1
|
Khanishayan A, Rahgozar S, Zebardast D. The significance of the activating transcription factor 6 gene in the pathogenesis of drug resistant cancer. Tissue Cell 2025; 93:102786. [PMID: 39946967 DOI: 10.1016/j.tice.2025.102786] [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: 11/15/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025]
Abstract
Chemotherapy remains a cornerstone in cancer therapy, but its effectiveness is often hindered by the development of drug resistance, a significant factor contributing to over 90 % of cancer-related deaths worldwide. A critical aspect of this resistance involves chronic endoplasmic reticulum stress, which activates the unfolded protein response (UPR), mainly through the activation of Activating Transcription Factor 6 (ATF6). Elevated ATF6 expression has been found to correlate with poor survival outcomes and increased resistance to chemotherapy across several malignancies. This study specifically investigates the role of ATF6 in cancer pathogenesis, focusing on its involvement in resistance mechanisms and the progression of the disease. Given the complex interactions between the UPR and other cellular pathways, including the DNA damage response (DDR), our findings emphasize the potential of targeting ATF6 and UPR and DDR pathways as a novel therapeutic strategy. This approach could potentially overcome chemoresistance and improve outcomes in cancer treatment.
Collapse
Affiliation(s)
- Ashkan Khanishayan
- University of Isfahan, Isfahan, Iran, and University of Arizona, United States
| | | | | |
Collapse
|
2
|
Aschner M, Skalny AV, Martins AC, Tizabi Y, Zaitseva IP, Santamaria A, Lu R, Gluhcheva YY, Tinkov AA. The role of NLRP3 inflammasome activation in proinflammatory and cytotoxic effects of metal nanoparticles. Arch Toxicol 2025; 99:1287-1314. [PMID: 39960653 DOI: 10.1007/s00204-025-03972-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/27/2025] [Indexed: 04/04/2025]
Abstract
Exposure to metal nanoparticles (NPs) is known to induce inflammatory responses in various tissues, thus limiting their therapeutic potential. NOD-like receptor protein 3 (NLRP3) inflammasome activation is an essential component of innate immunity playing a significant role in inflammation and development of inflammatory diseases. Therefore, the objective of the present review was to summarize data on the role of NLRP3 inflammasome in proinflammatory effects induced by metal NPs, and to discuss the underlying molecular mechanisms, including its dependence on the physical and chemical properties of metal NPs. Titanium, zinc, silver, aluminum, iron, cobalt, nickel, vanadium, and tungsten nanoparticles, as well as metal-based quantum dots have all been shown to induce NLRP3 inflammasome activation in vitro in macrophages and monocytes, dendritic cells, keratinocytes, hepatocytes, enterocytes, microglia, astrocytes, lung epithelial cells, endotheliocytes, as well as certain types of cancer cells. In vivo studies confirmed the role of NLRP3 pathway activation in development of colitis, pulmonary inflammation, liver damage, osteolysis, and neuroinflammation induced by various metal nanoparticles. Briefly, particle endocytosis with subsequent lysosomal damage, induction of ROS formation, K+ efflux, increased intracellular Ca2+ levels, and NF-κB pathway activation results in NLRP3 inflammasome complex assembly, caspase-1 activation, and cleavage of pro-IL-1β and pro-IL-18 to mature proinflammatory cytokines, while gasdermin D cleavage induces pyroptotic cell death. Moreover, small-sized and rod-shaped metal NPs exert a more profound stimulatory effect on NLRP3 inflammasome activation, but contrary findings have also been reported. Taken together, it is concluded that NLRP3 inflammasome may mediate both adverse proinflammatory effects of metal nanoparticles, as well as their beneficial effect when used as antitumor agents.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Anatoly V Skalny
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Irina P Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control and Department of Physical Education, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yordanka Y Gluhcheva
- Institute of Experimental Morphology, Pathology and Anthropology With Museum, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str., Bl. 25, 1113, Sofia, Bulgaria
| | - Alexey A Tinkov
- Institute of Bioelementology, Orenburg State University, Orenburg, 460018, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119146, Russia
- Laboratory of Ecobiomonitoring and Quality Control and Department of Physical Education, Yaroslavl State University, Yaroslavl, 150003, Russia
| |
Collapse
|
3
|
Zhu D, Li Y, Liu M, Yang Y, Fu J, Su L, Wang F, Cen Y, Zhou Y, Li Y. Study on the role of mitophagy and pyroptosis induced by nano-silver in testicular injury. Food Chem Toxicol 2025; 197:115245. [PMID: 39788479 DOI: 10.1016/j.fct.2025.115245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Silver nanoparticles(AgNPs)have been widely used in biomedicine and industry. Growing studies have shown that AgNPs can induce sperm motility decrease and spermiogenesis disorders. In this study, animal experiments were used to investigate the role of mitophagy and pyroptosis caused by AgNPs (25.93 nm) in testicular injury. Results showed that AgNPs induced the production of NLRP3, IL-1β and IL-18, activated Caspase-1, increased the expression of GSDMD protein, and activated the PINK1/Parkin signaling pathway, which induced in mitophagy in mice testicle tissue. In summary, AgNPs induced mitophagy and pyroptosis in mice testis at the highest dose, which lead to damage of testis tissue.
Collapse
Affiliation(s)
- Deyu Zhu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China; Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, Guizhou, 563000, PR China
| | - Yingyi Li
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Min Liu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yufen Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Jiayu Fu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Liyu Su
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Feng Wang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yuyan Cen
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yanna Zhou
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China
| | - Yan Li
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, 563000, PR China; Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, Guizhou, 563000, PR China
| |
Collapse
|
4
|
Rana SVS. Mechanistic paradigms of immunotoxicity, triggered by nanoparticles - a review. Toxicol Mech Methods 2025; 35:262-278. [PMID: 39585654 DOI: 10.1080/15376516.2024.2431687] [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: 07/22/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Nanoparticles (NPs) possess the ability to penetrate cells and elicit a rapid and targeted immune response, influenced by their distinct physicochemical properties. These particles can engage with both micro and macromolecules, thereby impacting various downstream signaling pathways that may lead to cell death. This review provides a comprehensive overview of the primary mechanisms contributing to the immunotoxicity of both organic and inorganic nanoparticles. The effects of carbon-based nanomaterials (CNMs), including single-walled carbon nanotubes, multi-walled carbon nanotubes, graphene, and metal oxide nanoparticles, on various immune cell types such as macrophages, neutrophils, monocytes, dendritic cells (DCs), antigen-presenting cells (APCs), and RAW 264.7 cells are examined. The immune responses discussed encompass inflammation, oxidative stress, autophagy, and apoptosis. Additionally, the roles of pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α, and IFN-γ, along with JAK/STAT signaling pathways, are highlighted. The interaction of NPs with oxidative stress pathways, including MAPK signaling and Nrf2/ARE signaling, is also explored. Furthermore, the mechanisms by which nanoparticles induce damage to organelles such as lysosomes, the endoplasmic reticulum, exosomes, and Golgi bodies within the immune system are addressed. The review also emphasizes the genotoxic and epigenetic mechanisms associated with the immunotoxicity of NPs. Recent advancements regarding the immunotherapeutic potential of engineered NPs are reported. The roles of autophagy and apoptosis in the immunotoxicity of NPs merit further investigation. In conclusion, understanding how engineered nanoparticles modulate immune responses may facilitate the prevention and treatment of human diseases, including cancer and autoimmune disorders.
Collapse
Affiliation(s)
- S V S Rana
- Department of Toxicology, Chaudhary Charan Singh University, Meerut, India
| |
Collapse
|
5
|
Kuc-Ciepluch D, Ciepluch K, Augustyniak D, Guła G, Maciejewska B, Kowalik A, Jop E, Drulis-Kawa Z, Arabski M. Exploiting gasdermin-mediated pyroptosis for enhanced antimicrobial activity of phage endolysin against Pseudomonas aeruginosa. mSystems 2025; 10:e0110624. [PMID: 39714210 PMCID: PMC11748493 DOI: 10.1128/msystems.01106-24] [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: 08/14/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024] Open
Abstract
Pyroptosis is an inflammatory immune response of eukaryotic cells to bacterial lipopolysaccharide (LPS) and other pathological stimuli, leading to the activation of the gasdermin D (GSDMD) and secretion of pore-forming domain GSDMDNterm, facilitating the release of cytokines. Additionally, GSDMDNterm exhibits antibacterial properties through interactions with bacterial outer membranes (OM). We explored alternative antimicrobial strategy to determine whether inducing natural pyroptosis via GSDMD activation by P. aeruginosa LPS could enhance the effectiveness of recombinant phage endopeptidase KP27 (peptidoglycan-degrading enzyme) against P. aeruginosa, enabling penetration through OM and bacterial killing synergistically. Our findings demonstrated that recombinant GSDMD alone exhibited antibacterial effects against wild-type P. aeruginosa with smooth LPS, while recombinant GSDMDNterm efficiently permeabilized both smooth LPS-bearing and O-chain-deficient P. aeruginosa potentially synergizing with endolysin KP27. Transcriptomic analyses revealed the activation of the immune system pathways in response to LPS, mainly in monocytic cells, in contrast to epithelial A549 or HeLa cell lines. LPS-induced pyroptosis in monocytes led to GSDMD cleavage and the release of interleukins, regardless of the nature/origin of the LPS used. However, the pyroptosis stimulation by LPS in the antibacterial assay was not effective enough for bacterial OM permeabilization and enhancement of endolysin activity. We assume that leveraging pyroptosis induction in monocytic cells to augment the bactericidal activity of endolysins may be limited. IMPORTANCE Recombinant GSDMDNterm protein was able to efficiently permeabilize P. aeruginosa outer membranes and increase endolysin activity against bacteria, producing either long LPS O-chains or lack them entirely. The obtained results suggest the limited possibility of using the natural process of pyroptosis occurring in monocytic cells to enhance the bactericidal effect of recombinant phage endolysins against Gram-negative bacteria infection.
Collapse
Affiliation(s)
- Dorota Kuc-Ciepluch
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Karol Ciepluch
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Daria Augustyniak
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Grzegorz Guła
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Barbara Maciejewska
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Artur Kowalik
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland
| | - Ewelina Jop
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, University of Wroclaw, Wroclaw, Poland
| | - Michał Arabski
- Division of Medical Biology, Jan Kochanowski University in Kielce, Kielce, Poland
| |
Collapse
|
6
|
Fu W, Liu M, Wang Y, Yang H, Ye A, Wu J, Li Y, Yu Z, Qiu Y, Xu L. Nano titanium dioxide induces HaCaT cell pyroptosis via regulating the NLRP3/caspase-1/GSDMD pathway. Toxicol Lett 2024; 402:27-37. [PMID: 39547316 DOI: 10.1016/j.toxlet.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Nano-titanium dioxide (Nano-TiO2) is extensively utilized across various industries and has the capacity to penetrate human tissues through multiple biological barriers. The HaCaT cell line, as one of human immortalized keratinocytes, is usually used as a model for studying skin drug toxicology. The objective was to assess the toxic effects of nano-TiO2 on HaCaT cells and to trigger pyroptosis. We used MTT method to evaluate the effects of three nano-TiO2 particle sizes (15 nm, 30 nm and 80 nm) on cell viability at different concentrations. Subsequently, we used LDH, Hoechst 33342 and propidium iodide (PI) double staining, scanning electron microscopy (SEM), Western blotting (WB) and real-time quantitative polymerase chain reaction (RT-qPCR) to evaluate the effects of different particle sizes on cells at the same concentration. Our findings indicated that HaCaT cell viability diminished with increasing nano-TiO2 concentrations. Moreover, nano-TiO2 increased LDH level in cellular supernatant. Fluorescence double staining, SEM, WB and RT-qPCR showed that nano-TiO2 induced cell membrane damage by activating pyroptosis pathway of NLRP3/caspase-1/GSDMD. These results suggest that nano-TiO2 toxicity in HaCaT cells is influenced by both dose and particle size, and is associated with the induction of pyroptosis. Frequent and large exposures to nano- TiO2 in daily life may cause serious health hazards.
Collapse
Affiliation(s)
- Wanting Fu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mingxue Liu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yu Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Huimin Yang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Aoqi Ye
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jianhong Wu
- Wuhan Institute for Drug and Medical Device Control, Wuhan 430075, China
| | - Yang Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zejun Yu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yinsheng Qiu
- School of Animal Science and Nutrition Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Lingyun Xu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
7
|
Lee CK, Wang FT, Huang CH, Chan WH. Dose-dependent effects of silver nanoparticles on cell death modes in mouse blastocysts induced via endoplasmic reticulum stress and mitochondrial apoptosis. Toxicol Res (Camb) 2024; 13:tfae158. [PMID: 39371680 PMCID: PMC11447381 DOI: 10.1093/toxres/tfae158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/19/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024] Open
Abstract
In view of the rapidly expanding medical and commercial applications of silver nanoparticles (AgNPs), their potential health risks and environmental effects are a significant growing concern. Earlier research by our group uncovered the embryotoxic potential of AgNPs, showing detrimental impacts of these nanoparticles on both pre- and post-implantation embryonic development. In the current study, we showed that low (50-100 μM) and high (200-400 μM) dose ranges of AgNPs trigger distinct cell death programs affecting mouse embryo development and further explored the underlying mechanisms. Treatment with low concentrations of AgNPs (50-100 μM) triggered ROS generation, in turn, inducing mitochondria-dependent apoptosis, and ultimately, harmful effects on embryo implantation, post-implantation development, and fetal development. Notably, high concentrations of AgNPs (200-400 μM) evoked more high-level ROS generation and endoplasmic reticulum (ER) stress-mediated necrosis. Interestingly, pre-incubation with Trolox, a strong antioxidant, reduced ROS generation in the group treated with 200-400 μM AgNPs to the level induced by 50-100 μM AgNPs, resulting in switching of the cell death mode from necrosis to apoptosis and a significant improvement in the impairment of embryonic development. Our findings additionally indicate that activation of PAK2 is a crucial step in AgNP-triggered apoptosis and sequent detrimental effects on embryonic development. Based on the collective results, we propose that the levels of ROS generated by AgNP treatment of embryos serve as a critical regulator of cell death type, leading to differential degrees of damage to embryo implantation, post-implantation development and fetal development through triggering apoptosis, necrosis or other cell death signaling cascades.
Collapse
Affiliation(s)
- Cheng-Kai Lee
- Department of Obstetrics and Gynecology, Taoyuan General Hospital, Ministry of Health & Welfare, Zhongshan Road, Taoyuan District, Taoyuan City 33004, Taiwan
| | - Fu-Ting Wang
- Rehabilitation and Technical Aid Center, Taipei Veterans General Hospital, Section 2, Shipai Road, Beitou District, Taipei City 11217, Taiwan
| | - Chien-Hsun Huang
- Hungchi Gene IVF Center, Taoyuan District, Daxing West Road, Taoyuan District, Taoyuan City 330012, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City 32023, Taiwan
| |
Collapse
|
8
|
Snyder CM, Mateo B, Patel K, Fahrenholtz CD, Rohde MM, Carpenter R, Singh RN. Enhancement of Triple-Negative Breast Cancer-Specific Induction of Cell Death by Silver Nanoparticles by Combined Treatment with Proteotoxic Stress Response Inhibitors. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1564. [PMID: 39404291 PMCID: PMC11477547 DOI: 10.3390/nano14191564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Metal nanoparticles have been tested for therapeutic and imaging applications in pre-clinical models of cancer, but fears of toxicity have limited their translation. An emerging concept in nanomedicine is to exploit the inherent drug-like properties of unmodified nanomaterials for cancer therapy. To be useful clinically, there must be a window between the toxicity of the nanomaterial to cancer and toxicity to normal cells. This necessitates identification of specific vulnerabilities in cancers that can be targeted using nanomaterials without inducing off-target toxicity. Previous studies point to proteotoxic stress as a driver of silver nanoparticle (AgNPs) toxicity. Two key cell stress responses involved in mitigating proteotoxicity are the heat shock response (HSR) and the integrated stress response (ISR). Here, we examine the role that these stress responses play in AgNP-induced cytotoxicity in triple-negative breast cancer (TNBC) and immortalized mammary epithelial cells. Furthermore, we investigate HSR and ISR inhibitors as potential drug partners to increase the anti-cancer efficacy of AgNPs without increasing off-target toxicity. We showed that AgNPs did not strongly induce the HSR at a transcriptional level, but instead decreased expression of heat shock proteins (HSPs) at the protein level, possibly due to degradation in AgNP-treated TNBC cells. We further showed that the HSR inhibitor, KRIBB11, synergized with AgNPs in TNBC cells, but also increased off-target toxicity in immortalized mammary epithelial cells. In contrast, we found that salubrinal, a drug that can sustain pro-death ISR signaling, enhanced AgNP-induced cell death in TNBC cells without increasing toxicity in immortalized mammary epithelial cells. Subsequent co-culture studies demonstrated that AgNPs in combination with salubrinal selectively eliminated TNBCs without affecting immortalized mammary epithelial cells grown in the same well. Our findings provide additional support for proteotoxic stress as a mechanism by which AgNPs selectively kill TNBCs and will help guide future efforts to identify drug partners that would be beneficial for use with AgNPs for cancer therapy.
Collapse
Affiliation(s)
- Christina M. Snyder
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.M.S.); (B.M.); (K.P.); (C.D.F.); (M.M.R.)
| | - Beatriz Mateo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.M.S.); (B.M.); (K.P.); (C.D.F.); (M.M.R.)
| | - Khushbu Patel
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.M.S.); (B.M.); (K.P.); (C.D.F.); (M.M.R.)
| | - Cale D. Fahrenholtz
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.M.S.); (B.M.); (K.P.); (C.D.F.); (M.M.R.)
- Fred Wilson School of Pharmacy, High Point University, High Point, NC 27268, USA
| | - Monica M. Rohde
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.M.S.); (B.M.); (K.P.); (C.D.F.); (M.M.R.)
| | - Richard Carpenter
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Bloomington, IN 47405, USA;
| | - Ravi N. Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (C.M.S.); (B.M.); (K.P.); (C.D.F.); (M.M.R.)
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| |
Collapse
|
9
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
| | | | | | - Weiqi Hong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
10
|
Skóra B, Piechowiak T, Szychowski KA. Engagement of specific intracellular pathways in the inflammation-based reprotoxicity effect of small-size silver nanoparticles on spermatogonia and spermatocytes invitro cell models. CHEMOSPHERE 2024; 363:142897. [PMID: 39029710 DOI: 10.1016/j.chemosphere.2024.142897] [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: 02/18/2024] [Revised: 04/25/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Male infertility is a serious ongoing problem, whose causes have not yet been clearly identified. However, since human exposure to silver nanoparticles (AgNPs) has recently increased due to their beneficial properties, the present study aimed to determine the impact of small-size AgNPs on mouse spermatogonia (GC-1 spg) and spermatocytes [GC-2 spd(ts)] in vitro models as well as the ability of these nanostructures to induce inflammation. The results showed a significant dose- and time-dependent decrease in the metabolic activity in both cell models, which was correlated with an increase in the intracellular ROS level. Moreover, increased activity of caspase-9 and -3, together with enhanced expression of CASP3 and p(S15)-p53 proteins, was detected. Further studies indicated a decrease in ΔΨm after the AgNP-treatment, which proves induction of apoptosis with engagement of an intrinsic pathway. The PARP1 protein expression, the activity and protein expression of antioxidant enzymes, the GSH level, and the increased level of p-ERK1/2 indicate not only the engagement of DNA damage but also the occurrence of oxidative stress. The small-size AgNPs were able to induce inflammation, proved by increased protein expression of NF-κB, p-IκBα, and NLRP3, which indicate damage to spermatogonia and spermatocyte cells. Moreover, the PGC-1α/PPARγ and NRF2/Keap1 pathways were engaged in the observed effect. The spermatogonial cells were characterized by a stronger inflammation-based response to AgNPs, which may be correlated with the TNFα/TRAF2-based pathway. Summarizing, the obtained results prove that AgNPs impair the function of testis-derived cells by inducing the redox imbalance and inflammation process; therefore, these NPs should be carefully implemented in the human environment.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszów, St. Sucharskiego 2, 35-225, Rzeszów, Poland.
| | - Tomasz Piechowiak
- Department of Chemistry and Food Toxicology, Institute of Food Technology and Nutrition, University of Rzeszów, St. Ćwiklinskiej 1A, 35-601, Rzeszów, Poland
| | - Konrad A Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszów, St. Sucharskiego 2, 35-225, Rzeszów, Poland
| |
Collapse
|
11
|
Guo S, Tong Y, Li T, Yang K, Gao W, Peng F, Zou X. Endoplasmic Reticulum Stress-Mediated Cell Death in Renal Fibrosis. Biomolecules 2024; 14:919. [PMID: 39199307 PMCID: PMC11352060 DOI: 10.3390/biom14080919] [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: 05/17/2024] [Revised: 07/04/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
The endoplasmic reticulum (ER) is indispensable for maintaining normal life activities. Dysregulation of the ER function results in the accumulation of harmful proteins and lipids and the disruption of intracellular signaling pathways, leading to cellular dysfunction and eventual death. Protein misfolding within the ER disrupts its delicate balance, resulting in the accumulation of misfolded or unfolded proteins, a condition known as endoplasmic reticulum stress (ERS). Renal fibrosis, characterized by the aberrant proliferation of fibrotic tissue in the renal interstitium, stands as a grave consequence of numerous kidney disorders, precipitating a gradual decline in renal function. Renal fibrosis is a serious complication of many kidney conditions and is characterized by the overgrowth of fibrotic tissue in the glomerular and tubular interstitium, leading to the progressive failure of renal function. Studies have shown that, during the onset and progression of kidney disease, ERS causes various problems in the kidneys, a process that can lead to kidney fibrosis. This article elucidates the underlying intracellular signaling pathways modulated by ERS, delineating its role in triggering diverse forms of cell death. Additionally, it comprehensively explores a spectrum of potential pharmacological agents and molecular interventions aimed at mitigating ERS, thereby charting novel research avenues and therapeutic advancements in the management of renal fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China; (S.G.); (Y.T.); (T.L.); (K.Y.); (W.G.); (F.P.)
| |
Collapse
|
12
|
Liu SF, Li MJ, Liang B, Sun W, Shao Y, Hu X, Xing D. Breaking the barrier: Nanoparticle-enhanced radiotherapy as the new vanguard in brain tumor treatment. Front Pharmacol 2024; 15:1394816. [PMID: 39021831 PMCID: PMC11252536 DOI: 10.3389/fphar.2024.1394816] [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: 03/02/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
The pursuit of effective treatments for brain tumors has increasingly focused on the promising area of nanoparticle-enhanced radiotherapy (NERT). This review elucidates the context and significance of NERT, with a particular emphasis on its application in brain tumor therapy-a field where traditional treatments often encounter obstacles due to the blood-brain barrier (BBB) and tumor cells' inherent resistance. The aims of this review include synthesizing recent advancements, analyzing action mechanisms, and assessing the clinical potential and challenges associated with nanoparticle (NP) use in radiotherapy enhancement. Preliminary preclinical studies have established a foundation for NERT, demonstrating that nanoparticles (NPs) can serve as radiosensitizers, thereby intensifying radiotherapy's efficacy. Investigations into various NP types, such as metallic, magnetic, and polymeric, have each unveiled distinct interactions with ionizing radiation, leading to an augmented destruction of tumor cells. These interactions, encompassing physical dose enhancement and biological and chemical radio sensitization, are crucial to the NERT strategy. Although clinical studies are in their early phases, initial trials have shown promising results in terms of tumor response rates and survival, albeit with mindful consideration of toxicity profiles. This review examines pivotal studies affirming NERT's efficacy and safety. NPs have the potential to revolutionize radiotherapy by overcoming challenges in targeted delivery, reducing off-target effects, and harmonizing with other modalities. Future directions include refining NP formulations, personalizing therapies, and navigating regulatory pathways. NERT holds promise to transform brain tumor treatment and provide hope for patients.
Collapse
Affiliation(s)
- Shi feng Liu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Meng Jiao Li
- The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Wenshe Sun
- The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Yingchun Shao
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Xiaokun Hu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| |
Collapse
|
13
|
Mishra T, Sengupta P, Basu S. Biomaterials for Targeting Endoplasmic Reticulum in Cancer. Chem Asian J 2024; 19:e202400250. [PMID: 38602248 DOI: 10.1002/asia.202400250] [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: 03/06/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/12/2024]
Abstract
Endoplasmic reticulum (ER) is one of the most important sub-cellular organelles which controls myriads of biological functions including protein biosynthesis with proper functional folded form, protein misfolding, protein transport into Golgi body for secretion, Ca2+ homeostasis and so on. Subsequently, dysregulation in ER function leads to ER stress followed by disease pathology like cancer. Hence, targeting ER in the cancer cells emerged as one of the futuristic strategies for cancer treatment. However, the major challenge is to selectively and specifically target ER in the sub-cellular milieu in the cancer tissues, due to the lack of ER targeting chemical moieties to recognize the ER markers. To address this, in the last decade, numerous biomaterials were explored to selectively impair and image ER in cancer cells to induce ER stress. This review outlines those biomaterials which consists of carbon and silicon materials, lipid nanoparticles (liposomes and micelles), supramolecular self-assembled nanostructures, cell membrane-coated nanoparticles and metallic nanoparticles. Moreover, we also discuss the challenges and possible solutions of this promising field to usher the readers towards next-generation ER targeted cancer therapy.
Collapse
Affiliation(s)
- Tripti Mishra
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| | - Poulomi Sengupta
- Department of Chemistry, Indrashil University, Rajpur, Kadi, Mehsana, Gujarat, 382740, India
| | - Sudipta Basu
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, 382355, India
| |
Collapse
|
14
|
Qiao S, Kang Y, Tan X, Zhou X, Zhang C, Lai S, Liu J, Shao L. Nanomaterials-induced programmed cell death: Focus on mitochondria. Toxicology 2024; 504:153803. [PMID: 38616010 DOI: 10.1016/j.tox.2024.153803] [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: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Nanomaterials are widely utilized in several domains, such as everyday life, societal manufacturing, and biomedical applications, which expand the potential for nanomaterials to penetrate biological barriers and interact with cells. Multiple studies have concentrated on the particular or improper utilization of nanomaterials, resulting in cellular death. The primary mode of cell death caused by nanotoxicity is programmable cell death, which includes apoptosis, ferroptosis, necroptosis, and pyroptosis. Based on our prior publications and latest research, mitochondria have a vital function in facilitating programmed cell death caused by nanomaterials, as well as initiating or transmitting death signal pathways associated with it. Therefore, this review takes mitochondria as the focal point to investigate the internal molecular mechanism of nanomaterial-induced programmed cell death, with the aim of identifying potential targets for prevention and treatment in related studies.
Collapse
Affiliation(s)
- Shijia Qiao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiyuan Kang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiner Tan
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xinru Zhou
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Can Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shulin Lai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
15
|
Li J, Li M, Wang R, Lan J, Yu L, Gao J, Lü H, Fang Q, Wang F. Mitophagy protects against silver nanoparticle-induced hepatotoxicity by inhibiting mitochondrial ROS and the NLRP3 inflammasome. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116137. [PMID: 38417314 DOI: 10.1016/j.ecoenv.2024.116137] [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: 11/08/2023] [Revised: 01/26/2024] [Accepted: 02/19/2024] [Indexed: 03/01/2024]
Abstract
Silver nanoparticles (AgNPs) have wide clinical applications because of their excellent antibacterial properties; however, they can cause liver inflammation in animals. Macrophages are among the main cells mediating inflammation and are also responsible for the phagocytosis of nanomaterials. The NLRP3 inflammasome is a major mechanism of inflammation, and its activation both induces cytokine release and triggers inflammatory cell death (i.e., pyroptosis). In previous studies, we demonstrated that mitophagy activation plays a protective role against AgNP-induced hepatotoxicity. However, the exact molecular mechanisms underlying these processes are not fully understood. In this study, we demonstrate that AgNP exposure induces NLRP3 inflammasome activation, mitochondrial damage and pyroptosis in vivo and in vitro. NLRP3 silencing or inhibiting mitochondrial reactive oxygen species (ROS) overproduction reduces PINK1-Parkin-mediated mitophagy. Meanwhile, the inhibition of mitophagy ROS production, mitochondrial, NLRP3-mediated inflammation, and pyroptosis in RAW264.7 cells were more pronounced than in the control group. These results suggest that PINK1-Parkin-mediated mitophagy plays a protective role by reducing AgNP-induced mitochondrial ROS and subsequent NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Jiangyan Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, China
| | - Ming Li
- Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Ruirui Wang
- Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Jiaqi Lan
- Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Lian Yu
- Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Jie Gao
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hezuo Lü
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Qiang Fang
- Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Fengchao Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| |
Collapse
|
16
|
Canup B, Rogers P, Paredes A, Manheng W, Lyn-Cook B, Fahmi T. Investigation of sex-based differences in the immunotoxicity of silver nanoparticles. Nanotoxicology 2024; 18:134-159. [PMID: 38444264 DOI: 10.1080/17435390.2024.2323070] [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: 11/14/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024]
Abstract
The growing application of silver nanoparticles (AgNPs) in consumer, healthcare, and industrial products has raised concern over potential health implications due to increasing exposure. The evaluation of the immune response to nanomaterials is one of the key criteria to assess their biocompatibility. There are well-recognized sex-based differences in innate and adaptive immune responses. However, there is limited information available using human models. The aim was to investigate the potential sex-based differences in immune functions after exposure to AgNPs using human peripheral blood mononuclear cells (PBMCs) and plasma from healthy donors. These functions include inflammasome activation, cytokine expression, leukocyte proliferation, chemotaxis, plasma coagulation, and complement activation. AgNPs were characterized by dynamic light scattering and transmission electron microscopy. Inflammasome activation by AgNPs was measured after 6- and 24-hours incubations. AgNPs-induced inflammasome activation was significantly higher in the females, especially for the 6-hour exposure. No sex-based differences were observed for Ag ions controls. Younger donors exhibited significantly more inflammasome activation than older donors after 24-hours exposure. IL-10 was significantly suppressed in males and females after exposure. AgNPs suppressed leukocyte proliferation similarly in males and females. No chemoattractant effects, no alterations in plasma coagulation, or activation of the complement were observed after AgNPs exposure. In conclusion, the results highlight that there are distinct sex-based differences in inflammasome activation after exposure to AgNPs in human PBMCs. The results highlight the importance of considering sex-based differences in inflammasome activation induced by exposure to AgNPs in any future biocompatibility assessment for products containing AgNPs.
Collapse
Affiliation(s)
- Brandon Canup
- Division of Biochemical Toxicology, Office of Research, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Paul Rogers
- Division of Bioinformatics and Biostatistics, Office of Research, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Angel Paredes
- Nanotechnology Core Facility, Office of Scientific Coordination, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Wimolnut Manheng
- Division of Hematology Oncology Toxicology, Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Beverly Lyn-Cook
- Division of Biochemical Toxicology, Office of Research, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| | - Tariq Fahmi
- Division of Biochemical Toxicology, Office of Research, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
17
|
Cheng C, Yuan Y, Yuan F, Li X. Acute kidney injury: exploring endoplasmic reticulum stress-mediated cell death. Front Pharmacol 2024; 15:1308733. [PMID: 38434710 PMCID: PMC10905268 DOI: 10.3389/fphar.2024.1308733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/31/2024] [Indexed: 03/05/2024] Open
Abstract
Acute kidney injury (AKI) is a global health problem, given its substantial morbidity and mortality rates. A better understanding of the mechanisms and factors contributing to AKI has the potential to guide interventions aimed at mitigating the risk of AKI and its subsequent unfavorable outcomes. Endoplasmic reticulum stress (ERS) is an intrinsic protective mechanism against external stressors. ERS occurs when the endoplasmic reticulum (ER) cannot deal with accumulated misfolded proteins completely. Excess ERS can eventually cause pathological reactions, triggering various programmed cell death (autophagy, ferroptosis, apoptosis, pyroptosis). This article provides an overview of the latest research progress in deciphering the interaction between ERS and different programmed cell death. Additionally, the report consolidates insights into the roles of ERS in AKI and highlights the potential avenues for targeting ERS as a treatment direction toward for AKI.
Collapse
Affiliation(s)
- Cong Cheng
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Yuan
- Department of Emergency, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, Hunan, China
| | - Fang Yuan
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan, China
| | - Xin Li
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan, China
| |
Collapse
|
18
|
Talarska P, Błaszkiewicz P, Kostrzewa A, Wirstlein P, Cegłowski M, Nowaczyk G, Dudkowiak A, Grabarek BO, Głowacka-Stalmach P, Szarpak A, Żurawski J. Effects of Spherical and Rod-like Gold Nanoparticles on the Reactivity of Human Peripheral Blood Leukocytes. Antioxidants (Basel) 2024; 13:157. [PMID: 38397755 PMCID: PMC10885998 DOI: 10.3390/antiox13020157] [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: 12/30/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Gold nanoparticles (GNPs) are widely used in the technological and biomedical industries, which is a major driver of research on these nanoparticles. The main goal of this study was to determine the influence of GNPs (at 20, 100, and 200 μg/mL concentrations) on the reactivity of human peripheral blood leukocytes. Flow cytometry was used to evaluate the respiratory burst activity and pyroptosis in monocytes and granulocytes following incubation with GNPs for 30 and 60 min. Furthermore, the concentration of interleukin-1β (IL-1β) in human blood samples was assessed using enzyme-linked immunosorbent assay (ELISA) after their incubation with GNPs for 24 h. Under the conditions tested in the study, the GNPs did not significantly affect the production of reactive oxygen species in the granulocytes and monocytes that were not stimulated using phorbol 12-myristate 13-acetate (PMA) in comparison to the samples exposed to PMA (p < 0.05). Compared to the control sample, the greatest significant increase in the mean fluorescence intensity of the granulocytes occurred in the samples incubated with CGNPs = 100 and 200 µg/mL for tinc = 30 and 60 min (p < 0.05). From our results, we conclude that the physicochemical properties of the nanoparticles, chemical composition, and the type of nanoparticles used in the unit, along with the unit and incubation time, influence the induced toxicity.
Collapse
Affiliation(s)
- Patrycja Talarska
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (A.K.); (J.Ż.)
| | - Paulina Błaszkiewicz
- Faculty of Materials Engineering and Technical Physics, Poznan University of Technology, 60-965 Poznan, Poland; (P.B.); (A.D.)
| | - Artur Kostrzewa
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (A.K.); (J.Ż.)
| | - Przemysław Wirstlein
- Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznan, Poland;
| | - Michał Cegłowski
- Faculty of Chemistry, Adam Mickiewicz University Poznań, 61-614 Poznan, Poland;
| | - Grzegorz Nowaczyk
- NanoBioMedical Centre, Adam Mickiewicz University Poznań, 61-614 Poznan, Poland;
| | - Alina Dudkowiak
- Faculty of Materials Engineering and Technical Physics, Poznan University of Technology, 60-965 Poznan, Poland; (P.B.); (A.D.)
| | | | | | - Agnieszka Szarpak
- Faculty of Medicine, Uczelnia Medyczna im. Marii Skłodowskiej-Curie, 00-136 Warszawa, Poland;
| | - Jakub Żurawski
- Department of Immunobiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland; (A.K.); (J.Ż.)
| |
Collapse
|
19
|
Wang Z, Wang M, Zeng X, Yue X, Wei P. Nanomaterial-induced pyroptosis: a cell type-specific perspective. Front Cell Dev Biol 2024; 11:1322305. [PMID: 38264354 PMCID: PMC10803419 DOI: 10.3389/fcell.2023.1322305] [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: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 01/25/2024] Open
Abstract
This review presents the advancements in nanomaterial (NM)-induced pyroptosis in specific types of cells. We elucidate the relevance of pyroptosis and delineate its mechanisms and classifications. We also retrospectively analyze pyroptosis induced by various NMs in a broad spectrum of non-tumorous cellular environments to highlight the multifunctionality of NMs in modulating cell death pathways. We identify key knowledge gaps in current research and propose potential areas for future exploration. This review emphasizes the need to focus on less-studied areas, including the pathways and mechanisms of NM-triggered pyroptosis in non-tumor-specific cell types, the interplay between biological and environmental factors, and the interactions between NMs and cells. This review aims to encourage further investigations into the complex interplay between NMs and pyroptosis, thereby providing a basis for developing safer and more effective nanomedical therapeutic applications.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Min Wang
- Department of Pharmaceutics, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xuan Zeng
- Department of Pharmaceutics, Guangdong Provincial People’s Hospital Zhuhai Hospital, Zhuhai, China
| | - Xupeng Yue
- College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Pei Wei
- Department of Immunology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| |
Collapse
|
20
|
Lin X, Chen T. A Review of in vivo Toxicity of Quantum Dots in Animal Models. Int J Nanomedicine 2023; 18:8143-8168. [PMID: 38170122 PMCID: PMC10759915 DOI: 10.2147/ijn.s434842] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
Tremendous research efforts have been devoted to nanoparticles for applications in optoelectronics and biomedicine. Over the past decade, quantum dots (QDs) have become one of the fastest growing areas of research in nanotechnology because of outstanding photophysical properties, including narrow and symmetrical emission spectrum, broad fluorescence excitation spectrum, the tenability of the emission wavelength with the particle size and composition, anti-photobleaching ability and stable fluorescence. These characteristics are suitable for optical imaging, drug delivery and other biomedical applications. Research on QDs toxicology has demonstrated QDs affect or damage the biological system to some extent, and this situation is generally caused by the metal ions and some special properties in QDs, which hinders the further application of QDs in the biomedical field. The toxicological mechanism mainly stems from the release of heavy metal ions and generation of reactive oxygen species (ROS). At the same time, the contact reaction with QDs also cause disorders in organelles and changes in gene expression profiles. In this review, we try to present an overview of the toxicity and related toxicity mechanisms of QDs in different target organs. It is believed that the evaluation of toxicity and the synthesis of environmentally friendly QDs are the primary issues to be addressed for future widespread applications. However, considering the many different types and potential modifications, this review on the potential toxicity of QDs is still not clearly elucidated, and further research is needed on this meaningful topic.
Collapse
Affiliation(s)
- Xiaotan Lin
- School of Basic Medicine, Guangdong Medical University, DongGuan, People’s Republic of China
- Department of Family Planning, Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, People’s Republic of China
| | - Tingting Chen
- School of Basic Medicine, Guangdong Medical University, DongGuan, People’s Republic of China
| |
Collapse
|
21
|
Girigoswami K, Pallavi P, Girigoswami A. Intricate subcellular journey of nanoparticles to the enigmatic domains of endoplasmic reticulum. Drug Deliv 2023; 30:2284684. [PMID: 37990530 PMCID: PMC10987057 DOI: 10.1080/10717544.2023.2284684] [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: 08/24/2023] [Accepted: 11/05/2023] [Indexed: 11/23/2023] Open
Abstract
It is evident that site-specific systemic drug delivery can reduce side effects, systemic toxicity, and minimal dosage requirements predominantly by delivering drugs to particular pathological sites, cells, and even subcellular structures. The endoplasmic reticulum (ER) and associated cell organelles play a vital role in several essential cellular functions and activities, such as the synthesis of lipids, steroids, membrane-associated proteins along with intracellular transport, signaling of Ca2+, and specific response to stress. Therefore, the dysfunction of ER is correlated with numerous diseases where cancer, neurodegenerative disorders, diabetes mellitus, hepatic disorder, etc., are very common. To achieve satisfactory therapeutic results in certain diseases, it is essential to engineer delivery systems that can effectively enter the cells and target ER. Nanoparticles are highly biocompatible, contain a variety of cargos or payloads, and can be modified in a pliable manner to achieve therapeutic effectiveness at the subcellular level when delivered to specific organelles. Passive targeting drug delivery vehicles, or active targeting drug delivery systems, reduce the nonselective accumulation of drugs while reducing side effects by modifying them with small molecular compounds, antibodies, polypeptides, or isolated bio-membranes. The targeting of ER and closely associated organelles in cells using nanoparticles, however, is still unsymmetrically understood. Therefore, here we summarized the pathophysiological prospect of ER stress, involvement of ER and mitochondrial response, disease related to ER dysfunctions, essential therapeutics, and nanoenabled modulation of their delivery to optimize therapy.
Collapse
Affiliation(s)
- Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| | - Pragya Pallavi
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| |
Collapse
|
22
|
Strużyńska L. Dual Implications of Nanosilver-Induced Autophagy: Nanotoxicity and Anti-Cancer Effects. Int J Mol Sci 2023; 24:15386. [PMID: 37895066 PMCID: PMC10607027 DOI: 10.3390/ijms242015386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
In recent years, efforts have been made to identify new anti-cancer therapies. Various types of nanomaterials, including silver nanoparticles (AgNPs), are being considered as an option. In addition to its well-known antibacterial activity, AgNPs exhibit cytotoxic potential in both physiological and cancer cells by inducing stress-mediated autophagy and apoptotic cell death. A rapidly growing collection of data suggests that the proper regulation of autophagic machinery may provide an efficient tool for suppressing the development of cancer. In this light, AgNPs have emerged as a potential anti-cancer agent to support therapy of the disease. This review summarizes current data indicating the dual role of AgNP-induced autophagy and highlights factors that may influence its protective vs. its toxic potential. It also stresses that our understanding of the cellular and molecular mechanisms of autophagy machinery in cancer cells, as well as AgNP-triggered autophagy in both normal and diseased cells, remains insufficient.
Collapse
Affiliation(s)
- Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego str., 02-106 Warsaw, Poland
| |
Collapse
|
23
|
Wang K, Wang S, Yin J, Yang Q, Yu Y, Chen L. Long-term application of silver nanoparticles in dental restoration materials: potential toxic injury to the CNS. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:52. [PMID: 37855967 PMCID: PMC10587321 DOI: 10.1007/s10856-023-06753-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/20/2023] [Indexed: 10/20/2023]
Abstract
Silver nanoparticles (AgNPs) have durable and remarkable antimicrobial effects on pathogenic microorganisms, such as bacteria and fungi, in dental plaques. As such, they are widely added to dental restoration materials, including composite resins, denture bases, adhesives, and implants, to solve the problems of denture stomatitis, peri-implant inflammation, and oral infection caused by the long-term use of these dental restoration materials. However, AgNPs can be absorbed into the blood circulatory system through the nasal/oral mucosa, lungs, gastrointestinal tract, skin, and other pathways and then distributed into the lungs, kidneys, liver, spleen, and testes, thereby causing toxic injury to these tissues and organs. It can even be transported across the blood-brain barrier (BBB) and continuously accumulate in brain tissues, causing injury and dysfunction of neurons and glial cells; consequently, neurotoxicity occurs. Other nanomaterials with antibacterial or remineralization properties are added to dental restoration materials with AgNPs. However, studies have yet to reveal the neurotoxicity caused by dental restoration materials containing AgNPs. In this review, we summarize the application of AgNPs in dental restoration materials, the mechanism of AgNPs in cytotoxicity and toxic injury to the BBB, and the related research on the accumulation of AgNPs to cause changes of neurotoxicity. We also discuss the mechanisms of neurotoxicity caused by AgNPs and the mode and rate of AgNPs released from dental restorative materials added with AgNPs to evaluate the probability of neurotoxic injury to the central nervous system (CNS), and then provide a theoretical basis for developing new composite dental restoration materials. Mechanism of neurotoxicity caused by AgNPs: AgNPs in the blood circulation enter the brain tissue after being transported across the BBB through transendothelial cell pathway and paracellular transport pathway, and continuously accumulate in brain tissue, causing damage and dysfunction of neurons and glial cells which ultimately leads to neurotoxicity. The uptake of AgNPs by neurons, astrocytes and microglia causes damage to these cells. AgNPs with non-neurotoxic level often increases the secretion of a variety of cytokines, up-regulates the expression of metallothionein in glial cells, even up-regulates autophagy and inflammation response to protect neurons from the toxic damage of AgNPs. However, the protective effect of glial cells induced by AgNPs exposure to neurotoxic levels is insufficient, which leads to neuronal damage and dysfunction and even neuronal programmed cell death, eventually cause neurotoxicity.
Collapse
Affiliation(s)
- Kaimei Wang
- Guiyang Hospital of Stomatology, Guiyang, Guizhou Province, 563000, China
| | - Shiqi Wang
- The Medical unit of 65651 troops of Chinese people's Liberation Army, Jinzhou, Liaoning Province, 121100, China
| | - Jingju Yin
- Fujian Medical University; Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350002, China
| | - Qiankun Yang
- The Southwest Hospital of Army Medical University, Chongqing, 400038, China
| | - Yi Yu
- Guiyang Hospital of Stomatology, Guiyang, Guizhou Province, 563000, China
| | - Lin Chen
- Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563100, China.
| |
Collapse
|
24
|
Bi J, Mo C, Li S, Huang M, Lin Y, Yuan P, Liu Z, Jia B, Xu S. Immunotoxicity of metal and metal oxide nanoparticles: from toxic mechanisms to metabolism and outcomes. Biomater Sci 2023. [PMID: 37161951 DOI: 10.1039/d3bm00271c] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The influence of metal and metal oxide nanomaterials on various fields since their discovery has been remarkable. They have unique properties, and therefore, have been employed in specific applications, including biomedicine. However, their potential health risks cannot be ignored. Several studies have shown that exposure to metal and metal oxide nanoparticles can lead to immunotoxicity. Different types of metals and metal oxide nanoparticles may have a negative impact on the immune system through various mechanisms, such as inflammation, oxidative stress, autophagy, and apoptosis. As an essential factor in determining the function and fate of immune cells, immunometabolism may also be an essential target for these nanoparticles to exert immunotoxic effects in vivo. In addition, the biodegradation and metabolic outcomes of metal and metal oxide nanoparticles are also important considerations in assessing their immunotoxic effects. Herein, we focus on the cellular mechanism of the immunotoxic effects and toxic effects of different types of metal and metal oxide nanoparticles, as well as the metabolism and outcomes of these nanoparticles in vivo. Also, we discuss the relationship between the possible regulatory effect of nanoparticles on immunometabolism and their immunotoxic effects. Finally, we present perspectives on the future research and development direction of metal and metal oxide nanomaterials to promote scientific research on the health risks of nanomaterials and reduce their adverse effects on human health.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Mingshu Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
Tkachenko A, Onishchenko A, Myasoedov V, Yefimova S, Havranek O. Assessing regulated cell death modalities as an efficient tool for in vitro nanotoxicity screening: a review. Nanotoxicology 2023; 17:218-248. [PMID: 37083543 DOI: 10.1080/17435390.2023.2203239] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Nanomedicine is a fast-growing field of nanotechnology. One of the major obstacles for a wider use of nanomaterials for medical application is the lack of standardized toxicity screening protocols for assessing the safety of newly synthesized nanomaterials. In this review, we focus on less frequently studied nanomaterials-induced regulated cell death (RCD) modalities, including eryptosis, necroptosis, pyroptosis, and ferroptosis, as a tool for in vitro nanomaterials safety evaluation. We summarize the latest insights into the mechanisms that mediate these RCDs in response to nanomaterials exposure. Comprehensive data from reviewed studies suggest that ROS (reactive oxygen species) overproduction and ROS-mediated pathways play a central role in nanomaterials-induced RCDs activation. On the other hand, studies also suggest that individual properties of nanomaterials, including size, shape, or surface charge, could determine specific toxicity pathways with consequent RCD induction as well. We anticipate that the evaluation of RCDs can become one of the mechanism-based screening methods in nanotoxicology. In addition to the toxicity assessment, evaluation of necroptosis-, pyroptosis-, and ferroptosis-promoting capacity of nanomaterials could simultaneously provide useful information for specific medical applications as could be their anti-tumor potential. Moreover, a detailed understanding of molecular mechanisms driving nanomaterials-mediated induction of immunogenic RCDs will substantially aid novel anti-tumor nanodrugs development.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Anatolii Onishchenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Valeriy Myasoedov
- Department of Medical Biology, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Svetlana Yefimova
- Institute for Scintillation Materials, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
26
|
Gong J, Cheng X, Zuo J, Zhang Y, Lin J, Liu M, Jiang Y, Long Y, Si H, Gao X, Guo D, Gu N. Silver nanoparticles combat Salmonella Typhimurium: Suppressing intracellular infection and activating dendritic cells. Colloids Surf B Biointerfaces 2023; 226:113307. [PMID: 37068446 DOI: 10.1016/j.colsurfb.2023.113307] [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: 11/10/2022] [Revised: 02/16/2023] [Accepted: 04/08/2023] [Indexed: 04/19/2023]
Abstract
Salmonella Typhimurium (ST) can hide inside cells, avoid antibiotic therapy and being killed by host's immune system to cause persistent infection in humans and animals. Metal nanoparticles are regarded as an alternative to overcome the above limitations, silver nanoparticles especially have been applied in combating drug-resistant bacteria. However, the therapeutic effects of silver nanoparticles against intracellular infection and their impacts on host immunity remain an area of further investigation. In this work, we synthesized Ganoderma extract-capped silver nanoparticles (Ag@Ge) and explored the therapeutic potential and immune adjuvant effects of Ag@Ge against intracellular ST. Firstly, Ag@Ge had a small particle size of 35.52±7.46 nm, good stability, and biocompatibility. Then, Ag@Ge effectively entered RAW 264.7 cells, suppressed intracellular ST infection. Furthermore, Ag@Ge activated mouse dendritic cells (DCs) in vitro, evidenced by increased phenotypic markers (CD80/CD86/CD40/major compatibility complex II (MHCII)) expression and cytokine and chemokine (interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), chemokine (C-C motif) ligand 2 (CCL-2), and chemokine (C-C motif) receptor-7 (CCR-7)) transcription. More notably, the combination of Ag@Ge with inactivated ST recruited intestinal DCs to mitigate ST infection in mice, evidenced by decreased body weight loss and bacterial loads in the tissues (liver, jejunum, and colon), and improved platelets count. The above findings indicate that Ag@Ge has the potential as an alternative nano-antibiotic against intracellular ST infection.
Collapse
Affiliation(s)
- Jiahao Gong
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Xingxing Cheng
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Jinjiao Zuo
- College of Life Sciences, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Yan Zhang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Jian Lin
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; College of Life Sciences, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Moxin Liu
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Yan Jiang
- Animal, Plant and Food Inspection Center of Nanjing Customs District, 39 Chuangzhi Road, Nanjing 210000, China
| | - Yunfeng Long
- Animal, Plant and Food Inspection Center of Nanjing Customs District, 39 Chuangzhi Road, Nanjing 210000, China
| | - Hongbin Si
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Xiuge Gao
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Dawei Guo
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China.
| | - Ning Gu
- Medical School, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| |
Collapse
|
27
|
Noga M, Milan J, Frydrych A, Jurowski K. Toxicological Aspects, Safety Assessment, and Green Toxicology of Silver Nanoparticles (AgNPs)—Critical Review: State of the Art. Int J Mol Sci 2023; 24:ijms24065133. [PMID: 36982206 PMCID: PMC10049346 DOI: 10.3390/ijms24065133] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
In recent years, research on silver nanoparticles (AgNPs) has attracted considerable interest among scientists because of, among other things, their alternative application to well-known medical agents with antibacterial properties. The size of the silver nanoparticles ranges from 1 to 100 nm. In this paper, we review the progress of research on AgNPs with respect to the synthesis, applications, and toxicological safety of AgNPs, and the issue of in vivo and in vitro research on silver nanoparticles. AgNPs’ synthesis methods include physical, chemical, and biological routes, as well as “green synthesis”. The content of this article covers issues related to the disadvantages of physical and chemical methods, which are expensive and can also have toxicity. This review pays special attention to AgNP biosafety concerns, such as potential toxicity to cells, tissues, and organs.
Collapse
Affiliation(s)
- Maciej Noga
- Department of Regulatory and Forensic Toxicology, Institute of Medical Expertise, Łódź, ul. Aleksandrowska 67/93, 91-205 Łódź, Poland
| | - Justyna Milan
- Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Studies, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Adrian Frydrych
- Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Studies, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
| | - Kamil Jurowski
- Department of Regulatory and Forensic Toxicology, Institute of Medical Expertise, Łódź, ul. Aleksandrowska 67/93, 91-205 Łódź, Poland
- Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Studies, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959 Rzeszów, Poland
- Correspondence: or
| |
Collapse
|
28
|
Zhang Z, Zhou Y, Zhao S, Ding L, Chen B, Chen Y. Nanomedicine-Enabled/Augmented Cell Pyroptosis for Efficient Tumor Nanotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203583. [PMID: 36266982 PMCID: PMC9762308 DOI: 10.1002/advs.202203583] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/23/2022] [Indexed: 05/19/2023]
Abstract
The terrible morbidity and mortality of malignant tumors urgently require innovative therapeutics, especially for apoptosis-resistant tumors. Pyroptosis, a pro-inflammatory form of programmed cell death (PCD), is featured with pore formation in plasma membrane, cell swelling with giant bubbles, and leakage of cytoplasmic pro-inflammatory cytokines, which can remodel the tumor immune microenvironment by stimulating a "cold" tumor microenvironment to be an immunogenic "hot" tumor microenvironment, and consequently augment the therapeutic efficiency of malignant tumors. Benefiting from current advances in nanotechnology, nanomedicine is extensively applied to potentiate, enable, and augment pyroptosis for enhancing cancer-therapeutic efficacy and specificity. This review provides a concentrated summary and discussion of the most recent progress achieved in this emerging field, highlighting the nanomedicine-enabled/augmented specific pyroptosis strategy for favoring the construction of next-generation nanomedicines to efficiently induce PCD. It is highly expected that the further clinical translation of nanomedicine can be accelerated by inducing pyroptotic cell death based on bioactive nanomedicines.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiang212000P. R. China
| | - Yajun Zhou
- Department of UltrasoundThe Fourth Affiliated HospitalNanjing Medical UniversityNanjing210029P. R. China
| | - Shuangshuang Zhao
- Department of UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiang212000P. R. China
| | - Li Ding
- Tongji University School of MedicineShanghai Tenth People's HospitalTongji University Cancer CenterShanghai Engineering Research Center of Ultrasound Diagnosis and TreatmentNational Clinical Research Center of Interventional MedicineShanghai200072P. R. China
| | - Baoding Chen
- Department of UltrasoundAffiliated Hospital of Jiangsu UniversityZhenjiang212000P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| |
Collapse
|
29
|
Zhao H, Yang Y, Si X, Liu H, Wang H. The Role of Pyroptosis and Autophagy in Ischemia Reperfusion Injury. Biomolecules 2022; 12:biom12071010. [PMID: 35883566 PMCID: PMC9313059 DOI: 10.3390/biom12071010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023] Open
Abstract
Pyroptosis is a process of programmed cell death mediated by gasdermin (GSDM) found in recent years. In the process of pyroptosis, caspase-1 or caspase-11/4/5 is activated, which cleaves gasdermin D and separates its N-terminal pore-forming domain (PFD). The oligomers of PFD bind to the cell membrane and form macropores on the membrane, resulting in cell swelling and membrane rupture. Increasing evidence indicates that pyroptosis is involved in many diseases, including ischemia reperfusion injury. Autophagy is a highly conserved catabolic process in eukaryotic cells. It plays an important role in the survival and maintenance of cells by degrading organelles, proteins, and macromolecules in the cytoplasm and recycling degradation products. Increasing evidence shows that dysfunctional autophagy participates in many diseases. Recently, autophagy and pyroptosis have been reported to play a vital role in the process of ischemia/reperfusion injury, but the related mechanisms are not completely clear. Therefore, this article reviews the role of autophagy and pyroptosis in ischemia–reperfusion injury and analyzes the related mechanisms to provide a basis for future research.
Collapse
Affiliation(s)
- Huijie Zhao
- Institute of Chronic Disease Risks Assessment, Henan University, Jinming Avenue, Kaifeng 475004, China;
| | - Yihan Yang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
| | - Xinya Si
- School of Stomatology, Henan University, Kaifeng 475004, China;
| | - Huiyang Liu
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
| | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
- Correspondence:
| |
Collapse
|
30
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
31
|
Huang CH, Wang FT, Chan WH. Low-dose silver nanoparticles plus methyl mercury exert embryotoxic effects on mouse blastocysts via endoplasmic reticulum stress and mitochondrial apoptosis. Toxicol Res (Camb) 2022; 11:460-474. [PMID: 35782646 PMCID: PMC9244727 DOI: 10.1093/toxres/tfac028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 07/30/2023] Open
Abstract
The health and environmental impacts of the increasing commercial use of silver nanoparticles (AgNPs) are a growing concern. Methyl mercury (MeHg) is a potent toxin that biotransforms from mercury or inorganic mercury compounds in waterways and causes dangerous environmental contamination. However, the potential interactions and combined effects of AgNPs and MeHg are yet to be established. In the current study, we showed that low/non-embryotoxic doses of AgNPs and MeHg interact synergistically to induce embryotoxicity and further explored the underlying mechanisms affecting mouse embryo development. Notably, co-treatment with noncytotoxic concentrations of AgNPs (10 μM) and MeHg (0.1 μM) triggered apoptotic processes and embryotoxicity in mouse blastocysts and evoked intracellular reactive oxygen species (ROS) generation, which was effectively blocked by preincubation with 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (trolox), a classic antioxidant. Further experiments demonstrated that ROS serve as a key upstream inducer of endoplasmic reticulum (ER) stress and mitochondria-dependent apoptotic processes in AgNP/MeHg-induced injury of mouse embryo implantation and pre- and postimplantation development. Our results collectively indicate that AgNP and MeHg at non-embryotoxic concentrations can synergistically evoke ROS, ultimately causing embryotoxicity through promotion of ER stress and mitochondria-dependent apoptotic signaling cascades.
Collapse
Affiliation(s)
- Chien-Hsun Huang
- Department of Obstetrics and Gynecology, Taoyuan General Hospital, Ministry of Health & Welfare, Zhongshan Road, Taoyuan District, Taoyuan City 33004, Taiwan
| | - Fu-Ting Wang
- Rehabilitation and Technical Aid Center, Taipei Veterans General Hospital, Section 2, Shipai Road, Beitou District, Taipei City 11217, Taiwan
| | - Wen-Hsiung Chan
- Corresponding author: Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City 32023, Taiwan.
| |
Collapse
|
32
|
Lu N, Cheng W, Liu D, Liu G, Cui C, Feng C, Wang X. NLRP3-Mediated Inflammation in Atherosclerosis and Associated Therapeutics. Front Cell Dev Biol 2022; 10:823387. [PMID: 35493086 PMCID: PMC9045366 DOI: 10.3389/fcell.2022.823387] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/24/2022] [Indexed: 11/15/2022] Open
Abstract
The NLRP3 inflammasome is a crucial constituent of the body’s innate immune system, and a multiprotein platform which is initiated by pattern recognition receptors (PRRs). Its activation leads to caspase-1 maturation and release of inflammatory cytokines, interleukin-1β (IL-1β) and IL-18, and subsequently causes pyroptosis. Recently, the excess activation of NLRP3 inflammasome has been confirmed to mediate inflammatory responses and to participate in genesis and development of atherosclerosis. Therefore, the progress on the discovery of specific inhibitors against the NLRP3 inflammasome and the upstream and downstream inflammatory factors has become potential targets for clinical treatment. Here we review the recently described mechanisms about the NLRP3 inflammasome activation, and discuss emphatically the pharmacological interventions using statins and natural medication for atherosclerosis associated with NLRP3 inflammasome.
Collapse
Affiliation(s)
- Na Lu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Weijia Cheng
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Can Cui
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Chaoli Feng
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- *Correspondence: Xianwei Wang,
| |
Collapse
|
33
|
Shi M, Chai Y, Zhang J, Chen X. Endoplasmic Reticulum Stress-Associated Neuronal Death and Innate Immune Response in Neurological Diseases. Front Immunol 2022; 12:794580. [PMID: 35082783 PMCID: PMC8784382 DOI: 10.3389/fimmu.2021.794580] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
Neuronal death and inflammatory response are two common pathological hallmarks of acute central nervous system injury and chronic degenerative disorders, both of which are closely related to cognitive and motor dysfunction associated with various neurological diseases. Neurological diseases are highly heterogeneous; however, they share a common pathogenesis, that is, the aberrant accumulation of misfolded/unfolded proteins within the endoplasmic reticulum (ER). Fortunately, the cell has intrinsic quality control mechanisms to maintain the proteostasis network, such as chaperone-mediated folding and ER-associated degradation. However, when these control mechanisms fail, misfolded/unfolded proteins accumulate in the ER lumen and contribute to ER stress. ER stress has been implicated in nearly all neurological diseases. ER stress initiates the unfolded protein response to restore proteostasis, and if the damage is irreversible, it elicits intracellular cascades of death and inflammation. With the growing appreciation of a functional association between ER stress and neurological diseases and with the improved understanding of the multiple underlying molecular mechanisms, pharmacological and genetic targeting of ER stress are beginning to emerge as therapeutic approaches for neurological diseases.
Collapse
Affiliation(s)
- Mingming Shi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Yan Chai
- Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
34
|
Targeting autophagy, oxidative stress, and ER stress for neurodegenerative diseases treatment. J Control Release 2022; 345:147-175. [DOI: 10.1016/j.jconrel.2022.03.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/13/2022]
|
35
|
The consequences of particle uptake on immune cells. Trends Pharmacol Sci 2022; 43:305-320. [DOI: 10.1016/j.tips.2022.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/11/2022]
|
36
|
Mohammapdour R, Ghandehari H. Mechanisms of immune response to inorganic nanoparticles and their degradation products. Adv Drug Deliv Rev 2022; 180:114022. [PMID: 34740764 PMCID: PMC8898339 DOI: 10.1016/j.addr.2021.114022] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/24/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023]
Abstract
Careful assessment of the biological fate and immune response of inorganic nanoparticles is crucial for use of such carriers in drug delivery and other biomedical applications. Many studies have elucidated the cellular and molecular mechanisms of the interaction of inorganic nanoparticles with the components of the immune system. The biodegradation and dissolution of inorganic nanoparticles can influence their ensuing immune response. While the immunological properties of inorganic nanoparticles as a function of their physicochemical properties have been investigated in detail, little attention has been paid to the immune adverse effects towards the degradation products of these nanoparticles. To fill this gap, we herein summarize the cellular mechanisms of immune response to inorganic nanoparticles and their degradation products with specific focus on immune cells. We also accentuate the importance of designing new methods and instruments for the in situ characterization of inorganic nanoparticles in order to assess their safety as a result of degradation. This review further sheds light on factors that need to be considered in the design of safe and effective inorganic nanoparticles for use in delivery of bioactive and imaging agents.
Collapse
Affiliation(s)
- Raziye Mohammapdour
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA.
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
37
|
Marczynski M, Lieleg O. Forgotten but not gone: Particulate matter as contaminations of mucosal systems. BIOPHYSICS REVIEWS 2021; 2:031302. [PMID: 38505633 PMCID: PMC10903497 DOI: 10.1063/5.0054075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/14/2021] [Indexed: 03/21/2024]
Abstract
A decade ago, environmental issues, such as air pollution and the contamination of the oceans with microplastic, were prominently communicated in the media. However, these days, political topics, as well as the ongoing COVID-19 pandemic, have clearly taken over. In spite of this shift in focus regarding media representation, researchers have made progress in evaluating the possible health risks associated with particulate contaminations present in water and air. In this review article, we summarize recent efforts that establish a clear link between the increasing occurrence of certain pathological conditions and the exposure of humans (or animals) to airborne or waterborne particulate matter. First, we give an overview of the physiological functions mucus has to fulfill in humans and animals, and we discuss different sources of particulate matter. We then highlight parameters that govern particle toxicity and summarize our current knowledge of how an exposure to particulate matter can be related to dysfunctions of mucosal systems. Last, we outline how biophysical tools and methods can help researchers to obtain a better understanding of how particulate matter may affect human health. As we discuss here, recent research has made it quite clear that the structure and functions of those mucosal systems are sensitive toward particulate contaminations. Yet, our mechanistic understanding of how (and which) nano- and microparticles can compromise human health via interacting with mucosal barriers is far from complete.
Collapse
|
38
|
Copper Oxide Nanoparticle-Induced Acute Inflammatory Response and Injury in Murine Lung Is Ameliorated by Synthetic Secoisolariciresinol Diglucoside (LGM2605). Int J Mol Sci 2021; 22:ijms22179477. [PMID: 34502389 PMCID: PMC8430773 DOI: 10.3390/ijms22179477] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022] Open
Abstract
Metal-oxide nanoparticles (MO-NPs), such as the highly bioreactive copper-based nanoparticles (CuO-NPs), are widely used in manufacturing of hundreds of commercial products. Epidemiological studies correlated levels of nanoparticles in ambient air with a significant increase in lung disease. CuO-NPs, specifically, were among the most potent in a set of metal-oxides and carbons studied in parallel regarding DNA damage and cytotoxicity. Despite advances in nanotoxicology research and the characterization of their toxicity, the exact mechanism(s) of toxicity are yet to be defined. We identified chlorination toxicity as a damaging consequence of inflammation and myeloperoxidase (MPO) activation, resulting in macromolecular damage and cell damage/death. We hypothesized that the inhalation of CuO-NPs elicits an inflammatory response resulting in chlorination damage in cells and lung tissues. We further tested the protective action of LGM2605, a synthetic small molecule with known scavenging properties for reactive oxygen species (ROS), but most importantly, for active chlorine species (ACS) and an inhibitor of MPO. CuO-NPs (15 µg/bolus) were instilled intranasally in mice and the kinetics of the inflammatory response in lungs was evaluated 1, 3, and 7 days later. Evaluation of the protective action of LGM2605 was performed at 24 h post-challenge, which was selected as the peak acute inflammatory response to CuO-NP. LGM2605 was given daily via gavage to mice starting 2 days prior to the time of the insult (100 mg/kg). CuO-NPs induced a significant inflammatory influx, inflammasome-relevant cytokine release, and chlorination damage in mouse lungs, which was mitigated by the action of LGM2605. Preventive action of LGM2605 ameliorated the adverse effects of CuO-NP in lung.
Collapse
|
39
|
Low Doses of Silver Nanoparticles Selectively Induce Lipid Peroxidation and Proteotoxic Stress in Mesenchymal Subtypes of Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13164217. [PMID: 34439373 PMCID: PMC8393662 DOI: 10.3390/cancers13164217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022] Open
Abstract
Molecular profiling of tumors shows that triple-negative breast cancer (TNBC) can be stratified into mesenchymal (claudin-low breast cancer; CLBC) and epithelial subtypes (basal-like breast cancer; BLBC). Subtypes differ in underlying genetics and in response to therapeutics. Several reports indicate that therapeutic strategies that induce lipid peroxidation or proteotoxicity may be particularly effective for various cancers with a mesenchymal phenotype such as CLBC, for which no specific treatment regimens exist and outcomes are poor. We hypothesized that silver nanoparticles (AgNPs) can induce proteotoxic stress and cause lipid peroxidation to a greater extent in CLBC than in BLBC. We found that AgNPs were lethal to CLBCs at doses that had little effect on BLBCs and were non-toxic to normal breast epithelial cells. Analysis of mRNA profiles indicated that sensitivity to AgNPs correlated with expression of multiple CLBC-associated genes. There was no correlation between sensitivity to AgNPs and sensitivity to silver cations, uptake of AgNPs, or proliferation rate, indicating that there are other molecular factors driving sensitivity to AgNPs. Mechanistically, we found that the differences in sensitivity of CLBC and BLBC cells to AgNPs were driven by peroxidation of lipids, protein oxidation and aggregation, and subsequent proteotoxic stress and apoptotic signaling, which were induced in AgNP-treated CLBC cells, but not in BLBC cells. This study shows AgNPs are a specific treatment for CLBC and indicates that stratification of TNBC subtypes may lead to improved outcomes for other therapeutics with similar mechanisms of action.
Collapse
|
40
|
Li J, Chang X, Shang M, Niu S, Zhang W, Li Y, Sun Z, Wu T, Kong L, Zhang T, Tang M, Xue Y. The crosstalk between DRP1-dependent mitochondrial fission and oxidative stress triggers hepatocyte apoptosis induced by silver nanoparticles. NANOSCALE 2021; 13:12356-12369. [PMID: 34254625 DOI: 10.1039/d1nr02153b] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Previous studies have revealed that the liver is the main target organ of deposition for engineered nanoparticles. The hepatotoxicity of silver nanoparticles (AgNPs), the widely used antimicrobial nanoparticles, has been of great interest. However, little is known about the regulatory mechanism of the mitochondria in AgNP-induced hepatotoxicity. In the present study, we found that AgNPs, rather than silver ions, induced mitochondrial dynamics disorders, oxidative stress, and mitochondria-dependent hepatocyte apoptosis in mice. Using human hepatocellular carcinoma (HepG2) cells, we confirmed that the interaction between dynamin-related protein 1 (DRP1)-dependent mitochondrial fission and oxidative stress promoted mitochondrial damage and mitochondria-dependent apoptosis induced by AgNPs, as determined by the elimination of DRP1 or addition of N-acetylcysteine (NAC). Interestingly, the crosstalk between DRP1-dependent mitochondrial fission and oxidative stress also activated mitophagy and autophagy flux blocking. Phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1) gene silencing contributed to the aggravation of mitochondrial damage, oxidative stress, and apoptosis. These results revealed that the interplay between mitochondrial fission and oxidative stress induced mitophagy defects and triggered AgNP-induced mitochondria-dependent apoptosis in liver cells both in vivo and in vitro. Our findings provide a perspective for the mechanism of hepatotoxicity induced by exposure to metal NPs.
Collapse
Affiliation(s)
- Jiangyan Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Rao BR, Kumar R, Haque S, Kumar JM, Rao TN, Kothapalli RVSN, Patra CR. Ag 2[Fe(CN) 5NO]-Fabricated Hydrophobic Cotton as a Potential Wound Healing Dressing: An In Vivo Approach. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10689-10704. [PMID: 33621045 DOI: 10.1021/acsami.0c19904] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
There have been reports of different types of wound dressings for various functions and purposes. Cotton being one of the most widely used wound dressing material due to its non-toxic, biodegradable, and other properties is used for fabrication as well as in the form of scaffolds for faster and effective wound closure. Our research team has already demonstrated the role of silver nitroprusside nanoparticles (SNPNPs) for wound healing and antibacterial activity. In the current study, we have developed cotton fabric impregnated with SNPNPs (SNPCFs) which remain photo inert and displayed long-term antimicrobial activity due to the surface modification with the silver nitroprusside complex. These SNPCFs were characterized by various analytical techniques (XRD, FTIR, UV spectroscopy, TGA, TEM, FESEM, EDAX, ICP-OES). The fabricated cotton dressings with nanoparticles showed an improved water contact angle (113-130°) than that of bare cotton gauze (60°) and exhibited more antibacterial property in case of both Gram-negative bacteria (Klebsiella aerogenes and Escherichia coli) and Gram-positive bacteria (Pseudomonas aeruginosa and Bacillus subtilis) even after several washings. The biocompatible nature of SNPCFs was assessed by in vivo chorioallantoic membrane assay that showed no obstruction in the formation of blood vessels. The SNPCFs exhibited better wound healing activity compared to the bare cotton and AgCFs as observed in the C57BL6/J mouse. The histopathological investigation reveals increase in re-epithelialization and deposition of connective tissue. The macrophage (M2) counts in SNPCF-treated skin tissues were supportive of more wound healing activity than mice treated with cotton fabric impregnated with chemically synthesized silver nanoparticles. Based on biodistribution analysis using ICP-OES, the data illustrated that a significant amount of silver is absorbed in the skin tissues of mice as compared to the blood and kidney. Furthermore, the absence of silver from the vital organs (heart, liver, and kidney) corroborates our hypothesis that the SNPCFs can act excellently in treating wounds when topically applied over skin. Thereafter, all these results highlight a strong possibility that SNPCFs exemplify the potential as a new antimicrobial and wound healing agent in future times.
Collapse
Affiliation(s)
- Bonda Rama Rao
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Rajnish Kumar
- Polymers & Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Shagufta Haque
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Jerald Mahesh Kumar
- Animal House, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - T Nageswara Rao
- Department of Analytical, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Raju V S N Kothapalli
- Polymers & Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
42
|
Tian B, Hua Z, Wang Z, Wang J. RETRACTED ARTICLE: Physcion 8-O-β-glucopyranoside mediates the NLRP3-associated pyroptosis and cell metastasis in the human osteosarcoma cells via ER stress activation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:555. [PMID: 32072190 DOI: 10.1007/s00210-020-01836-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Baogang Tian
- Department of Orthopedics, Wuxi Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, No.8 Zhong'nan Road, Wuxi, 214071, Jiangsu, China
| | - Zhen Hua
- Department of Orthopedics, Wuxi Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, No.8 Zhong'nan Road, Wuxi, 214071, Jiangsu, China
| | - Zhijiong Wang
- Department of Orthopedics, Wuxi Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, No.8 Zhong'nan Road, Wuxi, 214071, Jiangsu, China
| | - Jianwei Wang
- Department of Orthopedics, Wuxi Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, No.8 Zhong'nan Road, Wuxi, 214071, Jiangsu, China.
| |
Collapse
|
43
|
Hou J, Zhao L, Tang H, He X, Ye G, Shi F, Kang M, Chen H, Li Y. Silver Nanoparticles Induced Oxidative Stress and Mitochondrial Injuries Mediated Autophagy in HC11 Cells Through Akt/AMPK/mTOR Pathway. Biol Trace Elem Res 2021; 199:1062-1073. [PMID: 32666434 DOI: 10.1007/s12011-020-02212-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Silver nanoparticles (AgNPs) are widely used in industrial products, and they have good antibacterial properties, with potential for prevention and treatment of cow mastitis. However, concerns exist about the cytotoxicity of AgNPs. Thus, we have studied the role of autophagy in AgNP-induced cytotoxicity in mouse HC11 mammary epithelium cells. We found that AgNPs injured HC11 cells, with release of lactate dehydrogenase (LDH). AgNPs also induced autophagy in HC11 cells, which was associated with oxidative stress, as indicated by increased reactive oxygen species (ROS) and increased expression of hemoxygenase-1(HO-1) and Nrf2. Mitochondria were altered by AgNPs: mitochondrial membrane potential (MMP) was decreased and the expression of PINK1 and Parkin was increased. AgNPs also increased the expression of p-AMPK and decreased the expression of p-Akt and p-mTOR. The addition of 3-methyl adenine inhibited autophagy and enhanced the cytotoxicity of AgNPs, indicating that autophagy is protective against AgNP-induced cell death. In summary, AgNPs induced protective autophagy in HC11 cells via the Akt/AMPK/mTOR pathway, associated with cellular oxidative stress and mitochondrial alterations. Our research confirms that AgNPs may damage the breast tissue in clinical applications and should be used with caution. Further research is necessary to clarify whether the damage caused by AgNPs will affect the lactation function of the mammary glands and possible residues in milk.
Collapse
Affiliation(s)
- Jin Hou
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Xiaoli He
- College of Science, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Gang Ye
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Fei Shi
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Min Kang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Helin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
44
|
Tarrahi R, Mahjouri S, Khataee A. A review on in vivo and in vitro nanotoxicological studies in plants: A headlight for future targets. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111697. [PMID: 33396028 DOI: 10.1016/j.ecoenv.2020.111697] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/01/2020] [Accepted: 11/18/2020] [Indexed: 06/12/2023]
Abstract
Owing to the unique properties and useful applications in numerous fields, nanomaterials (NMs) received a great attention. The mass production of NMs has raised major concern for the environment. Recently, some altered growth patterns in plants have been reported due to the plant-NMs interactions. However, for NMs safe applications in agriculture and medicine, a comprehensive understanding of bio-nano interactions is crucial. The main goal of this review article is to summarize the results of the toxicological studies that have shown the in vitro and in vivo interactions of NMs with plants. The toxicity mechanisms are briefly discussed in plants as the defense mechanism works to overcome the stress caused by NMs implications. Indeed, the impact of NMs on plants varies significantly with many factors including physicochemical properties of NMs, culture media, and plant species. To investigate the impacts, dose metrics is an important analysis for assaying toxicity and is discussed in the present article to broadly open up different aspects of nanotoxicological investigations. To access reliable quantification and measurement in laboratories, standardized methodologies are crucial for precise dose delivery of NMs to plants during exposure. Altogether, the information is significant to researchers to describe restrictions and future perspectives.
Collapse
Affiliation(s)
- Roshanak Tarrahi
- Health Promotion Research Center, Iran University of Medical Sciences, 14496-14535 Tehran, Iran
| | - Sepideh Mahjouri
- Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran; Рeoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow 117198, Russian Federation.
| |
Collapse
|
45
|
Wu D, Wang S, Yu G, Chen X. Cell Death Mediated by the Pyroptosis Pathway with the Aid of Nanotechnology: Prospects for Cancer Therapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202010281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Dan Wu
- College of Materials Science and Engineering Zhejiang University of Technology Hangzhou 310014 P. R. China
| | - Sheng Wang
- School of Life Sciences Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology Tianjin 300072 P. R. China
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda Maryland 20892 USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda Maryland 20892 USA
- Yong Loo Lin School of Medicine and Faculty of Engineering National University of Singapore Singapore 117597 Singapore
| |
Collapse
|
46
|
Wu D, Wang S, Yu G, Chen X. Cell Death Mediated by the Pyroptosis Pathway with the Aid of Nanotechnology: Prospects for Cancer Therapy. Angew Chem Int Ed Engl 2020; 60:8018-8034. [PMID: 32894628 DOI: 10.1002/anie.202010281] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/18/2020] [Indexed: 12/20/2022]
Abstract
Pyroptosis, a unique form of programmed cell death (PCD) that is characterized by DNA fragmentation, chromatin condensation, cellular swelling with big bubbles, and leakage of cell content, has been proven to have a close relationship with human diseases, such as inflammatory diseases and malignant tumors. Since a new gasdermin-D (GSDMD) protein was identified in 2015, various strategies have been developed to induce pyroptosis for cancer therapy, including ions, small-molecule drugs and nanomaterials. Although there are a number of reviews about the close relationship between the pyroptosis mechanism and the occurrence of various cancers, a summary covering recent progress in the field of nanomedicines in pyroptosis-based cancer therapy has not yet been presented. Therefore, it is urgent to fill this gap and light up future directions for the use of this powerful tool to combat cancer. In this Minireview, recent progress in cancer treatment based on pyroptosis induced by nanoparticles will be described in detail, the design highlights and the therapeutic advantages are emphasized, and future perspectives in this emerging area are proposed.
Collapse
Affiliation(s)
- Dan Wu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Sheng Wang
- School of Life Sciences, Tianjin University and Tianjin Engineering Center of Micro-Nano Biomaterials and Detection-Treatment Technology, Tianjin, 300072, P. R. China
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, 20892, USA.,Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
47
|
Zakeri-Siavashani A, Chamanara M, Nassireslami E, Shiri M, Hoseini-Ahmadabadi M, Paknejad B. Three dimensional spongy fibroin scaffolds containing keratin/vanillin particles as an antibacterial skin tissue engineering scaffold. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1817021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mahdi Shiri
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Babak Paknejad
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Ye G, Bao F, Zhang X, Song Z, Liao Y, Fei Y, Bunpetch V, Heng BC, Shen W, Liu H, Zhou J, Ouyang H. Nanomaterial-based scaffolds for bone tissue engineering and regeneration. Nanomedicine (Lond) 2020; 15:1995-2017. [PMID: 32812486 DOI: 10.2217/nnm-2020-0112] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The global incidence of bone tissue injuries has been increasing rapidly in recent years, making it imperative to develop suitable bone grafts for facilitating bone tissue regeneration. It has been demonstrated that nanomaterials/nanocomposites scaffolds can more effectively promote new bone tissue formation compared with micromaterials. This may be attributed to their nanoscaled structural and topological features that better mimic the physiological characteristics of natural bone tissue. In this review, we examined the current applications of various nanomaterial/nanocomposite scaffolds and different topological structures for bone tissue engineering, as well as the underlying mechanisms of regeneration. The potential risks and toxicity of nanomaterials will also be critically discussed. Finally, some considerations for the clinical applications of nanomaterials/nanocomposites scaffolds for bone tissue engineering are mentioned.
Collapse
Affiliation(s)
- Guo Ye
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Fangyuan Bao
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xianzhu Zhang
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zhe Song
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Youguo Liao
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Yang Fei
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Varitsara Bunpetch
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing, PR China
| | - Weiliang Shen
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Hua Liu
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Jing Zhou
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| | - Hongwei Ouyang
- Dr Li Dak Sum & Yip Yio Chin Center for Stem Cells & Regenerative Medicine & Department of Orthopedic Surgery of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine & Key Laboratory of Tissue Engineering & Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, PR China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, PR China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, PR China
| |
Collapse
|
49
|
Wu C, Xu H, Li J, Hu X, Wang X, Huang Y, Li Y, Sheng S, Wang Y, Xu H, Ni W, Zhou K. Baicalein Attenuates Pyroptosis and Endoplasmic Reticulum Stress Following Spinal Cord Ischemia-Reperfusion Injury via Autophagy Enhancement. Front Pharmacol 2020; 11:1076. [PMID: 32903577 PMCID: PMC7438740 DOI: 10.3389/fphar.2020.01076] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022] Open
Abstract
Background Spinal cord ischemia-reperfusion injury (SCIRI) is the main complication after the repair of a complex thoracoabdominal aortic aneurysm. Many clinical treatments are not ideal due to the complex pathophysiological process of this injury. Baicalein (BA), a component derived from the roots of the herb Scutellaria baicalensis, may contribute to the successful treatment of ischemia/reperfusion injury. Purpose In the present study, the effects of BA on spinal cord ischemia-reperfusion injury and the underlying mechanisms were assessed. Materials and Methods Spinal cord ischemia was induced in C57BL/6 mice by blocking the aortic arch. Fifty-five mice were then randomly divided into four groups: Sham, SCIR+Vehicle, SCIR+BA, and SCIR+BA +3MA groups. At 0 and 24 h pre-SCIRI and at 24 h and 7 days post-SCIRI, evaluations with the Basso mouse scale (BMS) were performed. On postoperative 24 h, the spinal cord was harvested to assess pyroptosis, endoplasmic reticulum stress mediated apoptosis and autophagy. Results BA enhanced the functional recovery of spinal cord ischemia-reperfusion injury. In addition, BA attenuated pyroptosis, alleviated endoplasmic reticulum stress-mediated apoptosis, and activated autophagy. However, the effects of BA on the functional recovery of SCIRI, pyroptosis and endoplasmic reticulum stress-mediated apoptosis were reversed by the inhibition of autophagy. Conclusions In general, our findings revealed that BA enhances the functional recovery of spinal cord ischemia-reperfusion injury by dampening pyroptosis and alleviating endoplasmic reticulum stress-mediated apoptosis, which are mediated by the activation of autophagy.
Collapse
Affiliation(s)
- Chenyu Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Hui Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Jiafeng Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Xinli Hu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Xingyu Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yijia Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Sunren Sheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yongli Wang
- Department of Orthopaedics, Huzhou Central Hospital, Huzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Wenfei Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| |
Collapse
|
50
|
Quan JH, Gao FF, Lee M, Yuk JM, Cha GH, Chu JQ, Wang H, Lee YH. Involvement of endoplasmic reticulum stress response and IRE1-mediated ASK1/JNK/Mcl-1 pathways in silver nanoparticle-induced apoptosis of human retinal pigment epithelial cells. Toxicology 2020; 442:152540. [PMID: 32717251 DOI: 10.1016/j.tox.2020.152540] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
Silver nanoparticles (AgNPs) have cytotoxic effects on various human cell types. The endoplasmic reticulum (ER) is very sensitive to cytotoxic damage. Retina tissue is easily affected by internal and external stimuli. However, the effect of AgNPs on human retinal cells is not known. This study examined the effect of AgNPs on ER stress induction and their mechanism of action in human retinal pigment epithelium (RPE) ARPE-19 cells. We found that AgNPs significantly increased ARPE-19 cell cytotoxicity and stimulated caspase-3 and poly (ADP-ribose) polymerase (PARP) cleavage, as well as mitochondrial membrane potential (MMP) depolarization, in ARPE-19 cells in a dose-dependent manner (0.2-5 μg/mL for 18 h). AgNPs (5 μg/mL for 18 h) induced several signature ER stress markers, as indicated by the upregulated expressions of CCAAT/enhancer-binding protein-homologous protein (CHOP), phosphorylated protein kinase RNA-like ER kinase (PERK), eukaryotic initiation factor 2α (eIF2α), and inositol-requiring protein 1 (IRE1), and cleaved activating transcription factor 6 (ATF6). AgNPs also activated ASK1 and JNK in ARPE-19 cells, and induced increases in Bax and Puma expressions, as well as a decrease in Mcl-1 expression. However, inhibition of the ER stress response by pretreatment with 4-PBA included apparently and dose-dependently reduced levels of p-PERK, p-IRE1, CHOP, cleaved ATF6, p-ASK1, p-JNK, cleaved caspase-3, procaspase-12, and MMP depolarization in AgNP-treated ARPE-19 cells; it also led to significantly increased Mcl-1 protein levels in a dose-dependent manner in ARPE-19 cells. Pretreatment with JNK inhibitor SP600125 significantly attenuated caspase-3 cleavage and MMP depolarization and increased Mcl-1 protein levels in AgNPs-treated ARPE-19 cells in a dose-dependent manner. Hence, our study demonstrated that AgNPs induced apoptosis in human RPE ARPE-19 cells by ER stress response and ER stress-dependent mitochondrial apoptosis via the IRE1/ASK1/JNK/Mcl-1 pathways.
Collapse
Affiliation(s)
- Juan-Hua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Fei Fei Gao
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Mina Lee
- Department of Obstetrics and Gynecology, Chungnam National University, 33, Munhwa-ro, Jung-gu, Deajeon 35015, Republic of Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Guang-Ho Cha
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Jia-Qi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Hao Wang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| | - Young-Ha Lee
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea.
| |
Collapse
|