1
|
Tian L, Cao M, Cheng H, Wang Y, He C, Shi X, Li T, Li Z. Plasmon-Stimulated Colorimetry Biosensor Array for the Identification of Multiple Metabolites. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6849-6858. [PMID: 38293917 DOI: 10.1021/acsami.3c16561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Rationally designing highly catalytic and stable nanozymes for metabolite monitoring is of great importance because of their huge potential in early disease diagnosis. Herein, a novel nanozyme based on hierarchically structured CuS/ZnS with a highly efficient peroxidase (POD)-mimic capability was developed and synthesized for multiple metabolite determination and recognition via the plasmon-stimulated biosensor array strategy. The designed nanozyme can simultaneously harvest plasmon triggered hot electron-hole pairs and generate photothermal properties, leading to a sharply boosted POD-mimic capability under 808 nm laser irradiation. Interestingly, because of the interaction diversity of the metabolite with POD-like nanomaterials, the unique inhibitory effect of metabolites on the POD-mimic activity could be the signal response as the differentiation. Thus, utilizing TMB as a typical chromogenic substrate in the addition of H2O2, the designed colorimetric biosensor array can produce diverse fingerprints for the three vital metabolisms (cysteine (Cys), ascorbic acid (AA), and glutathione (GSH)), which can be precisely identified by principal component analysis (PCA). Notably, a distinct fingerprint of a single metabolite with different levels and metabolite mixtures is also achieved with a detection limit of 1 μM. Most importantly, cell lysis could be effectively discriminated by the biosensor assay, implying its great potential in clinical diagnosis.
Collapse
Affiliation(s)
- Lin Tian
- School of Materials and Chemical Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
- School of Food (Biology) Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Ming Cao
- School of Materials and Chemical Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Haorong Cheng
- School of Materials and Chemical Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Yanfei Wang
- School of Materials and Chemical Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Changchun He
- School of Materials and Chemical Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Xinxin Shi
- School of Food (Biology) Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Tongxiang Li
- School of Food (Biology) Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| | - Zhao Li
- School of Materials and Chemical Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
- School of Food (Biology) Engineering, Xuzhou University of Technology, Xuzhou 221018, PR China
| |
Collapse
|
2
|
Desprez C, Danovi D, Knowles CH, Day RM. Cell shape characteristics of human skeletal muscle cells as a predictor of myogenic competency: A new paradigm towards precision cell therapy. J Tissue Eng 2023; 14:20417314221139794. [PMID: 36949843 PMCID: PMC10026113 DOI: 10.1177/20417314221139794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/02/2022] [Indexed: 03/18/2023] Open
Abstract
Skeletal muscle-derived cells (SMDC) hold tremendous potential for replenishing dysfunctional muscle lost due to disease or trauma. Current therapeutic usage of SMDC relies on harvesting autologous cells from muscle biopsies that are subsequently expanded in vitro before re-implantation into the patient. Heterogeneity can arise from multiple factors including quality of the starting biopsy, age and comorbidity affecting the processed SMDC. Quality attributes intended for clinical use often focus on minimum levels of myogenic cell marker expression. Such approaches do not evaluate the likelihood of SMDC to differentiate and form myofibres when implanted in vivo, which ultimately determines the likelihood of muscle regeneration. Predicting the therapeutic potency of SMDC in vitro prior to implantation is key to developing successful therapeutics in regenerative medicine and reducing implementation costs. Here, we report on the development of a novel SMDC profiling tool to examine populations of cells in vitro derived from different donors. We developed an image-based pipeline to quantify morphological features and extracted cell shape descriptors. We investigated whether these could predict heterogeneity in the formation of myotubes and correlate with the myogenic fusion index. Several of the early cell shape characteristics were found to negatively correlate with the fusion index. These included total area occupied by cells, area shape, bounding box area, compactness, equivalent diameter, minimum ferret diameter, minor axis length and perimeter of SMDC at 24 h after initiating culture. The information extracted with our approach indicates live cell imaging can detect a range of cell phenotypes based on cell-shape alone and preserving cell integrity could be used to predict propensity to form myotubes in vitro and functional tissue in vivo.
Collapse
Affiliation(s)
- Charlotte Desprez
- Centre for Precision Healthcare, UCL
Division of Medicine, University College London, London, UK
- Department of Digestive Physiology,
Rouen University Hospital, Rouen, France
- On behalf of the EC Horizon 2020 AMELIE
consortium. Details of the AMELIE consortium is provided in the
Acknowledgements
| | - Davide Danovi
- Centre for Gene Therapy and
Regenerative Medicine, King’s College London, London, UK
- bit.bio, The Dorithy Hodgkin Building,
Babraham Research Campus, Cambridge
| | - Charles H Knowles
- On behalf of the EC Horizon 2020 AMELIE
consortium. Details of the AMELIE consortium is provided in the
Acknowledgements
- Blizard Institute, Centre for
Neuroscience, Surgery & Trauma, Queen Mary University of London, London,
UK
| | - Richard M Day
- Centre for Precision Healthcare, UCL
Division of Medicine, University College London, London, UK
- On behalf of the EC Horizon 2020 AMELIE
consortium. Details of the AMELIE consortium is provided in the
Acknowledgements
- Richard M Day, Centre for Precision
Healthcare, UCL Division of Medicine, University College London, Gower Street,
London WC1E 6JJ, UK.
| |
Collapse
|
3
|
An integral perspective of canonical cigarette and e-cigarette-related cardiovascular toxicity based on the adverse outcome pathway framework. J Adv Res 2022:S2090-1232(22)00193-X. [PMID: 35998874 DOI: 10.1016/j.jare.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Nowadays, cigarette smoking remains the leading cause of chronic disease and premature death, especially cardiovascular disease. As an emerging tobacco product, e-cigarettes have been advocated as alternatives to canonical cigarettes, and thus may be an aid to promote smoking cessation. However, recent studies indicated that e-cigarettes should not be completely harmless to the cardiovascular system. AIM OF REVIEW This review aimed to build up an integral perspective of cigarettes and e-cigarettes-related cardiovascular toxicity. KEY SCIENTIFIC CONCEPTS OF REVIEW This review adopted the adverse outcome pathway (AOP) framework as a pivotal tool and aimed to elucidate the association between the molecular initiating events (MIEs) induced by cigarette and e-cigarette exposure to the cardiovascular adverse outcome. Since the excessive generation of reactive oxygen species (ROS) has been widely approved to play a critical role in cigarette smoke-related CVD and may also be involved in e-cigarette-induced toxic effects, the ROS overproduction and subsequent oxidative stress are regarded as essential parts of this framework. As far as we know, this should be the first AOP framework focusing on cigarette and e-cigarette-related cardiovascular toxicity, and we hope our work to be a guide in exploring the biomarkers and novel therapies for cardiovascular injury.
Collapse
|
4
|
Rath S, Chakraborty D, Pradhan J, Imran Khan M, Dandapat J. Epigenomic interplay in tumor heterogeneity: Potential of epidrugs as adjunct therapy. Cytokine 2022; 157:155967. [PMID: 35905624 DOI: 10.1016/j.cyto.2022.155967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 11/28/2022]
Abstract
"Heterogeneity" in tumor mass has immense importance in cancer progression and therapy. The impact of tumor heterogeneity is an emerging field and not yet fully explored. Tumor heterogeneity is mainly considered as intra-tumor heterogeneity and inter-tumor heterogeneity based on their origin. Intra-tumor heterogeneity refers to the discrepancy within the same cancer mass while inter-tumor heterogeneity refers to the discrepancy between different patients having the same tumor type. Both of these heterogeneity types lead to variation in the histopathological as well as clinical properties of the cancer mass which drives disease resistance towards therapeutic approaches. Cancer stem cells (CSCs) act as pinnacle progenitors for heterogeneity development along with various other genetic and epigenetic parameters that are regulating this process. In recent times epigenetic factors are one of the most studied parameters that drive oxidative stress pathways essential during cancer progression. These epigenetic changes are modulated by various epidrugs and have an impact on tumor heterogeneity. The present review summarizes various aspects of epigenetic regulation in the tumor microenvironment, oxidative stress, and progression towards tumor heterogeneity that creates complications during cancer treatment. This review also explores the possible role of epidrugs in regulating tumor heterogeneity and personalized therapy against drug resistance.
Collapse
Affiliation(s)
- Suvasmita Rath
- Center of Environment, Climate Change and Public Health, Utkal University, Vani Vihar, Bhubaneswar 751004, Odisha, India
| | - Diptesh Chakraborty
- Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Jyotsnarani Pradhan
- Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Mohammad Imran Khan
- Department of Biochemistry, King Abdulaziz University (KAU), Jeddah 21577, Saudi Arabia; Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jagneshwar Dandapat
- Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India; Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India.
| |
Collapse
|
5
|
Liao K, Lv DY, Yu HL, Chen H, Luo SX. iNOS regulates activation of the NLRP3 inflammasome through the sGC/cGMP/PKG/TACE/TNF-α axis in response to cigarette smoke resulting in aortic endothelial pyroptosis and vascular dysfunction. Int Immunopharmacol 2021; 101:108334. [PMID: 34768128 DOI: 10.1016/j.intimp.2021.108334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cigarette smoke (CS) is associated with vascular injury and dysfunction, which may be mediated by iNOS and NLRP3. However, the exact mechanism is unknown. METHODS iNOS-knockout and NLRP3-knockout C57BL/6 mice were exposed to air or CS. The vascular structure was examined by hematoxylin-eosin staining. The vascular tension was measured by a vascular reactivity assay. The expression of iNOS, NLRP3, caspase-1p20, IL-1β and eNOS were measured by western blotting. Human aortic endothelial cells (HAECs) were exposed to L-NIL (iNOS inhibitor), MCC950 (NLRP3 inhibitor), ODQ (sGC inhibitor), KT5823 (PKG inhibitor) or TAPI-1 (TACE/ADAM17 inhibitor) for 1 h prior to cigarette smoke extract (CSE) treatment. The cell viability and lactate dehydrogenase activity were assessed and pyroptosis was determined by scanning electron microscopy. The mRNA expression of TNF-α, and protein expression of iNOS, active-TACE, NLRP3, caspase-1p20, IL-1β, and eNOS were measured. RESULTS CS resulted in shrinkage of endothelial cells, impaired aorta relaxation, reduced eNOS expression, and induced expression of iNOS, NLRP3, caspase-1p20 and IL-1β, which could be prevented by knockdown of iNOS and NLRP3. CSE reduced cell viability, induced LDH release and pyroptosis, and promoted iNOS, NLRP3, caspase-1p20, and IL-1β expression and reduced eNOS reduction, which could be reversed by inhibition of iNOS or NLRP3 in HAECs. Altogether, activation of the NLRP3 inflammasome by iNOS in CS-exposed HAECs may be mediated by the sGC/cGMP/PKG/TACE/TNF- α pathway. CONCLUSION These results link iNOS to NLRP3 in CSE-stimulated HAECs through the sGC/cGMP/PKG/TACE/TNF-α pathway. The findings identify a mechanism through which iNOS and NLRP3 contribute to the pathogenesis of CS-induced pyroptosis and impaired aorta relaxation in HAECs.
Collapse
Affiliation(s)
- Ke Liao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Ding-Yi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hui-Lin Yu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China
| | - Hong Chen
- Department of Respiratory, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| | - Su-Xin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China.
| |
Collapse
|
6
|
Abstract
Purpose: Nicotine causes tendon degeneration, whereas ascorbic acid imparts beneficial effects on tendon cells. Tendon stem cells (TSCs) play a vital role in maintaining tissue integrity and promoting restoration of structure and function after tendon injury. In the present study, cell culture experiments were performed to determine the effects of nicotine on TSCs and whether ascorbic acid supplementation could antagonize the action of high concentration nicotine. Methods: After treatment with nicotine and ascorbic acid, TSC proliferation, migration, stemness, apoptosis, and differentiation were analyzed. Results: TSC proliferation and expression of stem cell markers were significantly impaired by a high concentration of nicotine (1000 ng/mL), but a lower concentration (100 ng/mL) induced proliferative effects in TSCs. Moreover, the highest concentration of nicotine tested (1000 ng/mL) significantly inhibited the migratory ability of TSCs, while relatively high concentrations (100 and 1000 ng/mL) significantly (p < 0.05) up-regulated non-tenocyte genes. When ascorbic acid was added, the inhibitory effects of nicotine on the proliferation, migration, and stemness of TSCs were reversed. In addition, flow cytometry analysis showed that these nicotine concentrations could induce cell apoptosis, while the addition of ascorbic acid inhibited apoptosis. Conclusion: Addition of ascorbic acid partially reversed the inhibitory effect of a high concentration of nicotine. These findings indicate that while nicotine impairs the biological characteristics of TSCs, ascorbic acid can mitigate these deleterious effects and, therefore, may be useful for decreasing nicotine-induced tendon degeneration.
Collapse
Affiliation(s)
- Zhengzhou Shi
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Qi Wang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| | - Dapeng Jiang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| |
Collapse
|
7
|
Li X, Ding D, Chen W, Liu Y, Pan H, Hu J. Growth differentiation factor 11 mitigates cardiac radiotoxicity via activating AMPKα. Free Radic Res 2021; 55:176-185. [PMID: 33557626 DOI: 10.1080/10715762.2021.1885653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac radiotoxicity largely impedes the therapeutic benefits of radiotherapy to malignancies. Growth differentiation factor 11 (GDF11) is implicated in the pathogenesis of cardiac diseases under different pathological conditions. This study aims to investigate the role and underlying mechanisms of GDF11 on cardiac radiotoxicity. Mice were injected with cardiotropic adeno-associated virus 9 carrying the full-length mouse GDF11 gene or negative control under a cTnT promoter from the tail vein, and then received a single dose of 20 Gray (Gy) whole-heart irradiation (WHI) for 16 weeks to imitate cardiac radiotoxicity. Compound C (CC, 20 mg/kg) was intraperitoneally injected every two days at 1 week before WHI stimulation to inhibit 5' AMP-activated protein kinase α (AMPKα). Cardiac GDF11 expression was significantly suppressed at both the protein and mRNA levels. GDF11 overexpression decreased oxidative stress, apoptosis, and fibrosis in radiated hearts, thereby mitigating cardiac radiotoxicity, and dysfunction. Further detection revealed that GDF11 activated AMPKα to reduce radiation-induced oxidative damage and that AMPKα inhibition by CC offset the cardioprotective effects by GDF11. GDF11 mitigates cardiac radiotoxicity via activating AMPKα and it is a promising candidate to treat cardiac radiotoxicity.
Collapse
Affiliation(s)
- Xia Li
- Department of Ultrasound Imaging, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Dong Ding
- Department of Radiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Wei Chen
- Department of Ultrasound Imaging, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Yu Liu
- Department of Radiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Haisong Pan
- Department of Radiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
| | - Jun Hu
- Department of Radiology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, PR China
| |
Collapse
|
8
|
Caliri AW, Tommasi S, Besaratinia A. Relationships among smoking, oxidative stress, inflammation, macromolecular damage, and cancer. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 787:108365. [PMID: 34083039 PMCID: PMC8287787 DOI: 10.1016/j.mrrev.2021.108365] [Citation(s) in RCA: 231] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Smoking is a major risk factor for a variety of diseases, including cancer and immune-mediated inflammatory diseases. Tobacco smoke contains a mixture of chemicals, including a host of reactive oxygen- and nitrogen species (ROS and RNS), among others, that can damage cellular and sub-cellular targets, such as lipids, proteins, and nucleic acids. A growing body of evidence supports a key role for smoking-induced ROS and the resulting oxidative stress in inflammation and carcinogenesis. This comprehensive and up-to-date review covers four interrelated topics, including 'smoking', 'oxidative stress', 'inflammation', and 'cancer'. The review discusses each of the four topics, while exploring the intersections among the topics by highlighting the macromolecular damage attributable to ROS. Specifically, oxidative damage to macromolecular targets, such as lipid peroxidation, post-translational modification of proteins, and DNA adduction, as well as enzymatic and non-enzymatic antioxidant defense mechanisms, and the multi-faceted repair pathways of oxidized lesions are described. Also discussed are the biological consequences of oxidative damage to macromolecules if they evade the defense mechanisms and/or are not repaired properly or in time. Emphasis is placed on the genetic- and epigenetic alterations that may lead to transcriptional deregulation of functionally-important genes and disruption of regulatory elements. Smoking-associated oxidative stress also activates the inflammatory response pathway, which triggers a cascade of events of which ROS production is an initial yet indispensable step. The release of ROS at the site of damage and inflammation helps combat foreign pathogens and restores the injured tissue, while simultaneously increasing the burden of oxidative stress. This creates a vicious cycle in which smoking-related oxidative stress causes inflammation, which in turn, results in further generation of ROS, and potentially increased oxidative damage to macromolecular targets that may lead to cancer initiation and/or progression.
Collapse
Affiliation(s)
- Andrew W Caliri
- Department of Preventive Medicine, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, USA
| | - Stella Tommasi
- Department of Preventive Medicine, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, USA
| | - Ahmad Besaratinia
- Department of Preventive Medicine, USC Keck School of Medicine, University of Southern California, M/C 9603, Los Angeles, CA 90033, USA.
| |
Collapse
|