin Ⅱ type 1 receptor (AT-1R) expression and interacted with AT-1R to promote the splenic monocyte mobilization following acute MI. MIF derived from bone marrow cells (KOWT mice) had stronger systemic and local inflammatory responses and augmented mobilization of splenic monocytes. In contrast, deficiency of MIF in leukocytes (WTKO mice) increased Ly-6Clow monocyte accumulation, M2 macrophage infiltration, and degree of myocardial fibrosis in infarcted myocardium. In vitro, MIF derived from ischemic heart enhanced M2 but impaired M1 macrophage marker expression in PBMCs. Anti-MIF treatment effectively attenuated splenic monocyte mobilization and both systemic and regional inflammatory responses post-MI without affecting the healing process, thereby improving the long-term prognosis. CONCLUSION Deletion of global and inflammatory-cell-derived MIF diminished inflammation following MI by inhibiting monocyte mobilization and downregulating pro-inflammatory mediators, while cardiac-derived MIF exerted anti-inflammatory influence and facilitated healing. Furthermore, MIF antibody therapy protected the heart from severe ischemic injury and improved long-term prognosis.
Collapse
Affiliation(s)
- Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China; Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Hong-Zhi Qi
- Department of Radiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chuan-Shan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Su Hu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Min-Tao Gai
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China.
| |
Collapse
|
4
|
Roa-Bruzón IY, Duany-Almira LF, Valle-Delgadillo YM, Flores-Salinas HE, Valdés-Alvarado E, Padilla-Gutiérrez JR. MMP3 as a Molecular Link: Unraveling the Connection Between Ankylosing Spondylitis and Acute Coronary Syndrome. Cells 2025; 14:597. [PMID: 40277922 PMCID: PMC12025634 DOI: 10.3390/cells14080597] [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: 02/17/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease that primarily affects the joints, limiting patients' mobility and quality of life. Recent studies have shown that patients with AS have a significantly higher risk of developing severe cardiovascular complications, such as acute coronary syndrome (ACS). A comprehensive review (2014-2024) included a study evaluating the significance of matrix metalloproteinase 3 (MMP-3) in cardiovascular risk among AS patients. The findings indicate that chronic inflammation in AS not only damages the joints but also contributes to the progression of cardiovascular diseases. At the molecular level, MMP-3 is instrumental in degrading the extracellular matrix, leading to instability in the atherosclerotic plaques and increasing the risk of ACS. Additionally, MMP-3 activation is related to the inflammatory pathways, such as tumor necrosis factor-alpha (TNF-α) and NF-κB, which amplify its effect on both joint destruction and vascular damage. This molecular approach offers new perspectives for understanding and treating AS and its cardiovascular complications, suggesting that MMP-3 inhibition could be a promising therapeutic strategy to mitigate cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Iliannis Y. Roa-Bruzón
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (I.Y.R.-B.); (L.F.D.-A.); (Y.M.V.-D.); (E.V.-A.)
- Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis F. Duany-Almira
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (I.Y.R.-B.); (L.F.D.-A.); (Y.M.V.-D.); (E.V.-A.)
- Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Yeminia M. Valle-Delgadillo
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (I.Y.R.-B.); (L.F.D.-A.); (Y.M.V.-D.); (E.V.-A.)
| | - Héctor E. Flores-Salinas
- Especialidad en Cardiología, Unidad Médica de Alta Especialidad, Centro Médico Nacional de Occidente (CMNO), Departamento de Cardiología, Instituto Mexicano Del Seguro Social (IMSS), Guadalajara 44340, Jalisco, Mexico;
| | - Emmanuel Valdés-Alvarado
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (I.Y.R.-B.); (L.F.D.-A.); (Y.M.V.-D.); (E.V.-A.)
| | - Jorge R. Padilla-Gutiérrez
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (I.Y.R.-B.); (L.F.D.-A.); (Y.M.V.-D.); (E.V.-A.)
| |
Collapse
|
5
|
Ioannou P, Katsoulieris E, Afratis NA. Matrix Dynamics and Microbiome Crosstalk: Matrix Metalloproteinases as Key Players in Disease and Therapy. Int J Mol Sci 2025; 26:3621. [PMID: 40332093 PMCID: PMC12027064 DOI: 10.3390/ijms26083621] [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: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix (ECM) remodeling, regulating a wide range of cellular and immune processes in both homeostatic and pathological conditions. Host-microbiota interactions play a critical role in maintaining ECM balance; however, during dysbiosis, this regulation is disrupted, leading to compromised barrier integrity, pathogen translocation into circulation, and the development of systemic diseases and cancer. This review highlights the bidirectional relationship between MMP expression/activity and microbiota dysbiosis, emphasizing tissue-specific alterations in MMP activity that contribute to disease progression. In addition, it integrates interdisciplinary evidence to illustrate the MMP-dependent mechanisms underlying various pathologies associated with oral and gut microbiome dysbiosis, including long-range effects through the gut-skin and gut-brain axes. Thus, this review introduces the emerging field of MatrixBiome, which explores the complex interactions between the ECM, microbiota, and host tissues. Finally, it also outlines therapeutic strategies to modulate MMP levels, either indirectly through microbiome-targeted approaches (e.g., prebiotics, probiotics, and postbiotics) or directly using MMP inhibitors, offering promising avenues for future clinical interventions.
Collapse
Affiliation(s)
- Paraskevi Ioannou
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
| | - Elias Katsoulieris
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
| | - Nikolaos A. Afratis
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| |
Collapse
|
6
|
Zhao X, Su S, Zhou J, Gao J, Tang X, Wen B. Metabolism and Excretion of 8-O-Acetylharpagide in Rats and Identification of Its Potential Anti-Breast Cancer Active Metabolites. Drug Des Devel Ther 2025; 19:2795-2815. [PMID: 40231196 PMCID: PMC11995923 DOI: 10.2147/dddt.s487898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Background Ajuga decumbens, a traditional Chinese medicine, possesses anti-breast cancer effects. Its main component, 8-O-acetylharpagide, exhibits potential anticancer activity; however, the active metabolites and mechanisms underlying its effects remain unclear. Methods The metabolism and excretion of 8-O-acetylharpagide in rats were investigated using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry analysis of bile, urine, and feces. Active metabolites were identified and evaluated using network pharmacology, molecular docking, and Western blotting assays. Results A total of 21 metabolites were identified, with demethylation, hydrolysis, and glucuronidation being the primary metabolic pathways. M3 and M5 were identified as key metabolites, showing strong binding affinity to cancer-related targets, such as AKT1, MMP9, and STAT3. M5 displayed strong pharmacokinetic properties, including better lipid solubility and reduced polarity. Network pharmacology analysis indicated that these metabolites exert anticancer effects by modulating the PI3K/AKT signaling pathway. In vivo experiments demonstrated that oral administration of 8-O-acetylharpagide significantly inhibited the proliferation of 4T1 tumor tissues by suppressing the expression of the AKT/NF-κB/MMP9 signaling axis. This may be related to inhibition of the expression of the AKT/NF-κB/MMP9 signaling axis in 4T1 tumor tissues after metabolism of 8-O-acetylharpagide to M5 and M3. Conclusion As a prodrug, 8-O-acetylharpagide is metabolized to M5, which may subsequently exert an anti-breast cancer effect through downregulation of the AKT/NF-κB/MMP9 signaling axis. This study provides a theoretical basis for the clinical application of Ajuga decumbens in breast cancer therapy.
Collapse
MESH Headings
- Animals
- Rats
- Female
- Rats, Sprague-Dawley
- Cell Proliferation/drug effects
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Antineoplastic Agents, Phytogenic/metabolism
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/chemistry
- Mice
- Drug Screening Assays, Antitumor
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Humans
- Molecular Structure
- Dose-Response Relationship, Drug
- Molecular Docking Simulation
- Administration, Oral
Collapse
Affiliation(s)
- Xinyu Zhao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Sijia Su
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Jingna Zhou
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Junfeng Gao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Xu Tang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Binyu Wen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| |
Collapse
|
7
|
Ohara R, Dario FL, Emílio-Silva MT, Assunção R, Rodrigues VP, Bueno G, Raimundo PR, Justulin LA, da Rocha LRM, Hiruma-Lima CA. A high-fat diet changes the interaction of the extracellular matrix, cytokines, and growth factors in gastric ulcer repair. Biochem Biophys Res Commun 2025; 755:151565. [PMID: 40043617 DOI: 10.1016/j.bbrc.2025.151565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Obesity is characterized by persistent low-grade inflammation that alters the gastrointestinal system and healing process. The link between obesity and the prevalence of stomach ulcers has not yet been fully established. AIMS We investigated the healing features of gastric lesions in male Swiss mice fed a standard diet (SD) or high-fat diet (HFD) using morphometric, biochemical, and molecular parameters. METHODS After 12 weeks on different diets, the animals underwent acetic acid-induced stomach ulcer surgery. To evaluate healing patterns, the stomachs of the animals were studied at five post-induction times, including the early, middle, and late phases of healing (1, 3, 7, 10, and 14 days). Morphometric features, activity of matrix metalloproteinases 2 and 9 (MMP-2 and 9), and measurement of inflammatory and growth factors were investigated using multiplex immunoassays. RESULTS Compared with the SD group, the HFD group demonstrated slowing of the early healing process. During the initial phase of the healing process, the SD group had significantly higher levels of EGF, VEGF-A, and VEGF-D than the HFD group. In the intermediate phase, only the SD group showed a 70 % increase in the regeneration area compared with the initial phase of the procedure. In this phase, the SD group also had higher levels of MMP-9, VEGF-D, and HGF than the HFD group. CONCLUSIONS HFD can have a negative impact on the healing process of gastric ulcers in animals by delaying repair in gastric tissue when compared with animals consuming SD.
Collapse
Affiliation(s)
- Rie Ohara
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| | - Felipe Lima Dario
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Maycon Tavares Emílio-Silva
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Renata Assunção
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Vinícius Peixoto Rodrigues
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Gabriela Bueno
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Priscila Romano Raimundo
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Morphology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Lúcia Regina Machado da Rocha
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Clelia Akiko Hiruma-Lima
- Department of Structural and Functional Biology, Physiology Sector, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
8
|
Alberti G, Russo E, Lo Iacono M, Di Pace MR, Grasso F, Baldanza F, Pensabene M, La Rocca G, Sergio M. Matrix Metalloproteinases in Ureteropelvic Junction Obstruction: Their Role in Pathogenesis and Their Use as Clinical Markers. Cells 2025; 14:520. [PMID: 40214474 PMCID: PMC11988040 DOI: 10.3390/cells14070520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
The obstruction of the urinary tract is responsible for obstructive nephropathy (ON), also known as uropathy, which may then evolve in a renal parenchymal disease (hydronephrosis). Regarding the etiology of ON, it has been linked to the perturbation of processes occurring during the urinary tract development such as morphogenesis, maturation, and growth. Despite the research carried out in recent years, there is still a pressing need to elucidate the molecular processes underlying the disease. This may then result in the definition of novel biomarkers that can help in patient stratification and the monitoring of therapeutic choices. Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases with key roles in extracellular matrix remodeling due to their wide cleavage specificity and ability to modulate the bioavailability of growth factors. Despite the known changes in the local tissue microenvironment at the site of the urinary tract obstruction, the role of MMPs in ureteropelvic junction obstruction (UPJO) and, therefore, in the pathogenesis of renal damage in ON is not well-documented. In this review, we underline the possible roles of MMPs both in the pathogenesis of UPJO and in the progression of related hydronephrosis. The potential use of MMPs as biomarkers detectable in bodily fluids (such as the patient's urine) is also discussed.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.L.I.)
| | - Eleonora Russo
- Departmental Faculty of Medicine, Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Melania Lo Iacono
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.L.I.)
| | - Maria Rita Di Pace
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (M.R.D.P.); (F.G.); (F.B.); (M.P.)
| | - Francesco Grasso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (M.R.D.P.); (F.G.); (F.B.); (M.P.)
| | - Fabio Baldanza
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (M.R.D.P.); (F.G.); (F.B.); (M.P.)
| | - Marco Pensabene
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (M.R.D.P.); (F.G.); (F.B.); (M.P.)
| | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (M.L.I.)
| | - Maria Sergio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (M.R.D.P.); (F.G.); (F.B.); (M.P.)
| |
Collapse
|
9
|
Gatlin V, Gupta S, Romero S, Chapkin RS, Cai JJ. Exploring cell-to-cell variability and functional insights through differentially variable gene analysis. NPJ Syst Biol Appl 2025; 11:29. [PMID: 40113778 PMCID: PMC11926233 DOI: 10.1038/s41540-025-00507-z] [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: 08/28/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of cellular variability by capturing gene expression profiles of individual cells. The importance of cell-to-cell variability in determining and shaping cell function has been widely appreciated. Nevertheless, differential expression (DE) analysis remains a cornerstone method in analytical practice. Current computational analyses overlook the rich information encoded by variability within the single-cell gene expression data by focusing exclusively on mean expression. To offer a deeper understanding of cellular systems, there is a need for approaches to assess data variability rather than just the mean. Here we present spline-DV, a statistical framework for differential variability (DV) analysis using scRNA-seq data. The spline-DV method identifies genes exhibiting significantly increased or decreased expression variability among cells derived from two experimental conditions. Case studies show that DV genes identified using spline-DV are representative and functionally relevant to tested cellular conditions, including obesity, fibrosis, and cancer.
Collapse
Affiliation(s)
- Victoria Gatlin
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
| | - Shreyan Gupta
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
| | - Selim Romero
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Robert S Chapkin
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - James J Cai
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA.
- CPRIT Single Cell Data Science Core, Texas A&M University, College Station, TX, 77843, USA.
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
10
|
Wang X, Rong C, Leng W, Niu P, He Z, Wang G, Qi X, Zhao D, Li J. Effect and mechanism of Dichloroacetate in the treatment of stroke and the resolution strategy for side effect. Eur J Med Res 2025; 30:148. [PMID: 40025562 PMCID: PMC11874805 DOI: 10.1186/s40001-025-02399-5] [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: 12/30/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Stroke is a serious disease that leads to high morbidity and mortality, and ischemic stroke accounts for more than 80% of strokes. At present, the only effective drug recombinant tissue plasminogen activator is limited by its indications, and its clinical application rate is not high. Therefore, it is urgent to develop effective new drugs according to the pathological mechanism. In the hypoxic state after ischemic stroke, anaerobic glycolysis has become the main way to provide energy to the brain. This process is essential for the maintenance of important brain functions and has important implications for recovery after stroke. However, acidosis caused by anaerobic glycolysis and lactic acid accumulation is an important pathological process after ischemic stroke. Dichloroacetate (DCA) is an orphan drug that has been used for decades to treat children with genetic mitochondrial diseases. Some studies have confirmed the role of DCA in stroke, but the conclusions are conflicting because some believe that DCA is not effective for ischemic stroke and may aggravate hemorrhagic stroke. This study reviews these studies and finds that DCA has a good effect on ischemic stroke. DCA can protect ischemic stroke by improving oxidative stress, reducing neuroinflammation, inhibiting apoptosis, protecting blood-brain barrier, and regulating metabolism. We also describe the differences in the outcomes of DCA in the treatment of ischemic stroke and the reasons why DCA aggravate hemorrhagic stroke. In addition, DCA, as a water disinfection byproduct, has been concerned about its toxicity. We describe the causes and solutions of peripheral neuropathy caused by DCA. In summary, this study analyzes the neuroprotective mechanism of DCA in ischemic stroke and the contradiction of the different research results, and discusses the causes and solutions of its adverse effects.
Collapse
Affiliation(s)
- Xu Wang
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chunshu Rong
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Wei Leng
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Ping Niu
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Ziqiao He
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Gaihua Wang
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Xin Qi
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - Dexi Zhao
- Department of Encephalopathy, Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
11
|
Taheri E, Raeeszadeh-Sarmazdeh M. Effect of TIMPs and their minimally engineered variants in blocking invasion and migration of brain cancer cells. Oncotarget 2025; 16:118-130. [PMID: 40019229 PMCID: PMC11870142 DOI: 10.18632/oncotarget.28691] [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: 06/05/2024] [Accepted: 01/03/2025] [Indexed: 03/01/2025] Open
Abstract
Matrix metalloproteinases (MMPs) are crucial in remodeling the extracellular matrix (ECM), modulating key processes involved in cancer progression, such as migration, invasion, angiogenesis, and metastasis. The overexpression of MMPs, particularly MMP-9, is markedly observed in glioblastoma multiforme (GBM), an aggressive primary brain tumor known for its diffuse and infiltrative nature. Tissue inhibitors of metalloproteinases (TIMPs), endogenous MMP inhibitors, offer significant therapeutic potential due to their wider interaction interfaces relative to small molecule inhibitors. Here, we studied the effect of wild-type human TIMP-1 and TIMP-3 and minimal TIMP variants (mTC1 and mTC3), previously engineered for MMP inhibition, on migration and invasion of GBM cells. Our study focused on minimal TIMP variants, due to their small molecular size and potential in higher cellular uptake and delivery, to assess their potential in cell-based assays. The results demonstrated that the minimal TIMP variants, mTC1, and mTC3, effectively inhibit MMP activity underscoring their potential to limit tumor invasion and progression. Given the lethal nature of GBM and the limited efficacy of current therapies, the application of TIMPs and their engineered minimal variants represents a novel and potentially transformative approach to regulating MMP activity in GBM.
Collapse
Affiliation(s)
- Elham Taheri
- Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | | |
Collapse
|
12
|
Wang C, Wu L, Li X, Mao S, Wang Z, Xie H, Xu J, Li D, Yang H, Wang T, Huang Q, Yang L, Zhang X, Liu Y, Wang J, Huang G, Xie W. Frizzled-7-targeting antibody-derived bifunctional protein retargets NK cells against triple-negative breast cancer cells via MICA-NKG2D axis. J Immunother Cancer 2025; 13:e009621. [PMID: 40021214 PMCID: PMC11873350 DOI: 10.1136/jitc-2024-009621] [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: 05/09/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Hypoxia is associated with the evasion of triple-negative breast cancer (TNBC) from immune surveillance. Hypoxia increases the subpopulation of putative TNBC stem-like cells (TNBCSCs) through activating Wnt/β-Catenin signaling. The shedding of MHC class I-related chain A (MICA) is particularly noteworthy in cancer stem cells (CSCs), promoting the resistance of CSCs to natural killer (NK) cell cytotoxicity. To reestablish MICA/NKG2D-mediated immunosurveillance, we proposed the design of a fusion protein (SHH002-hu1-MICA) which consists of Frizzled-7 (Fzd7)-targeting antibody and MICA, serving as an engager retargeting NK cells against TNBCs, especially TNBCSCs. METHODS Opal multicolor immunohistochemistry staining was used to validate the expression of membrane MICA (mMICA) and existence of NK cells in TNBC tumors; flow cytometry (FCM) assay was used to detect the expression of Fzd7/mMICA on TNBCs. Biolayer interferometry (BLI) and surface plasmon resonance (SPR) assays were executed to assess the affinity of SHH002-hu1-MICA towards rhFzd7/rhNKG2D; near-infrared imaging assay was used to evaluate the targeting capability. A cytotoxicity assay was conducted to assess the effects of SHH002-hu1-MICA on NK cell-mediated killing of TNBCs, and FCM assay to analyze the effects of SHH002-hu1-MICA on the degranulation of NK cells. Finally, TNBC cell-line-derived xenografts were established to evaluate the anti-tumor activities of SHH002-hu1-MICA in vivo. RESULTS The expression of mMICA is significantly downregulated in hypoxic TNBCs and TNBCSCs, leading to the evasion of immune surveillance exerted by NK cells. The expression of Fzd7 is significantly upregulated in TNBCSCs and exhibits a negative correlation with the expression of mMICA and infiltration level of NK cells. On accurate assembly, SHH002-hu1-MICA shows a strong affinity for rhFzd7/rhNKG2D, specifically targets TNBC tumor tissues, and disrupts Wnt/β-Catenin signaling. SHH002-hu1-MICA significantly enhances the cytotoxicity of NK cells against hypoxic TNBCs and TNBCSCs by inducing the degranulation of NK cells and promotes the infiltration of NK cells in CD44high regions within TNBC xenograft tumors, exhibiting superior anti-tumor activities than SHH002-hu1. CONCLUSIONS SHH002-hu1-MICA maintains the targeting property of SHH002-hu1, successfully activates and retargets NK cells against TNBCs, especially TNBCSCs, exhibiting superior antitumor activities than SHH002-hu1. SHH002-hu1-MICA represents a promising new engager for NK cell-based immunotherapy for TNBC.
Collapse
Affiliation(s)
- Chenyue Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lisha Wu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xingxing Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Shuyang Mao
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zitong Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Haiyan Xie
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jing Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Danfang Li
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Tong Wang
- Tulane University Biomedical Informatics & Genomics Center, New Orleans, Louisiana, USA
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey, USA
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Lin Yang
- Joint Innovation Laboratory for Cell Therapy Technology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiaofei Zhang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuxia Liu
- Department of Applied Chemistry, Shanghai Institute of Applied Physics Chinese Academy of Sciences, Shanghai, China
| | - Jin Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Wei Xie
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
13
|
Rhind SG, Shiu MY, Tenn C, Nakashima A, Jetly R, Sajja VSSS, Long JB, Vartanian O. Repetitive Low-Level Blast Exposure Alters Circulating Myeloperoxidase, Matrix Metalloproteinases, and Neurovascular Endothelial Molecules in Experienced Military Breachers. Int J Mol Sci 2025; 26:1808. [PMID: 40076437 PMCID: PMC11898641 DOI: 10.3390/ijms26051808] [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: 12/10/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Repeated exposure to low-level blast overpressure, frequently experienced during explosive breaching and heavy weapons use in training and operations, is increasingly recognised as a serious risk to the neurological health of military personnel. Although research on the underlying pathobiological mechanisms in humans remains limited, this study investigated the effects of such exposure on circulating molecular biomarkers associated with inflammation, neurovascular damage, and endothelial injury. Blood samples from military breachers were analysed for myeloperoxidase (MPO), matrix metalloproteinases (MMPs), and junctional proteins indicative of blood-brain barrier (BBB) disruption and endothelial damage, including occludin (OCLN), zonula occludens-1 (ZO-1), aquaporin-4 (AQP4), and syndecan-1 (SD-1). The results revealed significantly elevated levels of MPO, MMP-3, MMP-9, and MMP-10 in breachers compared to unexposed controls, suggesting heightened inflammation, oxidative stress, and vascular injury. Increased levels of OCLN and SD-1 further indicated BBB disruption and endothelial glycocalyx degradation in breachers. These findings highlight the potential for chronic neurovascular unit damage/dysfunction from repeated blast exposure and underscore the importance of early targeted interventions-such as reducing oxidative stress, reinforcing BBB integrity, and managing inflammation-that could be essential in mitigating the risk of long-term neurological impairment associated with blast exposure.
Collapse
Affiliation(s)
- Shawn G. Rhind
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON M3K 2C9, USA; (M.Y.S.); (O.V.)
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, ON M5S 2W6, Canada
| | - Maria Y. Shiu
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON M3K 2C9, USA; (M.Y.S.); (O.V.)
| | - Catherine Tenn
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB T1A 8K6, Canada;
| | - Ann Nakashima
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON M3K 2C9, USA; (M.Y.S.); (O.V.)
| | - Rakesh Jetly
- The Institute of Mental Health Research, University of Ottawa, Royal Ottawa Hospital, Ottawa, ON K1Z 7K4, Canada;
| | | | - Joseph B. Long
- Blast-Induced NeuroTrauma Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA (J.B.L.)
| | - Oshin Vartanian
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON M3K 2C9, USA; (M.Y.S.); (O.V.)
- Department of Psychology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
14
|
Oselusi SO, Sibuyi NR, Martin DR, Meyer M, Madiehe AM. Potential matrix metalloproteinase 2 and 9 inhibitors identified from Ehretia species for the treatment of chronic wounds - Computational drug discovery approaches. Comput Biol Med 2025; 185:109487. [PMID: 39637455 DOI: 10.1016/j.compbiomed.2024.109487] [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: 04/26/2024] [Revised: 10/29/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Matrix metalloproteinases (MMPs) serve as prognostic factors in several pathophysiological conditions, including chronic wounds. Therefore, they are considered important therapeutic targets in the intervention and treatment of these conditions. In this study, computational tools such as molecular docking and molecular dynamics simulations were used to gain insight into protein‒ligand interactions and determine the free binding energy between Ehretia species phytoconstituents and gelatinases (MMP2 and MMP9). A total of 74 phytoconstituents from Ehretia species were compiled from the literature, and 46 of these compounds were identified as potential inhibitors of at least one type of MMP. Molecular docking revealed that lithospermic acid B, rosmarinic acid, and danshensu had stronger binding affinities against the two enzymes than the reference ligands. Furthermore, (9S, 10E, 12Z, 15Z)-9-hydroxy-10,12,15-octadecatrienoic (∗-octadecatrienoic) had a higher binding energy for MMP2, whereas caffeic anhydride and caffeic acid established stronger binding energy with MMP9 than the reference ligand. These complexes also demonstrated relatively stable, favourable, and comparable conformational changes with those of unbound proteins at 500 ns. The free energy decomposition results further provide detailed insights into the contributions of active site residues and different types of interactions to the overall binding free energy. Finally, most of the hit phytoconstituents (rosmarinic acid, caffeic anhydride, caffeic acid, and danshensu) had good physicochemical, drug-likeness, and pharmacokinetic properties. Collectively, our findings showed that phytoconstituents from Ehretia species could be beneficial in the search for novel MMP inhibitors as therapeutic agents for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Samson O Oselusi
- Nanobiotechnology Research Group, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa; DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa
| | - Nicole Rs Sibuyi
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa; Health Platform, Advanced Materials Division, Mintek, 200 Malibongwe Drive, Randburg, South Africa
| | - Darius R Martin
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa
| | - Mervin Meyer
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa
| | - Abram M Madiehe
- Nanobiotechnology Research Group, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa; DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town, 7535, South Africa.
| |
Collapse
|
15
|
Wu L, Liu C, Hu W. Comprehensive investigation of matrix metalloproteinases in skin cutaneous melanoma: diagnostic, prognostic, and therapeutic insights. Sci Rep 2025; 15:2152. [PMID: 39820824 PMCID: PMC11739484 DOI: 10.1038/s41598-025-85887-2] [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: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
The dysregulation of matrix metalloproteinases (MMPs) in skin cutaneous melanoma (SKCM) represents a critical aspect of tumorigenesis. In this study, we investigated the diagnostic, prognostic, and therapeutic aspects of the MMPs in SKCM. Thirteen SKCM cell lines and seven normal skin cell lines were cultured under standard conditions for experimental analyses. RNA and DNA were extracted, followed by RT-qPCR to assess MMP expression and promoter methylation analysis to determine methylation levels. Functional assays, including cell proliferation, colony formation, and wound healing, were conducted post-MMP7 knockdown using siRNA in A375 cells. Databases like GEPIA2, HPA, MEXPRESS, and miRNet were employed for expression, survival, methylation, and miRNA-mRNA network analyses. We investigated the expression and promoter methylation landscape of MMPs in SKCM cell lines, revealing significant (p-value < 0.05) up-regulation of MMP1, MMP7, MMP9, MMP10, MMP11, MMP12, MMP13, MMP14, and MMP25, alongside down-regulation of MMP2, MMP3, and MMP21. Furthermore, our analysis demonstrated a significant (p-value < 0.05) inverse correlation between MMP expression levels and promoter methylation status, suggesting a potential regulatory role of DNA methylation in MMP dysregulation. Notably, MMP7, MMP11, and MMP14 exhibited significant (p-value < 0.05) associations with the overall survival of SKCM patients, emphasizing their prognostic significance. Additionally, Receiver operating characteristic (ROC) curve analysis highlighted the significant (p-value < 0.05) diagnostic potential of MMP7, MMP11, and MMP14 in distinguishing SKCM from normal individuals. Subsequent validation across multiple cohorts confirmed significant (p-value < 0.05) elevated MMP expression levels in SKCM tissues, particularly in advanced disease stages, further emphasizing their role in tumor progression. Furthermore, we elucidated potential regulatory pathways involving miR-22-3p, which targets MMP7, MMP11, and MMP14 genes in SKCM. Our findings also revealed associations between MMP expression and immune modulation, drug sensitivity, and functional states of SKCM cells. Lastly, MMP7 knockdown in A375 cells significantly significant (p-value < 0.05) impacted several characteristics, including cell proliferation, colony formation, and wound healing. Our findings highlight the diagnostic, prognostic, and therapeutic potential of MMP7, MMP11, and MMP14 in SKCM. These MMPs could serve as biomarkers for early detection and targets for therapy. Future efforts should focus on preclinical and clinical validation to translate these insights into personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lingxia Wu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China
| | - Chenxiaoxiao Liu
- The First Clinical Institute, Zunyi Medical University, Zunyi, 520300, Guizhou, China
| | - Weicai Hu
- Dermatology, Changzhi Second People's Hospital, Changzhi, 046000, Shanxi, China.
| |
Collapse
|
16
|
Fallahi F, Askari N, Jamali T, Parsapour S, Ghasemi H, Shams J, Yaraee R, Ghazanfari Z, Ghazanfari T. MMP-9 and TIMPs profiles in sulfur mustard-exposed individuals with serious lung complications. Int Immunopharmacol 2025; 145:113777. [PMID: 39657535 DOI: 10.1016/j.intimp.2024.113777] [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: 10/07/2024] [Revised: 11/13/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024]
Abstract
Sulfur mustard (SM), a chemical weapon used in the Iraq-Iran war, can pose severe health risks, especially to the lungs. Dysregulation of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) have been implicated in various inflammatory lung diseases. This study compares the levels of MMPs and TIMPs in the serum and sputum of veterans with serious lung complications to a control group. Serum and sputum samples were collected and analyzed using the ELISA sandwich method. Differences between SM-exposed and control groups were assessed statistically. The serum levels of TIMP-4 and MMP-9/TIMP-4 were significantly lower and higher in the SM-exposed group respectively compared to the control group. In SM-exposed individuals resembling Bronchiolitis Obliterans (BO), Chronic Bronchitis (CB), and Asthma, TIMP-4 levels were lower than controls, while TIMP-2 levels were higher in those with CB. Although the increased TIMP-2 levels in these patients align with COPD studies, differences were observed in other factors with COPD and asthma-related MMP-9 and TIMP-4 findings. Assessment of serum levels of these factors based on severity reveals lower MMP-9/TIMP-4 levels in the severe group compared to the mild-moderate group. Individuals exposed to SM exhibit distinct MMP and TIMP profiles, with significantly lower TIMP-4 levels and higher MMP-9/TIMP-4 ratios, compared to controls. These profiles vary across different lung conditions, indicating a unique disease mechanism in SM-exposed individuals. This distinctive profile supports the classification of this condition as 'Mustard Lung.' Further research is needed to elucidate these mechanisms for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Faramarz Fallahi
- Department of Cardiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Nayere Askari
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran; Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Tahereh Jamali
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | | | - Hassan Ghasemi
- Department of Ophthalmology, Shahed University, Tehran, Iran
| | - Jalaledin Shams
- Hematology-Oncology Unit, Internal Medicine Department, Shahed University, Tehran, Iran; Department of Oncology and Hematology, Shahed University, Tehran, Iran
| | - Roya Yaraee
- Department of Immunology, Shahed University, Tehran, Iran
| | - Zeinab Ghazanfari
- Department of Health Education and Promotion, Faculty of Health, Ilam University of Medical Sciences, Ilam, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Immunology, Shahed University, Tehran, Iran.
| |
Collapse
|
17
|
Gupta P, Rani V. Gel Diffusion-Based Gelatin Zymography to Analyze MMP-2 and MMP-9 Activity in Cell Culture. Methods Mol Biol 2025; 2917:213-223. [PMID: 40347344 DOI: 10.1007/978-1-0716-4478-2_18] [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: 05/12/2025]
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes regulating the degradation of collagen as well as other proteins in the extracellular matrix (ECM), tissue repair, and remodeling. Validating MMPs as current drug targets can potentially establish the clinical relevance of their quantitative expression and distribution and open new therapeutic venues in tumor and metastasis. Zymography is a technique used for the identification of the proteolytic activity of MMPs in diverse biological samples. In gel diffusion zymography, analysis of the functional activity of the gelatinases (MMP-2 and MMP-9) by the digestion of gelatin as a substrate, added to agarose gel to assess the active forms of MMPs, is done. Here we describe a detailed experimental protocol to quantitate the gelatinase activity within the cell culture.
Collapse
Affiliation(s)
- Priyadarshini Gupta
- Transcriptome Laboratory, Centre of Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Vibha Rani
- Transcriptome Laboratory, Centre of Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India.
| |
Collapse
|
18
|
Jain P, Gupta A, Mrinalini, Sharma M. Role of MMP inhibitors on levels of MMP-2 and MMP-9 in mammalian cell lines - A systematic review. J Oral Maxillofac Pathol 2025; 29:109-116. [PMID: 40248615 PMCID: PMC12002592 DOI: 10.4103/jomfp.jomfp_279_24] [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: 09/26/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 04/19/2025] Open
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes involved in extracellular matrix (ECM) degradation, contributing to various pathological conditions, including periapical lesions and periodontal diseases. This systematic review evaluates the inhibitory effects of different natural and synthetic MMP inhibitors on MMP-2 and MMP-9 activities in mammalian cells, which are critical enzymes implicated in ECM breakdown. A comprehensive literature search was performed across databases, including PubMed, Scopus, and Cochrane until June 2023, following PRISMA guidelines. The Office of Health Assessment and Translation (OHAT) risk of bias tool was used for quality assessment, revealing a low risk of bias across all studies. Our findings demonstrate that both natural and synthetic MMP inhibitors significantly reduce MMP-2 and MMP-9 activities in mammalian cells. These compounds offer potential therapeutic benefits in managing diseases characterized by excessive MMP activity, such as periapical lesions and periodontal disease. This review highlights the therapeutic potential of MMP inhibitors in dentistry, specifically focusing on the promising roles of natural and synthetic MMP inhibitors in protecting ECM integrity.
Collapse
Affiliation(s)
- Palak Jain
- Department of Conservative Dentistry and Endodontics, Manav Rachna Dental College, SDS, MRIIRS, Faridabad, Haryana, India
| | - Alpa Gupta
- Department of Conservative Dentistry and Endodontics, Manav Rachna Dental College, SDS, MRIIRS, Faridabad, Haryana, India
| | - Mrinalini
- Department of Conservative Dentistry and Endodontics, Manav Rachna Dental College, SDS, MRIIRS, Faridabad, Haryana, India
| | - Mohit Sharma
- Department of Oral Pathology, SGT Dental College Hospital and Research Institute, Gurugram, Haryana, India
| |
Collapse
|
19
|
Dutta S, Zhu Y, Almuntashiri S, Peh HY, Zuñiga J, Zhang D, Somanath PR, Ramírez G, Irineo-Moreno V, Jiménez-Juárez F, López-Salinas K, Regino N, Campero P, Crocker SJ, Owen CA, Wang X. PDGFRα-positive cell-derived TIMP-1 modulates adaptive immune responses to influenza A viral infection. Am J Physiol Lung Cell Mol Physiol 2025; 328:L60-L74. [PMID: 39585242 PMCID: PMC11905806 DOI: 10.1152/ajplung.00104.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Tissue inhibitor of metalloproteinases-1 (TIMP-1) is a physiologic inhibitor of the matrix metalloproteinases (MMPs), but little is known about the role of TIMP-1 in regulating the pathogenesis of influenza A virus (IAV) infection. Here, we performed both in vivo and in vitro experiments to investigate the regulation and function of TIMP-1 during IAV infection. Specifically, plasma levels of TIMP-1 are significantly increased in human subjects and wild-type (WT) mice infected with 2009 H1N1 IAV compared with levels in uninfected controls. Also, TIMP-1 is strikingly upregulated in PDGFRα positive (PDGFRα+) cells in IAV-infected murine lungs as demonstrated using conditional KO (cKO) mice with a specific deletion of Timp-1 in PDGFRα+ cells. Our in vitro data indicated that TIMP-1 is induced by transforming growth factor-β (TGF-β) during lipofibroblasts (lipoFBs)-to-myofibroblast (myoFB) transdifferentiation. Timp-1 deficiency protects mice from H1N1 IAV-induced weight loss, mortality, and lung injury. IAV-infected Timp-1-deficient mice showed increased macrophages, and B and T cell counts in bronchoalveolar lavage (BAL) on day 7 postinfection (p.i.), but reduced BAL neutrophil counts. Increased Cxcl12 levels were detected in both BAL cells and lungs from Timp-1-deficient mice on day 3 p.i. Taken together, our data strongly link TIMP-1 to IAV pathogenesis. We identified that PDGFRα-lineage cells are the main cellular source of elevated TIMP-1 during IAV infection. Loss of Timp-1 attenuates IAV-induced mortality and promotes T and B cell recruitment. Thus, TIMP-1 may be a novel therapeutic target for IAV infection.NEW & NOTEWORTHY Our data strongly link tissue inhibitor of metalloproteinases-1 (TIMP-1) to influenza A virus (IAV) pathogenesis. TIMP-1 is highly increased in PDGFRα-lineage cells during IAV infection. Transforming growth factor-β (TGF-β) induces TIMP-1 during lipofibroblast (lipoFB)-to- myofibroblast (myoFB) transdifferentiation. Timp-1 deficiency protects mice from H1N1 IAV-induced weight loss, mortality, and lung injury. TIMP-1 may be a novel therapeutic target for IAV infection.
Collapse
Affiliation(s)
- Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| | - Joaquin Zuñiga
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
| | - Gustavo Ramírez
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Karen López-Salinas
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Nora Regino
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Mexico City, Mexico
| | - Paloma Campero
- Laboratory of Immunobiology and Genetics and Intensive Care Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, United States
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, United States
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
20
|
Singh P, Kumari S, Chakravortty H, Pandey A, Dash D, Singh R. In vivo, in vitro, and in silico approaches in the detailed study of di-butyl phthalate (DBP), a plasticizer-induced lung fibrosis via Nrf-2/Keap-1/HO-1 pathway and its regulation. Bioorg Chem 2025; 154:107970. [PMID: 39591687 DOI: 10.1016/j.bioorg.2024.107970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
The alveolar epithelium is a crucial barrier against external threats, yet it becomes a key player in initiating pulmonary fibrosis when compromised. Despite its importance, the intricate relationship between, DBP exposure and alveolar epithelial cell injury ensuing pro-fibrotic effects remains poorly understood. Phthalates, ubiquitous in nature, pose a significant risk to lung health upon inhalation, acting as immune triggers that cause airway inflammation and epithelial damage. We aimed to investigate the impact of intranasal administration ofDi-butyl Phthalate (DBP) inhalation, and its probable effects on normal and asthmatic lungs. DBP was administered via intranasal route in normal and OVA-induced asthmatic mice. DBP exposure enhanced oxidative stress and inflammatory parameters, leading to exacerbated asthmatic response and oxidative lung damage. Enhanced accumulation of immune cells, bronchial thickening, and collagen deposition was noted in histopathological investigations of DBP-exposed lung sections. Curcumin, a plant-derived molecule, significantly mitigated DBP-exposed asthma exacerbations by suppressing NF-κB expression and enhancing NRF2 levels via the Nrf-2/Keap-1/HO-1 signaling pathway. FACS analysis revealed increased CD11b+ cells (32 %) in asthmatic mice which were reduced in the curcumin pre-treatment group (10.5 %). Enhanced epithelial to mesenchymal transition (EMT) was noted in mice lungs and A549 cells where E-cadherin expression was reduced as compared to Vimentin, and α-SMA. Apart from aggravated airway inflammation, DBP exposure damages healthy lungs also. MMP-9/TIMP-1 ratios and collagen-1 levels were restored which were enhanced after DBP exposure. Moreover, antioxidant enzyme levels such as NQO-1, HO-1, and Catalase were significantly enhanced (p < 0.01) and comparable to dexamethasone, a conventional corticosteroid. Notably, both dexamethasone and curcumin treatments effectively regulated the stimulation and accumulation of Nrf-2 in the nucleus, promoting antioxidant production and offering potential therapeutic benefits in mitigating pulmonary fibrosis. OVA and DBP alone caused DNA damage in the lung cells where increasedpercentage of damaged DNA movement in thetail, tail length, tail moment, and olive tail moment indicated severe damage in theDBP and OVA combined exposure strategies. Dexamethasone and Curcumin treatments reduced theextent of the DNA damage indicating anti-inflammatory and ant-oxidative potentials. Moreover, in silico studies are supportive of therapeutic potential of Curcumin and Dexamethasone in DBP-induced lung inflammation and fibrosis.
Collapse
Affiliation(s)
- Payal Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Sneha Kumari
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Harshika Chakravortty
- Department of Bioinformatics, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India
| | - Ajai Pandey
- Department of Kaychikitsa, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - D Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rashmi Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
21
|
Shoari A, Ashja Ardalan A, Dimesa AM, Coban MA. Targeting Invasion: The Role of MMP-2 and MMP-9 Inhibition in Colorectal Cancer Therapy. Biomolecules 2024; 15:35. [PMID: 39858430 PMCID: PMC11762759 DOI: 10.3390/biom15010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the most prevalent and lethal cancers worldwide, prompting ongoing research into innovative therapeutic strategies. This review aims to systematically evaluate the role of gelatinases, specifically MMP-2 and MMP-9, as therapeutic targets in CRC, providing a critical analysis of their potential to improve patient outcomes. Gelatinases, specifically MMP-2 and MMP-9, play critical roles in the processes of tumor growth, invasion, and metastasis. Their expression and activity are significantly elevated in CRC, correlating with poor prognosis and lower survival rates. This review provides a comprehensive overview of the pathophysiological roles of gelatinases in CRC, highlighting their contribution to tumor microenvironment modulation, angiogenesis, and the metastatic cascade. We also critically evaluate recent advancements in the development of gelatinase inhibitors, including small molecule inhibitors, natural compounds, and novel therapeutic approaches like gene silencing techniques. Challenges such as nonspecificity, adverse side effects, and resistance mechanisms are discussed. We explore the potential of gelatinase inhibition in combination therapies, particularly with conventional chemotherapy and emerging targeted treatments, to enhance therapeutic efficacy and overcome resistance. The novelty of this review lies in its integration of recent findings on diverse inhibition strategies with insights into their clinical relevance, offering a roadmap for future research. By addressing the limitations of current approaches and proposing novel strategies, this review underscores the potential of gelatinase inhibitors in CRC prevention and therapy, inspiring further exploration in this promising area of oncological treatment.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Arghavan Ashja Ardalan
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | | | - Mathew A. Coban
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
22
|
Garajová I, Giovannetti E. Targeting Perineural Invasion in Pancreatic Cancer. Cancers (Basel) 2024; 16:4260. [PMID: 39766161 PMCID: PMC11674953 DOI: 10.3390/cancers16244260] [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: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Pancreatic cancer is an aggressive tumor with dismal prognosis. Neural invasion is one of the pathological hallmarks of pancreatic cancer. Peripheral nerves can modulate the phenotype and behavior of the malignant cells, as well as of different components of the tumor microenvironment, and thus affect tumor growth and metastasis. From a clinical point of view, neural invasion is translated into intractable pain and represents a predictor of tumor recurrence and poor prognosis. Several molecules are implicated in neural invasion and pain onset in PDAC, including neutrophins (e.g., NGF), chemokines, adhesion factors, axon-guidance molecules, different proteins, and neurotransmitters. In this review, we discuss the role of nerves within the pancreatic cancer microenvironment, highlighting how infiltrating nerve fibers promote tumor progression and metastasis, while tumor cells, in turn, drive nerve outgrowth in a reciprocal interaction that fuels tumor advancement. We outline key molecules involved in neural invasion in pancreatic cancer and, finally, explore potential therapeutic strategies to target neural invasion, aiming to both inhibit cancer progression and alleviate cancer-associated pain.
Collapse
Affiliation(s)
- Ingrid Garajová
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Lab of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1007 MB Amsterdam, The Netherlands;
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, San Giuliano Terme PI, 56017 Pisa, Italy
| |
Collapse
|
23
|
Kalantar M, Kalanther I, Kumar S, Buxton EK, Raeeszadeh-Sarmazdeh M. Determining key residues of engineered scFv antibody variants with improved MMP-9 binding using deep sequencing and machine learning. Comput Struct Biotechnol J 2024; 23:3759-3770. [PMID: 39525083 PMCID: PMC11550764 DOI: 10.1016/j.csbj.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Given the crucial role of specific matrix metalloproteinases (MMPs) in the extracellular matrix, an imbalance in the regulation of activation of matrix metalloproteinase-9 (MMP-9) zymogen and inhibition of the enzyme can result in various diseases, such as cancer, neurodegenerative, and gynecological diseases. Thus, developing novel therapeutics that target MMP-9 with single-chain antibody fragments (scFvs) is a promising approach. We used fluorescent-activated cell sorting (FACS) to screen a synthetic scFv antibody library displayed on yeast for enhanced binding to MMP-9. The screened scFv mutants demonstrated improved binding to MMP-9 compared to the natural inhibitor of MMPs, tissue inhibitor of metalloproteinases (TIMPs). To identify the molecular determinants of these engineered scFv variants that affect binding to MMP-9, we used next-generation DNA sequencing and computational protein structure analysis. Additionally, a deep-learning language model was trained on the screened scFv library of variants to predict the binding affinities of scFv variants based on their CDR-H3 sequences.
Collapse
Affiliation(s)
- Masoud Kalantar
- Department of Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | | | - Sachin Kumar
- Department of Chemical and Materials Engineering, University of Nevada, Reno, NV 89557, USA
| | | | | |
Collapse
|
24
|
Singh V, Panda SP. Nexus of NFκB/VEGF/MMP9 signaling in diabetic retinopathy-linked dementia: Management by phenolic acid-enabled nanotherapeutics. Life Sci 2024; 358:123123. [PMID: 39419266 DOI: 10.1016/j.lfs.2024.123123] [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: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
AIMS The purpose of this review is to highlight the therapeutic effectiveness of phenolic acids in slowing the progression of diabetic retinopathy (DR)-linked dementia by addressing the nuclear factor kappa B (NFκB)/matrix metalloproteinase-9 (MMP9)/vascular endothelial growth factor (VEGF) interconnected pathway. MATERIALS AND METHODS We searched 80 papers published in the last 20 years using terms like DR, dementia, phenolic acids, NFkB/VEFG/MMP9 signaling, and microRNAs (miRs) in databases including Pub-Med, WOS, and Google Scholar. By encasing phenolic acid in nanoparticles and then controlling its release into the targeted tissues, nanotherapeutics can increase their effectiveness. Results were summarized, and compared, and research gaps were identified throughout the data collection and interpretation. KEY FINDINGS Amyloid beta (Aβ) deposition in neuronal cells and drusen sites of the eye leads to the activation of NFkB/VEGF/MMP9 signaling and microRNAs (miR146a and miR155), which in turn energizes the accumulation of pro-inflammatory and pro-angiogenic microenvironments in the brain and retina leading to DR-linked dementia. This study demonstrates the potential of phenolic acid-enabled nanotherapeutics as a functional food or supplement for preventing and treating DR-linked dementia, and oxidative stress-related diseases. SIGNIFICANCE The retina has mechanisms to clear metabolic waste including Aβ, but the activation of NFkB/ MMP9/ VEGF signaling leads to fatal pathological consequences. Understanding the role of miR146a and miR155 provides potential therapeutic avenues for managing the complex pathology shared between DR and dementia. In particular, phenolic acid nanotherapeutics offer a dual benefit in retinal regeneration and dementia management.
Collapse
Affiliation(s)
- Vikrant Singh
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| |
Collapse
|
25
|
ElKabbany ZA, Ismail EAR, Hamed ET, Elbarbary NS. The impact of vildagliptin as an add-on therapy on matrix metalloproteinase-14 levels, liver stiffness and subclinical atherosclerosis in adolescents with type 1 diabetes and non-alcoholic steatohepatitis: A randomized controlled trial. Diabetes Obes Metab 2024; 26:5857-5869. [PMID: 39318059 DOI: 10.1111/dom.15958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024]
Abstract
AIM Many patients with type 1 diabetes mellitus (T1DM) met the histological criteria for non-alcoholic steatohepatitis (NASH), which leads to cardiovascular disease morbidity and mortality. Matrix metalloproteinase-14 (MMP-14) is involved in cardiovascular disease and atherosclerosis. OBJECTIVES To assess the impact of oral dipeptidyl peptidase-4 inhibitor, vildagliptin, as adjunctive therapy on NASH in adolescents with T1DM and its effect on glycaemic control, MMP-14 levels and carotid intima media thickness (CIMT). METHODS Sixty adolescents with T1DM and NASH were randomly assigned to receive oral vildagliptin (50 mg once daily) for 6 months or not. Glycated haemoglobin, lipid profile, hepatic steatosis index, triglyceride glucose (TyG) index and MMP-14 levels were assessed. Transient elastography with controlled attenuation parameter (CAP) was performed together with measuring CIMT. RESULTS By transient elastography, 12 (20%) patients with T1DM with NASH had elevated liver stiffness ≥7 kPa (F2 stage or higher). Baseline MMP-14 was positively correlated to insulin dose (p = 0.016), triglycerides and TyG index, CIMT, liver stiffness and CAP levels among the studied patients (p < 0.001 for all). After 6 months, patients with T1DM on vildagliptin therapy had significantly lower glycated haemoglobin, lipid profile, hepatic steatosis index and TyG index, as well as MMP-14 (p < 0.001). CIMT, liver stiffness and CAP were significantly decreased post-therapy compared with baseline levels and compared with the control group (p < 0.001). Vildagliptin was safe and well-tolerated. CONCLUSIONS Administration of vildagliptin for adolescents with T1DM and NASH improved glycaemic control, dyslipidaemia and MMP-14 levels and decreased liver stiffness and CIMT; hence, reducing subclinical atherosclerosis and disease progression.
Collapse
|
26
|
Gilmore BF, White TA, Busetti A, McAteer MI, Maggs CA, Thompson TP. Exiguolysin, a Novel Thermolysin (M4) Peptidase from Exiguobacterium oxidotolerans. Microorganisms 2024; 12:2311. [PMID: 39597700 PMCID: PMC11596557 DOI: 10.3390/microorganisms12112311] [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: 10/10/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
This study details a comprehensive biochemical and structural characterization of exiguolysin, a novel thermolysin-like, caseinolytic peptidase secreted by a marine isolate of Exiguobacterium oxidotolerans strain BW26. Exiguolysin demonstrated optimal proteolytic activity at 37 °C and pH 3, retaining 85% activity at 50 °C, highlighting its potential stability under broad reaction conditions. SDS-PAGE and LC-MS analysis identified the enzyme as a 32 kDa M4-family metalloprotease. Exiguolysin activity was inhibited by 1,10-phenanthroline, confirming its dependence on metal ions for activity. Zymographic analysis and substrate specificity assays revealed selective hydrolysis of matrix metalloproteinase (MMP) substrates but no activity against elastase substrates. Analysis of the predicted gene sequence and structural predictions using AlphaFold identified the presence and position of HEXXH and Glu-Xaa-Xaa-Xaa-Asp motifs, crucial for zinc binding and catalytic activity, characteristic of 'Glu-zincins' and members of the M4 peptidase family. High-throughput screening of a 20 × 20 N-alpha mercaptoamide dipeptide inhibitor library against exiguolysin identified SH-CH2-CO-Met-Tyr-NH2 as the most potent inhibitor, with a Ki of 1.95 μM. Notably, exiguolysin selectively inhibited thrombin-induced PAR-1 activation in PC-3 cells, potentially indicating a potential mechanism of virulence in modulating PAR-1 signalling during infection by disarming PARs. This is the first detailed characterization of a peptidase of the M4 (thermolysin) family in the genus Exiguobacterium which may have industrial application potential and relevance as a putative virulence factor.
Collapse
Affiliation(s)
- Brendan F. Gilmore
- Biofilm Research Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
- School of Medicine, University of Limerick, Limerick V94 T9PX, Ireland
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, UK
| | - Tracy A. White
- Biofilm Research Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Alessandro Busetti
- Biofilm Research Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Matthew I. McAteer
- Biofilm Research Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Christine A. Maggs
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, UK
| | - Thomas P. Thompson
- Biofilm Research Group, School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|
27
|
Gelsomino L, Caruso A, Tasan E, Leonetti AE, Malivindi R, Naimo GD, Giordano F, Panza S, Gu G, Perrone B, Giordano C, Mauro L, Nardo B, Filippelli G, Bonofiglio D, Barone I, Fuqua SAW, Catalano S, Andò S. Evidence that CRISPR-Cas9 Y537S-mutant expressing breast cancer cells activate Yes-associated protein 1 to driving the conversion of normal fibroblasts into cancer-associated fibroblasts. Cell Commun Signal 2024; 22:545. [PMID: 39543704 PMCID: PMC11566413 DOI: 10.1186/s12964-024-01918-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Endocrine therapy (ET) has improved the clinical outcomes of Estrogen receptor alpha-positive (ERɑ +) breast cancer (BC) patients, even though resistance to ET remains a clinical issue. Mutations in the hormone-binding domain of ERɑ represent an acquired intrinsic mechanism of ET resistance. However, the latter also depends on the multiple functional interactions between BC cells and the tumor microenvironment (TME). Here, we investigated how the most common Y537S-ERɑ mutation may influence the behavior of fibroblasts, the most prominent component of the TME. METHODS We conducted coculture experiments with normal human foreskin fibroblasts BJ1-hTERT (NFs), cancer-associated fibroblasts (CAFs), isolated from human BC specimens, and Y537S CRISPR-expressing MCF-7 BC cells (MCF-7YS). Mass spectrometry (MS) and Metacore analyses were performed to investigate how the functional interactions between BC cells/fibroblasts may affect their proteomic profile. The impact of fibroblasts on BC tumor growth and metastatic potential was evaluated in nude mice. RESULTS Mutant BC conditioned medium (CM) affected the morphology/proliferation/migration of both NFs and CAFs. 198 deregulated proteins signed the proteomic similarity profile of NFs exposed to the YS-CM and CAFs. Among the upregulated proteins, Yes-associated protein 1 (YAP1) was the main central hub in the direct interaction network. Increased YAP1 protein expression and activity were confirmed in NFs treated with MCF-7YS-CM. However, YAP1 activation appears to crosstalk with the insulin growth factor-1 receptor (IGF-1R). Higher amount of IGF-1 were noticed in the MCF-7YS-CM cells compared to the MCF-7P, and IGF-1 immunodepletion reversed the enhanced YAP1 expression and activity. Mutant cells upon exposure to the NF- and CAF-CM exhibited an enhanced proliferation/growth/migration/invasion compared to the MCF-7P. MCF-7YS cells when implanted with CAFs showed an early relative increased tumor volume compared to YS alone. No changes were observed when MCF-7P cells were co-implanted with CAFs. Compared with that in MCF-7P cells, the metastatic burden of MCF-7YS cells was intrinsically greater, and this effect was augmented upon treatment with NF-CM and further increased with CAF-CM. CONCLUSIONS YS mutant BC cells induced the conversion of fibroblasts into CAFs, via YAP, which represent a potential therapeutic target which interrupt the functional interactions between mutant cells/TME and to be implemented in the novel therapeutic strategy of a subset of metastatic BC patients carrying the frequent Y537S mutations.
Collapse
Affiliation(s)
- Luca Gelsomino
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Amanda Caruso
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Emine Tasan
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Adele Elisabetta Leonetti
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Rocco Malivindi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Clinical Laboratory Unit, A.O. "Annunziata", Cosenza, Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Francesca Giordano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Salvatore Panza
- Department of Experimental and Clinical Medicine, "Magna Graecia", University of Catanzaro, Catanzaro, Italy
| | - Guowei Gu
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Benedetta Perrone
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Clinical Laboratory Unit, A.O. "Annunziata", Cosenza, Italy
| | - Loredana Mauro
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Bruno Nardo
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Department of Surgery, General Surgery Unit, Annunziata Hospital, Cosenza, Italy
| | | | - Daniela Bonofiglio
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
| | - Suzanne A W Fuqua
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Stefania Catalano
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy
- Clinical Laboratory Unit, A.O. "Annunziata", Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy.
- Health Center, University of Calabria, Arcavacata Di Rende (CS), Cosenza, Italy.
| |
Collapse
|
28
|
Li W, Huang Y, Liu J, Zhou Y, Sun H, Fan Y, Liu F. Defective macrophage efferocytosis in advanced atherosclerotic plaque and mitochondrial therapy. Life Sci 2024; 359:123204. [PMID: 39491771 DOI: 10.1016/j.lfs.2024.123204] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease primarily affecting large and medium-sized arterial vessels, characterized by lipoprotein disorders, intimal thickening, smooth muscle cell proliferation, and the formation of vulnerable plaques. Macrophages (MΦs) play a vital role in the inflammatory response throughout all stages of atherosclerotic development and are considered significant therapeutic targets. In early lesions, macrophage efferocytosis rapidly eliminates harmful cells. However, impaired efferocytosis in advanced plaques perpetuates the inflammatory microenvironment of AS. Defective efferocytosis has emerged as a key factor in atherosclerotic pathogenesis and the progression to severe cardiovascular disease. Herein, this review probes into investigate the potential mechanisms at the cellular, molecular, and organelle levels underlying defective macrophage efferocytosis in advanced lesion plaques. In the inflammatory microenvironments of AS with interactions among diverse inflammatory immune cells, impaired macrophage efferocytosis is strongly linked to multiple factors, such as a lower absolute number of phagocytes, the aberrant expression of crucial molecules, and impaired mitochondrial energy provision in phagocytes. Thus, focusing on molecular targets to enhance macrophage efferocytosis or targeting mitochondrial therapy to restore macrophage metabolism homeostasis has emerged as a potential strategy to mitigate the progression of advanced atherosclerotic plaque, providing various treatment options.
Collapse
Affiliation(s)
- Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hongyu Sun
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yonghong Fan
- The General Hospital of Western Theater Command, Chengdu 610083, China.
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
29
|
Ding F, Zheng P, Yan XY, Chen HJ, Fang HT, Luo YY, Peng YX, Zhang L, Yan YE. Adipocyte-secreted PRELP promotes adipocyte differentiation and adipose tissue fibrosis by binding with p75 NTR to activate FAK/MAPK signaling. Int J Biol Macromol 2024; 279:135376. [PMID: 39244119 DOI: 10.1016/j.ijbiomac.2024.135376] [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: 05/03/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Adipocyte-secreted factors intricately regulate adipose tissue function, and the underlying molecular mechanisms are only partially understood. However, the function of PRELP, which is a key component of the extracellular matrix (ECM) in adipocytes, remains largely unknown. In this study, we demonstrate that PRELP was upregulated in both obese humans and mice, which exhibited a positive correlation with metabolic disorders. PRELP knockout could resist HFD-induced obesity and inhibit adipocyte differentiation. PRELP knockout improved glucose tolerance, insulin sensitivity and alleviated adipose tissue fibrosis. Mechanistically, PRELP was secreted into the ECM and bound to the extracellular domain of its receptor p75NTR in adipocytes, which further activated the FAK/MAPK (JNK, p38 MAPK, ERK1/2) signaling pathway, promoting adipocyte differentiation and exacerbating adipocyte fibrosis. Adipocyte PRELP plays a pivotal role in regulating obesity and adipose tissue fibrosis through an autocrine manner, and PRELP may be a therapeutic target for obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Fei Ding
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Peng Zheng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Xi-Yue Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hui-Jian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Hong-Ting Fang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yuan-Yuan Luo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Yu-Xuan Peng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Li Zhang
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - You-E Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China.
| |
Collapse
|
30
|
Gao Y, Wu M, Rizvi SAA, Wei Q. Exploring the key pathogenic mechanisms and potential intervention targets for Sophorae Flavescentis radix in managing bone metastasis of lung cancer based on network pharmacology and molecular docking techniques. Transl Cancer Res 2024; 13:5616-5626. [PMID: 39524998 PMCID: PMC11543056 DOI: 10.21037/tcr-24-1947] [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: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Background Lung cancer often metastasizes to the bone, which significantly complicates treatment and worsens patient prognosis. Thus, new therapeutic strategies need to be established. Using network pharmacology and bioinformatics analysis, this study sought to determine the molecular targets and associated mechanisms of the traditional Chinese medicine (TCM) Sophorae Flavescentis radix in the treatment of lung cancer bone metastasis. Methods The active components of Sophorae Flavescentis radix were screened using the TCM Systems Pharmacology (TCMSP) platform based on drug-likeness and oral bioavailability. The target genes of these active compounds were obtained from the DrugBank database. Differentially expressed genes (DEGs) between primary and bone metastatic lung cancer samples were screened in the GSE175601 dataset from the Gene Expression Omnibus (GEO) database using GEO2R. The intersecting DEGs from both groups were used to construct a Venn diagram to identify the candidate target genes. The expression and prognostic relevance of these genes were validated in The Cancer Genome Atlas (TCGA) database. The GeneMania and Search Tool for Recurring Instances of Neighbouring Genes (STRING) databases were used to generate the protein-protein interaction networks. Molecular docking was performed using the PubChem, Protein Data Bank (PDB), and CB-DOCK2 databases. A Gene Set Enrichment Analysis (GSEA) was conducted to explore the possible mechanisms of action. Results In the TCMSP database, 28 active compounds and 227 target genes of the Sophorae Flavescentis radix were identified. In total, 952 DEGs related to lung cancer bone metastasis were found in the GSE175601 dataset from the GEO database. Five common DEGs were identified via Venn diagram construction (i.e., F10, JUN, AKR1B1, MMP1, and CCND1). MMP1 was selected as the candidate gene. MMP1 was upregulated in lung cancer tissues, and patients with low MMP1 expression had better survival rates than those with high MMP1 expression (P<0.05). MMP1 has an affinity of -8.9 with luteolin. The GSEA results suggested that MMP1 might influence biological processes in lung cancer by participating in pathways such as chemokine signaling, apoptosis, Wingless/Integrated (Wnt) signaling, tumor protein p53-regulated cell cycle arrest, Hedgehog signaling, and mitogen-activated protein kinase signaling. Conclusions Patients with lower MMP1 levels had prolonged overall survival and may serve as a novel predictive biomarker for lung cancer. Sophorae Flavescentis radix appears to exert therapeutic effects on lung cancer bone metastasis by inhibiting MMP1 expression and modulating the abnormal activation of the Wnt pathway. Our findings further extend the understanding of the pathogenic mechanisms and potential therapeutic interventions of Sophorae Flavescentis radix in lung cancer bone metastasis, providing a theoretical basis for clinical diagnosis and treatment research.
Collapse
Affiliation(s)
- Yan Gao
- Department of Oncology, The First People’s Hospital of Lianyungang, Liangyungang, China
| | - Meng Wu
- Department of Oncology, The First People’s Hospital of Lianyungang, Liangyungang, China
| | - Syed A. A. Rizvi
- College of Biomedical Sciences, Larkin University, Miami, FL, USA
| | - Qiang Wei
- Department of Ultrasound, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Wang H, Li Q, Tang Q, Shi G, Wu G, Mao X, Wu C, Zhang L, Liu J, Li J, Li B. Role and therapeutic potential of E3s in the tumor microenvironment of hepatocellular carcinoma. Front Immunol 2024; 15:1483721. [PMID: 39544935 PMCID: PMC11560419 DOI: 10.3389/fimmu.2024.1483721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a high-incidence, poor-prognosis malignancy worldwide, requiring new strategies for treatment. Ubiquitination, especially ubiquitination through E3 ubiquitin ligases, plays an indispensable role in the development and progression of HCC. E3 ubiquitin ligases are crucial enzymes in ubiquitination, controlling the degradation of specific substrate proteins and influencing various cellular functions, such as tumor cell proliferation, apoptosis, migration, and immune evasion. In this review, we systematically summarize the mechanisms of E3 ubiquitin ligases in HCC, with a focus on the significance of RING, HECT, and RBR types in HCC progression. The review also looks at the potential for targeting E3 ligases to modulate the tumor microenvironment (TME) and increase immunotherapy efficacy. Future studies will optimize HCC treatment by formulating specific inhibitors or approaches that will be based on gene therapy targeting E3 ligases in order to overcome resistance issues with present treatments and create optimism in the journey of treatment for HCC patients.
Collapse
Affiliation(s)
- Hailin Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qiang Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Qinqin Tang
- Department of Dermatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Gang Shi
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guo Wu
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xingbo Mao
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Changkang Wu
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lixin Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jingdong Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
32
|
Di Marco T, Mazzoni M, Greco A, Cassinelli G. Non-oncogene dependencies: Novel opportunities for cancer therapy. Biochem Pharmacol 2024; 228:116254. [PMID: 38704100 DOI: 10.1016/j.bcp.2024.116254] [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: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Targeting oncogene addictions have changed the history of subsets of malignancies and continues to represent an excellent therapeutic opportunity. Nonetheless, alternative strategies are required to treat malignancies driven by undruggable oncogenes or loss of tumor suppressor genes and to overcome drug resistance also occurring in cancers addicted to actionable drivers. The discovery of non-oncogene addiction (NOA) uncovered novel therapeutically exploitable "Achilles' heels". NOA refers to genes/pathways not oncogenic per sé but essential for the tumor cell growth/survival while dispensable for normal cells. The clinical success of several classes of conventional and molecular targeted agents can be ascribed to their impact on both tumor cell-associated intrinsic as well as microenvironment-related extrinsic NOA. The integration of genetic, computational and pharmacological high-throughput approaches led to the identification of an expanded repertoire of synthetic lethality interactions implicating NOA targets. Only a few of them have been translated into the clinics as most NOA vulnerabilities are not easily druggable or appealing targets. Nonetheless, their identification has provided in-depth knowledge of tumor pathobiology and suggested novel therapeutic opportunities. Here, we summarize conceptual framework of intrinsic and extrinsic NOA providing exploitable vulnerabilities. Conventional and emerging methodological approaches used to disclose NOA dependencies are reported together with their limits. We illustrate NOA paradigmatic and peculiar examples and outline the functional/mechanistic aspects, potential druggability and translational interest. Finally, we comment on difficulties in exploiting the NOA-generated knowledge to develop novel therapeutic approaches to be translated into the clinics and to fully harness the potential of clinically available drugs.
Collapse
Affiliation(s)
- Tiziana Di Marco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Mara Mazzoni
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Angela Greco
- Integrated Biology of Rare Tumors Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy.
| |
Collapse
|
33
|
Cerbulescu T, Anghel A, Brie DA, Petraşcu FM, Salavat MC, Ardelean AI, Barac IR, Borugă O. The impact of matrix metalloproteinases and their tissue inhibitors in patients with chronic glaucoma - a literature review. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:557-565. [PMID: 39957016 PMCID: PMC11924901 DOI: 10.47162/rjme.65.4.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/29/2024] [Indexed: 02/18/2025]
Abstract
Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) play an important role in the pathophysiology of chronic glaucoma, as they are involved in extracellular matrix (ECM) remodeling in the trabecular meshwork (TM), affecting its ability to efficiently regulate intraocular pressure (IOP). Ensuring the balance between MMPs and TIMPs helps to maintain homeostasis in ocular tissues, which is essential to avoid glaucomatous lesions. Elevated levels of MMPs and increased degradation of the ECM, ultimately affecting aqueous humor outflow and increasing IOP, characterize glaucoma. In the current literature review, the impact and interactions of MMPs and TIMPs in chronic glaucoma have been emphasized, with multiple but still unelucidated roles in the mentioned pathology including their clinical implications, future research directions, and therapeutic approaches. Research to date indicates that the expression of TIMPs is altered in patients with chronic glaucoma, suggesting a compensatory response to increased MMPs activity. Certain drugs can influence the expression levels of MMPs and TIMPs, therefore therapeutic strategies can be developed to restore the balance between tissue enzymes and their inhibitors. Therefore, understanding the relationship between MMPs and TIMPs is a key factor in the pathogenesis of chronic glaucoma. Understanding the interplay between the two provides interesting insights into ECM remodeling in ocular tissues, highlighting the potential of targeted therapies to restore the balance between proteolytic enzymes and their inhibitors.
Collapse
Affiliation(s)
- Teodor Cerbulescu
- Department of Biochemistry, Faculty of Medicine, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania;
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Li F, Chen L, Xia Q, Feng Z, Li N. Combined knockdown of CD151 and MMP9 may inhibit the malignant biological behaviours of triple-negative breast cancer through the GSK-3β/β-catenin-related pathway. Sci Rep 2024; 14:21786. [PMID: 39294214 PMCID: PMC11411119 DOI: 10.1038/s41598-024-71533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
Triple-negative breast cancer (TNBC) represents a significant health concern for women worldwide, and the overproduction of MMP9 and CD151 is associated with various cancers, influencing tumour growth and progression. This study aimed to investigate how CD151 and MMP9 affect TNBC cell migration, apoptosis, proliferation, and invasion. Immunohistochemical experiments revealed that CD151 and MMP9 were positively expressed in triple-negative breast cancer, and lymph node metastasis, the histological grade, and CD151 and MMP9 expression were found to be independent prognostic factors for the survival of patients with triple-negative breast cancer. Cytological experiments indicated that the knockdown of CD151 or MMP9 slowed triple-negative breast cancer cell growth, migration, and invasion and increased the apoptosis rate. Compared with CD151 knockdown, double MMP9 and CD151 knockdown further promoted cell death and inhibited TNBC cell proliferation, migration, and invasion. Moreover, β-catenin and p-GSK-3β were significantly downregulated. In summary, simultaneously silencing CD151 and MMP9 further suppressed the proliferation, migration and invasion of TNBC cells and promoted their apoptosis. One possible strategy for inducing this effect is to block the GSK-3β/β-catenin pathway.
Collapse
Affiliation(s)
- Fan Li
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Liucheng Chen
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, Anhui, China
| | - Qing Xia
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Zhenzhong Feng
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Nan Li
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical University, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China.
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| |
Collapse
|
35
|
Wang D, Saleem S, Sullivan RD, Zhao T, Reed GL. Differences in Acute Expression of Matrix Metalloproteinases-9, 3, and 2 Related to the Duration of Brain Ischemia and Tissue Plasminogen Activator Treatment in Experimental Stroke. Int J Mol Sci 2024; 25:9442. [PMID: 39273389 PMCID: PMC11394866 DOI: 10.3390/ijms25179442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Matrix metalloproteinases (MMPs) such as MMP-9, 3, and 2 degrade the cellular matrix and are believed to play a crucial role in ischemic stroke. We examined how the duration of ischemia (up to 4 h) and treatment with recombinant tissue plasminogen activator altered the comparative expression of these MMPs in experimental ischemic stroke with reperfusion. Both prolonged ischemia and r-tPA treatment markedly increased MMP-9 expression in the ischemic hemisphere (all p < 0.0001). The duration of ischemia and r-tPA treatment also significantly increased MMP-2 expression (p < 0.01-0.001) in the ischemic hemisphere (p < 0.01) but to a lesser degree than MMP-9. In contrast, MMP-3 expression significantly decreased in the ischemic hemisphere (p < 0.001) with increasing duration of ischemia and r-tPA treatment (p < 0.05-0001). MMP-9 expression was prominent in the vascular compartment and leukocytes. MMP-2 expression was evident in the vascular compartment and MMP-3 in NeuN+ neurons. Prolonging the duration of ischemia (up to 4 h) before reperfusion increased brain hemorrhage, infarction, swelling, and neurologic disability in both saline-treated (control) and r-tPA-treated mice. MMP-9 and MMP-2 expression were significantly positively correlated with, and MMP-3 was significantly negatively correlated with, infarct volume, swelling, and brain hemorrhage. We conclude that in experimental ischemic stroke with reperfusion, the duration of ischemia and r-tPA treatment significantly altered MMP-9, 3, and 2 expression, ischemic brain injury, and neurological disability. Each MMP showed unique patterns of expression that are strongly correlated with the severity of brain infarction, swelling, and hemorrhage. In summary, in experimental ischemic stroke in male mice with reperfusion, the duration of ischemia, and r-tPA treatment significantly altered the immunofluorescent expression of MMP-9, 3, and 2, ischemic brain injury, and neurological disability. In this model, each MMP showed unique patterns of expression that were strongly correlated with the severity of brain infarction, swelling, and hemorrhage.
Collapse
Affiliation(s)
| | | | | | | | - Guy L. Reed
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ 85004, USA; (D.W.); (S.S.); (R.D.S.); (T.Z.)
| |
Collapse
|
36
|
Rodríguez-Cruz D, Boquet-Pujadas A, López-Muñoz E, Rincón-Heredia R, Paredes-Díaz R, Flores-Fortis M, Olivo-Marin JC, Guillén N, Aguilar-Rojas A. Three-dimensional cell culture conditions promoted the Mesenchymal-Amoeboid Transition in the Triple-Negative Breast Cancer cell line MDA-MB-231. Front Cell Dev Biol 2024; 12:1435708. [PMID: 39156975 PMCID: PMC11327030 DOI: 10.3389/fcell.2024.1435708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Breast cancer (BC) is the leading cause of death among women, primarily due to its potential for metastasis. As BC progresses, the extracellular matrix (ECM) produces more type-I collagen, resulting in increased stiffness. This alteration influences cellular behaviors such as migration, invasion, and metastasis. Specifically, cancer cells undergo changes in gene expression that initially promote an epithelial-to-mesenchymal transition (EMT) and subsequently, a transition from a mesenchymal to an amoeboid (MAT) migration mode. In this way, cancer cells can migrate more easily through the stiffer microenvironment. Despite their importance, understanding MATs remains challenging due to the difficulty of replicating in vitro the conditions for cell migration that are observed in vivo. Methods To address this challenge, we developed a three-dimensional (3D) growth system that replicates the different matrix properties observed during the progression of a breast tumor. We used this model to study the migration and invasion of the Triple-Negative BC (TNBC) cell line MDA-MB-231, which is particularly subject to metastasis. Results Our results indicate that denser collagen matrices present a reduction in porosity, collagen fiber size, and collagen fiber orientation, which are associated with the transition of cells to a rounder morphology with bleb-like protrusions. We quantified how this transition is associated with a more persistent migration, an enhanced invasion capacity, and a reduced secretion of matrix metalloproteinases. Discussion Our findings suggest that the proposed 3D growth conditions (especially those with high collagen concentrations) mimic key features of MATs, providing a new platform to study the physiology of migratory transitions and their role in BC progression.
Collapse
Affiliation(s)
- Daniela Rodríguez-Cruz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Aleix Boquet-Pujadas
- École Polytechnique Fédérale de Lausanne, Biomedical Imaging Group, Lausanne, Switzerland
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Eunice López-Muñoz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Rodolfo Paredes-Díaz
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Mauricio Flores-Fortis
- Cuajimalpa Unit, Engineering and Natural Science Doctoral Program, Metropolitan Autonomous University, Mexico City, Mexico
- Cuajimalpa Unit, Department of Natural Science, Metropolitan Autonomous University, Mexico City, Mexico
| | - Jean-Christophe Olivo-Marin
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Nancy Guillén
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS ERL9195, Paris, France
| | - Arturo Aguilar-Rojas
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
| |
Collapse
|
37
|
Ullah Z, Tao Y, Huang J. Integrated Bioinformatics-Based Identification and Validation of Neuroinflammation-Related Hub Genes in Primary Open-Angle Glaucoma. Int J Mol Sci 2024; 25:8193. [PMID: 39125762 PMCID: PMC11311784 DOI: 10.3390/ijms25158193] [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: 06/25/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Glaucoma is a leading cause of permanent blindness, affecting 80 million people worldwide. Recent studies have emphasized the importance of neuroinflammation in the early stages of glaucoma, involving immune and glial cells. To investigate this further, we used the GSE27276 dataset from the GEO (Gene Expression Omnibus) database and neuroinflammation genes from the GeneCards database to identify differentially expressed neuroinflammation-related genes associated with primary open-angle glaucoma (POAG). Subsequently, these genes were submitted to Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes for pathway enrichment analyses. Hub genes were picked out through protein-protein interaction networks and further validated using the external datasets (GSE13534 and GSE9944) and real-time PCR analysis. The gene-miRNA regulatory network, receiver operating characteristic (ROC) curve, genome-wide association study (GWAS), and regional expression analysis were performed to further validate the involvement of hub genes in glaucoma. A total of 179 differentially expressed genes were identified, comprising 60 upregulated and 119 downregulated genes. Among them, 18 differentially expressed neuroinflammation-related genes were found to overlap between the differentially expressed genes and neuroinflammation-related genes, with six genes (SERPINA3, LCN2, MMP3, S100A9, IL1RN, and HP) identified as potential hub genes. These genes were related to the IL-17 signaling pathway and tyrosine metabolism. The gene-miRNA regulatory network showed that these hub genes were regulated by 118 miRNAs. Notably, GWAS data analysis successfully identified significant single nucleotide polymorphisms (SNPs) corresponding to these six hub genes. ROC curve analysis indicated that our genes showed significant accuracy in POAG. The expression of these genes was further confirmed in microglia, Müller cells, astrocytes, and retinal ganglion cells in the Spectacle database. Moreover, three hub genes, SERPINA3, IL1R1, and LCN2, were validated as potential diagnostic biomarkers for high-risk glaucoma patients, showing increased expression in the OGD/R-induced glaucoma model. This study suggests that the identified hub genes may influence the development of POAG by regulation of neuroinflammation, and it may offer novel insights into the management of POAG.
Collapse
Affiliation(s)
| | | | - Jufang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, China; (Z.U.); (Y.T.)
| |
Collapse
|
38
|
Yang F, Yang K, Wang Y, Yao J, Hua X, Danso B, Wang Y, Liang H, Wang M, Chen J, Chen L, Xiao L, Zhang J. Insights into the discovery and intervention of metalloproteinase in marine hazardous jellyfish. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134526. [PMID: 38704908 DOI: 10.1016/j.jhazmat.2024.134526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
The proliferation of toxic organisms caused by changes in the marine environment, coupled with the rising human activities along the coastal lines, has resulted in an increasing number of stinging incidents, posing a serious threat to public health. Here, we evaluated the systemic toxicity of the venom in jellyfish Chrysaora quinquecirrha at both cellular and animal levels, and found that jellyfish tentacle extract (TE) has strong lethality accompanied by abnormal elevation of blood biochemical indicators and pathological changes. Joint analysis of transcriptome and proteome indicated that metalloproteinases are the predominant toxins in jellyfish. Specially, two key metalloproteinases DN6695_c0_g3 and DN8184_c0_g7 were identified by mass spectrometry of the red blood cell membrane and tetracycline hydrochloride (Tch) inhibition models. Structurally, molecular docking and kinetic analysis are employed and observed that Tch could inhibit the enzyme activity by binding to the hydrophobic pocket of the catalytic center. In this study, we demonstrated that Tch impedes the metalloproteinase activity thereby reducing the lethal effect of jellyfish, which suggests a potential strategy for combating the health threat of marine toxic jellyfish.
Collapse
Affiliation(s)
- Fengling Yang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Kai Yang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Yi Wang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Jinchi Yao
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; School of Life Sciences, Liaoning Normal University, Dalian 116081, China
| | - Xiaoyu Hua
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Blessing Danso
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Yongfang Wang
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Hongyu Liang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China; Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Mingke Wang
- Medical Care Center, Naval Medical Center of PLA, Naval Medical University, Shanghai 200052, China
| | - Jingbo Chen
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Lingxin Chen
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Research Center for Coastal Environmental Engineering and Technology, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| | - Liang Xiao
- Faculty of Naval Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Jing Zhang
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
39
|
Song WH, Lim YS, Kim JE, Kang HY, Lee C, Rajbongshi L, Hwang SY, Oh SO, Kim BS, Lee D, Song YJ, Yoon S. A Marine Collagen-Based 3D Scaffold for In Vitro Modeling of Human Prostate Cancer Niche and Anti-Cancer Therapeutic Discovery. Mar Drugs 2024; 22:295. [PMID: 39057404 PMCID: PMC11277582 DOI: 10.3390/md22070295] [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: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Recently, the need to develop a robust three-dimensional (3D) cell culture system that serves as a valuable in vitro tumor model has been emphasized. This system should closely mimic the tumor growth behaviors observed in vivo and replicate the key elements and characteristics of human tumors for the effective discovery and development of anti-tumor therapeutics. Therefore, in this study, we developed an effective 3D in vitro model of human prostate cancer (PC) using a marine collagen-based biomimetic 3D scaffold. The model displayed distinctive molecular profiles and cellular properties compared with those of the 2D PC cell culture. This was evidenced by (1) increased cell proliferation, migration, invasion, colony formation, and chemoresistance; (2) upregulated expression of crucial multidrug-resistance- and cancer-stemness-related genes; (3) heightened expression of key molecules associated with malignant progressions, such as epithelial-mesenchymal transition transcription factors, Notch, matrix metalloproteinases, and pluripotency biomarkers; (4) robust enrichment of prostate cancer stem cells (CSCs); and (5) enhanced expression of integrins. These results suggest that our 3D in vitro PC model has the potential to serve as a research platform for studying PC and prostate CSC biology, as well as for screening novel therapies targeting PC and prostate CSCs.
Collapse
Affiliation(s)
- Won Hoon Song
- Department of Urology, Pusan National University Yangsan Hospital and Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Ye Seon Lim
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Ji-Eun Kim
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Hae Yeong Kang
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Changyong Lee
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Lata Rajbongshi
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Seon Yeong Hwang
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea;
| | - Yong Jung Song
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital and Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea
| | - Sik Yoon
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| |
Collapse
|
40
|
Yin T, Chen S, Zhou R, Liu W, Diao M, Li L. Relationships of serum MMP-7 and clinical characteristics in choledochal cyst children. BMC Surg 2024; 24:195. [PMID: 38914992 PMCID: PMC11194885 DOI: 10.1186/s12893-024-02488-y] [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: 02/18/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Matrix metalloproteinase-7 (MMP-7) is associated with biliary injury. This study aimed to evaluate the relationships of serum MMP-7 with clinical characteristics in choledochal cysts (CDC) children. METHODS Between June 2020 and July 2022, we conducted a prospective study of CDCs who underwent one-stage definitive operation at our center. Serum MMP-7 was measured using an enzyme-linked immunosorbent assay. We evaluated the relationships between serum MMP-7 and age, laboratory tests, imaging examinations, liver fibrosis, MMP-7 expression, and perforation. RESULTS A total of 328 CDCs were enrolled in the study, with a median serum MMP-7 of 7.67 ng/mL. Higher serum MMP-7 was correlated with younger age at diagnosis (p < 0.001), larger cyst sizes (p < 0.001), higher liver fibrosis stages (p < 0.001), and higher incidence of perforation (p < 0.01). Liver MMP-7 was mainly expressed in intrahepatic and extrahepatic biliary epithelial cells. The area under the receiver operating characteristic curve (AUROC) was 0.630 (p < 0.001) for serum MMP-7 in predicting perforation. When serum MMP-7 was combined with γ-glutamyl transferase (GGT), the AUROC increased to 0.706 (p < 0.001). CONCLUSIONS Serum MMP-7 was associated with biliary obstruction in CDCs. Patients with high serum MMP-7 were more likely to have severe liver damage and biliary injury, with higher incidences of liver fibrosis and perforation.
Collapse
Affiliation(s)
- Tong Yin
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
- Department of General Surgery, Capital Institute of Pediatrics, Beijing, China
| | - Suyun Chen
- Department of Pediatric Urology, Fujian Children's Hospital, Fujian, China
| | - Ruijie Zhou
- Department of General Surgery, Capital Institute of Pediatrics, Beijing, China
| | - Wei Liu
- Department of General Surgery, Capital Institute of Pediatrics, Beijing, China
| | - Mei Diao
- Department of General Surgery, Capital Institute of Pediatrics, Beijing, China.
- Research Unit of Minimally Invasive Pediatric Surgery on Diagnosis and Treatment, Chinese Academy of Medical Sciences 2021RU015, Beijing, China.
| | - Long Li
- Department of General Surgery, Capital Institute of Pediatrics, Beijing, China.
- Research Unit of Minimally Invasive Pediatric Surgery on Diagnosis and Treatment, Chinese Academy of Medical Sciences 2021RU015, Beijing, China.
- Department of Pediatric Surgery, Tsinghua University Affiliated Beijing Tsinghua Changgung Hospital, Beijing, China.
| |
Collapse
|
41
|
Taheri E, Raeeszadeh-Sarmazdeh M. Effect of TIMPs and Their Minimally Engineered Variants in Blocking Invasion and Migration of Brain Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597644. [PMID: 38895489 PMCID: PMC11185677 DOI: 10.1101/2024.06.05.597644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Matrix metalloproteinases (MMPs) play a pivotal role in extracellular matrix (ECM) remodeling, influencing various aspects of cancer progression including migration, invasion, angiogenesis, and metastasis. Overexpression of MMPs, particularly MMP-2 and MMP-9, is notably pronounced in glioblastoma multiforme (GBM), a highly aggressive primary brain tumor characterized by diffuse and infiltrative behavior. Previous attempts to develop small molecule MMP inhibitors have failed in clinical trials, necessitating the exploration of more stable and selective alternatives. Tissue inhibitors of metalloproteinases (TIMPs), endogenous human proteins, offer promising potential due to their stability and broader interaction interfaces compared to small molecule inhibitors. In this study, we examined the effectiveness of wild-type human TIMP-1 and TIMP-3, alongside engineered minimal TIMP variants (mTC1 and mTC3), specifically designed for targeted MMP inhibition to reduce the migratory and invasive capabilities of GBM cells. Our investigation focused on these minimal TIMP variants, which provide enhanced tissue penetration and cellular uptake due to their small molecular weight, aiming to validate their potential as therapeutic agents. The results demonstrated that mTC1 and mTC3 effectively inhibit MMP activity, a critical factor in GBM aggressiveness, thereby highlighting their promise in controlling tumor spread. Given the lethality of GBM and the limited effectiveness of current treatments, the application of engineered TIMP variants represents a novel and potentially transformative therapeutic approach. By offering targeted MMP inhibition, these variants may significantly improve patient outcomes, providing new avenues for treatment and enhancing the survival and quality of life for patients with this devastating disease.
Collapse
|
42
|
Kalantar M, Kalanther I, Kumar S, Buxton EK, Raeeszadeh-Sarmazdeh M. Elucidating key determinants of engineered scFv antibody in MMP-9 binding using high throughput screening and machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597476. [PMID: 38895413 PMCID: PMC11185642 DOI: 10.1101/2024.06.04.597476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
An imbalance in matrix metalloproteinase-9 (MMP-9) regulation can lead to numerous diseases, including neurological disorders, cancer, and pre-term labor. Engineering single-chain antibody fragments (scFvs) Targeting MMP-9 to develop novel therapeutics for such diseases is desirable. We screened a synthetic scFv antibody library displayed on the yeast surface for binding improvement to MMP-9 using FACS (fluorescent-activated cell sorting). The scFv antibody clones isolated after FACS showed improvement in binding to MMP-9 compared to the endogenous inhibitor. To understand molecular determinants of binding between engineered scFv antibody variants and MMP-9, next-generation DNA sequencing, and computational protein structure analysis were used. Additionally, a deep-learning language model was trained on the synthetic library to predict the binding of scFv variants using their CDR-H3 sequences.
Collapse
|
43
|
Torelli FR, Rodrigues-Peres RM, Lopes-Cendes I, Bahamondes L, Juliato CRT. Gene expression associated with vaginal bleeding in women using the 52-mg levonorgestrel hormonal intrauterine device: A prospective study. Int J Gynaecol Obstet 2024; 165:1199-1209. [PMID: 38299835 DOI: 10.1002/ijgo.15357] [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: 05/05/2023] [Revised: 09/27/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024]
Abstract
OBJECTIVE To evaluate gene expression associated with vaginal bleeding in the 52-mg hormonal intrauterine device (IUD) users. MATERIALS AND METHODS We conducted a prospective study involving 100 women seeking to use the 52-mg hormonal IUD for contraception. We excluded women with a history or current condition of abnormal uterine bleeding and who were unable to attend a 1-year follow up. Women who expelled the device, removed it for reasons unrelated to vaginal bleeding, or were lost to follow up were discontinued. We collected endometrial biopsies immediately before IUD placement and assessed 20 selected genes using reverse transcription quantitative polymerase chain reaction. Users maintained a uterine bleeding diary for 12 months following IUD insertion. For statistical analysis, participants were categorized into groups with or without vaginal bleeding at 3 and 12 months. RESULTS Women with elevated CXCL9 expression had an 8.15-fold higher likelihood of experiencing vaginal bleeding at 3 months (odds ratio [OR] 8.15, 95% confidence interval [CI] 2.24-29.61, P = 0.001). At 12 months of follow up, women with increased TIMP1 expression had a 2.74-fold higher chance of experiencing vaginal bleeding (OR 2.74, 95% CI 1.08-6.95, P = 0.033). CXCL9 ≥ 1.5 and IL17A ≥ 0.68 were associated with a higher probability of vaginal bleeding at 3 months, while TIMP1 levels ≥0.943 were linked to an increased risk of bleeding at 12 months. CONCLUSION Users of the 52-mg hormonal IUD with elevated relative CXCL9 expression face an increased risk of vaginal bleeding at 3-month follow up, whereas those with heightened TIMP1 expression are more likely to experience vaginal bleeding at 12 months.
Collapse
Affiliation(s)
- Flávia R Torelli
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Raquel M Rodrigues-Peres
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Luis Bahamondes
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Cássia R T Juliato
- Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
44
|
Shimizu Y, Ntege EH, Inoue Y, Matsuura N, Sunami H, Sowa Y. Optimizing mesenchymal stem cell extracellular vesicles for chronic wound healing: Bioengineering, standardization, and safety. Regen Ther 2024; 26:260-274. [PMID: 38978963 PMCID: PMC11228664 DOI: 10.1016/j.reth.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Chronic wounds represent a significant global burden, afflicting millions with debilitating complications. Despite standard care, impaired healing persists due to factors like persistent inflammation and impaired tissue regeneration. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) offer an innovative regenerative medicine approach, delivering stem cell-derived therapeutic cargo in engineered nanoscale delivery systems. This review examines pioneering bioengineering strategies to engineer MSC-EVs into precision nanotherapeutics for chronic wounds. Emerging technologies like CRISPR gene editing, microfluidic manufacturing, and biomimetic delivery systems are highlighted for their potential to enhance MSC-EV targeting, optimize therapeutic cargo enrichment, and ensure consistent clinical-grade production. However, key hurdles remain, including batch variability, rigorous safety assessment for potential tumorigenicity, immunogenicity, and biodistribution profiling. Crucially, collaborative frameworks harmonizing regulatory science with bioengineering and patient advocacy hold the key to expediting global clinical translation. By overcoming these challenges, engineered MSC-EVs could catalyze a new era of off-the-shelf regenerative therapies, restoring hope and healing for millions afflicted by non-healing wounds.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| |
Collapse
|
45
|
Deng Z, Xu M, Ding Z, Kong J, Liu J, Zhang Z, Cao P. ID2 promotes tumor progression and metastasis in thyroid cancer. Endocrine 2024; 84:1051-1063. [PMID: 38195969 PMCID: PMC11208273 DOI: 10.1007/s12020-023-03674-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND Inhibitor of DNA Binding 2 (ID2) plays a crucial role in tumor cell proliferation, invasion, metastasis, and stemness. Aberrant ID2 expression is associated with poor prognosis in various cancers. However, the specific function of ID2 in thyroid cancer remain unclear. METHOD The TCGA database were utilized to explore the clinical relevance of ID2 in cancer. GO, KEGG, and TIMER were employed to predict the potential roles of ID2 in cancer. Functional analysis, including CCK-8, colony formation, transwell, wound healing, and sphere formation experiments, were conducted to determine the biological functions of ID2 in human cancers. Western blot (WB), RT-qPCR, and immunohistochemical (IHC) analyses were used to investigate the relationship between ID2 and downstream targets. RESULTS Our study revealed significant overexpression of ID2 in various malignant tumor cells. Knocking ID2 significantly inhibited cancer cell proliferation and invasion, while overexpressing ID2 enhanced these capabilities. Additionally, ID2 mediates resistance of cancer cells to protein kinase B (or Akt) inhibitions. Further WB and IHC experiments indicated that ID2 promotes the phosphorylation activation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, thereby upregulating the expression of downstream proliferation, epithelial-mesenchymal transition (EMT), and stemness-related markers. CONCLUSION We found that ID2 significantly promotes thyroid cancer cell proliferation, migration, EMT, and stemness through the PI3K/Akt pathway. Moreover, ID2 plays a crucial role in regulating cancer immune responses. It may serve as a potential biomarker for enhancing the efficacy of chemotherapy, targeted therapy, and immunotherapy against cancer.
Collapse
Affiliation(s)
- Zhongming Deng
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Min Xu
- Department of Anesthesiology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Zhenghua Ding
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Jianqiao Kong
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Juanjuan Liu
- Department of Anesthesiology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Zelin Zhang
- Department of Oncology Department, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| | - Ping Cao
- Department of Oncology Department, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| |
Collapse
|
46
|
Brown RB, Tozer DJ, Loubière L, Harshfield EL, Hong YT, Fryer TD, Williams GB, Graves MJ, Aigbirhio FI, O'Brien JT, Markus HS. MINocyclinE to Reduce inflammation and blood-brain barrier leakage in small Vessel diseAse (MINERVA): A phase II, randomized, double-blind, placebo-controlled experimental medicine trial. Alzheimers Dement 2024; 20:3852-3863. [PMID: 38629936 PMCID: PMC11180856 DOI: 10.1002/alz.13830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/13/2024] [Accepted: 03/16/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Cerebral small vessel disease (SVD) is a common cause of stroke/vascular dementia with few effective treatments. Neuroinflammation and increased blood-brain barrier (BBB) permeability may influence pathogenesis. In rodent models, minocycline reduced inflammation/BBB permeability. We determined whether minocycline had a similar effect in patients with SVD. METHODS MINERVA was a single-center, phase II, randomized, double-blind, placebo-controlled trial. Forty-four participants with moderate-to-severe SVD took minocycline or placebo for 3 months. Co-primary outcomes were microglial signal (determined using 11C-PK11195 positron emission tomography) and BBB permeability (using dynamic contrast-enhanced MRI). RESULTS Forty-four participants were recruited between September 2019 and June 2022. Minocycline had no effect on 11C-PK11195 binding (relative risk [RR] 1.01, 95% confidence interval [CI] 0.98-1.04), or BBB permeability (RR 0.97, 95% CI 0.91-1.03). Serum inflammatory markers were not affected. DISCUSSION 11C-PK11195 binding and increased BBB permeability are present in SVD; minocycline did not reduce either process. Whether these pathophysiological mechanisms are disease-causing remains unclear. INTERNATIONAL CLINICAL TRIALS REGISTRY PORTAL IDENTIFIER ISRCTN15483452 HIGHLIGHTS: We found focal areas of increased microglial signal and increased blood-brain barrier permeability in patients with small vessel disease. Minocycline treatment was not associated with a change in these processes measured using advanced neuroimaging. Blood-brain barrier permeability was dynamic but MRI-derived measurements correlated well with CSF/serum albumin ratio. Advanced neuroimaging is a feasible outcome measure for mechanistic clinical trials.
Collapse
Affiliation(s)
- Robin B. Brown
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Daniel J. Tozer
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | - Laurence Loubière
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| | | | - Young T. Hong
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Tim D. Fryer
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Guy B. Williams
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | - Martin J. Graves
- Department of RadiologyUniversity of CambridgeCambridgeCambridgeUK
| | - Franklin I. Aigbirhio
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUK
| | | | - Hugh S. Markus
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| |
Collapse
|
47
|
Vilardi A, Przyborski S, Mobbs C, Rufini A, Tufarelli C. Current understanding of the interplay between extracellular matrix remodelling and gut permeability in health and disease. Cell Death Discov 2024; 10:258. [PMID: 38802341 PMCID: PMC11130177 DOI: 10.1038/s41420-024-02015-1] [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: 01/18/2024] [Revised: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
The intestinal wall represents an interactive network regulated by the intestinal epithelium, extracellular matrix (ECM) and mesenchymal compartment. Under healthy physiological conditions, the epithelium undergoes constant renewal and forms an integral and selective barrier. Following damage, the healthy epithelium is restored via a series of signalling pathways that result in remodelling of the scaffolding tissue through finely-regulated proteolysis of the ECM by proteases such as matrix metalloproteinases (MMPs). However, chronic inflammation of the gastrointestinal tract, as occurs in Inflammatory Bowel Disease (IBD), is associated with prolonged disruption of the epithelial barrier and persistent damage to the intestinal mucosa. Increased barrier permeability exhibits distinctive signatures of inflammatory, immunological and ECM components, accompanied by increased ECM proteolytic activity. This narrative review aims to bring together the current knowledge of the interplay between gut barrier, immune and ECM features in health and disease, discussing the role of barrier permeability as a discriminant between homoeostasis and IBD.
Collapse
Affiliation(s)
- Aurora Vilardi
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Claire Mobbs
- Department of Biosciences, Durham University, Durham, DH1 3LE, United Kingdom
| | - Alessandro Rufini
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
- Department of Biosciences, University of Milan, Milan, 20133, Italy.
| | - Cristina Tufarelli
- Cancer Research Centre, University of Leicester, Leicester, LE2 7LX, United Kingdom.
| |
Collapse
|
48
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [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: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
49
|
Klimm W, Szamotulska K, Karwański M, Bartoszewicz Z, Witkowski W, Rozmyslowicz T, Niemczyk S. Tissue Inhibitors of Metalloproteinase 1 (TIMP-1) and 3 (TIMP-3) as New Markers of Acute Kidney Injury After Massive Burns. Med Sci Monit 2024; 30:e943500. [PMID: 38706186 PMCID: PMC11084814 DOI: 10.12659/msm.943500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/20/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common and serious complication after massive burn injury. One of the postulated etiologies is destruction of the extracellular matrix of nephrons, caused by a local imbalance between matrix metalloproteinases (MMPs) and specific inhibitors. The aim of this study was to analyze the dynamics of tissue inhibitors of metalloproteinases (TIMPs) during the first 5 days after massive thermal injury and the relationship with the risk of AKI. MATERIAL AND METHODS Thirty-three adults (22 men, 11 women) with severe burns were enrolled in the study. The values of TIMPs 1 to 4 were measured in blood serum and urine using the multiplex Luminex system. The associations between TIMPs and the risk of AKI were analyzed by using the generalized linear mixed models for repeated measurements. RESULTS Significant changes in serum and urine activities of TIMPs were confirmed, especially during the first 2 days after burn injury. Almost half of patients presented renal problems during the study. Significant differences between values of TIMPs in AKI and non-AKI status were also observed. However, a significant relationship between concentration of TIMPs and risk of AKI was confirmed only for urine TIMP-1 and serum TIMP-3. CONCLUSIONS The evaluation of TIMPs in the early stage after burn injury has potential benefits. The important roles of urine TIMP-1 and serum TIMP-3, as novel markers of the risk of AKI development, were confirmed. Other parameters require further analysis.
Collapse
Affiliation(s)
- Wojciech Klimm
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine – National Research Institute, Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Warsaw, Poland
| | - Marek Karwański
- Department of Applied Mathematics, University of Life Sciences, SGGW, Warsaw, Poland
| | - Zbigniew Bartoszewicz
- Department of Internal Diseases and Endocrinology, Medical University of Warsaw, Warsaw, Poland
| | - Wojciech Witkowski
- Department of Burns, Plastic and Reconstructive Surgery, Military Institute of Medicine – National Research Institute, Warsaw, Poland
| | - Tomasz Rozmyslowicz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine – National Research Institute, Warsaw, Poland
| |
Collapse
|
50
|
Alves R, Pires A, Jorge J, Balça-Silva J, Gonçalves AC, Sarmento-Ribeiro AB. Batimastat Induces Cytotoxic and Cytostatic Effects in In Vitro Models of Hematological Tumors. Int J Mol Sci 2024; 25:4554. [PMID: 38674139 PMCID: PMC11050270 DOI: 10.3390/ijms25084554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024] Open
Abstract
The role of metalloproteinases (MMPs) in hematological malignancies, like acute myeloid leukemia (AML), myelodysplastic neoplasms (MDS), and multiple myeloma (MM), is well-documented, and these pathologies remain with poor outcomes despite treatment advancements. In this study, we investigated the effects of batimastat (BB-94), an MMP inhibitor (MMPi), in single-administration and daily administration schemes in AML, MDS, and MM cell lines. We used four hematologic neoplasia cell lines: the HL-60 and NB-4 cells as AML models, the F36-P cells as an MDS model, and the H929 cells as a model of MM. We also tested batimastat toxicity in a normal human lymphocyte cell line (IMC cells). BB-94 decreases cell viability and density in a dose-, time-, administration-scheme-, and cell-line-dependent manner, with the AML cells displaying higher responses. The efficacy in inducing apoptosis and cell cycle arrests is dependent on the cell line (higher effects in AML cells), especially with lower daily doses, which may mitigate treatment toxicity. Furthermore, BB-94 activated apoptosis via caspases and ERK1/2 pathways. These findings highlight batimastat's therapeutic potential in hematological malignancies, with daily dosing emerging as a strategy to minimize adverse effects.
Collapse
Affiliation(s)
- Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Pires
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- HistologiX, BioCity, Innovation, Pennyfoot St., Nottingham NG1 1GF, UK
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Joana Balça-Silva
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- NOVA Medical School, New University of Lisbon, 1150-090 Lisbon, Portugal
| | - Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.P.); (J.J.); (J.B.-S.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Hematology Service, Centro Hospitalar Universitário de Coimbra, Unidade Local de Saúde de Coimbra, 3000-061 Coimbra, Portugal
| |
Collapse
|