1
|
Chierichetti M, Cristofani R, Crippa V, Ferrari V, Cozzi M, Casarotto E, Pramaggiore P, Cornaggia L, Patelli G, Mohamed A, Piccolella M, Galbiati M, Rusmini P, Tedesco B, Poletti A. Small heat shock protein B8: from cell functions to its involvement in diseases and potential therapeutic applications. Neural Regen Res 2025; 20:2872-2886. [PMID: 39435632 PMCID: PMC11826450 DOI: 10.4103/nrr.nrr-d-24-00517] [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: 05/06/2024] [Revised: 08/05/2024] [Accepted: 08/29/2024] [Indexed: 10/23/2024] Open
Abstract
Heat shock protein family B (small) member 8 (HSPB8) is a 22 kDa ubiquitously expressed protein belonging to the family of small heat shock proteins. HSPB8 is involved in various cellular mechanisms mainly related to proteotoxic stress response and in other processes such as inflammation, cell division, and migration. HSPB8 binds misfolded clients to prevent their aggregation by assisting protein refolding or degradation through chaperone-assisted selective autophagy. In line with this function, the pro-degradative activity of HSPB8 has been found protective in several neurodegenerative and neuromuscular diseases characterized by protein misfolding and aggregation. In cancer, HSPB8 has a dual role being capable of exerting either a pro- or an anti-tumoral activity depending on the pathways and factors expressed by the model of cancer under investigation. Moreover, HSPB8 exerts a protective function in different diseases by modulating the inflammatory response, which characterizes not only neurodegenerative diseases, but also other chronic or acute conditions affecting the nervous system, such as multiple sclerosis and intracerebellar hemorrhage. Of note, HSPB8 modulation may represent a therapeutic approach in other neurological conditions that develop as a secondary consequence of other diseases. This is the case of cognitive impairment related to diabetes mellitus, in which HSPB8 exerts a protective activity by assuring mitochondrial homeostasis. This review aims to summarize the diverse and multiple functions of HSPB8 in different pathological conditions, focusing on the beneficial effects of its modulation. Drug-based and alternative therapeutic approaches targeting HSPB8 and its regulated pathways will be discussed, emphasizing how new strategies for cell and tissue-specific delivery represent an avenue to advance in disease treatments.
Collapse
Affiliation(s)
- Marta Chierichetti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Riccardo Cristofani
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Valeria Crippa
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Veronica Ferrari
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Marta Cozzi
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Elena Casarotto
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Pramaggiore
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Laura Cornaggia
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Guglielmo Patelli
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Ali Mohamed
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Margherita Piccolella
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Mariarita Galbiati
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Paola Rusmini
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Barbara Tedesco
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| | - Angelo Poletti
- Laboratory of Experimental Biology, Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti” (DiSFeB), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Wang Z, Yang C, Wang X, Lyu W, Liao H, Liu X, Liu H, Zhang J, Shen H, Zhang L, Wang H. Decoding stress granules dynamics: Implications for neurodegenerative disease. Prog Neurobiol 2025; 248:102758. [PMID: 40132681 DOI: 10.1016/j.pneurobio.2025.102758] [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: 01/23/2025] [Revised: 03/01/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Stress granules (SGs) are membrane-less cytoplasmic structures formed by cells in response to external stress, primarily composed of mRNA and proteins. The dynamic properties of their assembly, maintenance, and disassembly play crucial roles in cellular homeostasis. Recent studies have increasingly revealed that aberrations in SGs dynamics are closely related to the pathogenesis of various neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. This review summarizes the latest research progress on SGs dynamics in neurodegenerative diseases. It begins with an overview of the basic biological characteristics of SGs and their functions in neurons, followed by an in-depth exploration of the mechanisms and regulatory pathways of SGs dynamics. The review then summarizes potential therapeutic strategies targeting SGs dynamics abnormalities, particularly through small molecule drugs to modulate SGs formation and disassembly, aiming to delay or halt the progression of neurodegenerative diseases. The review also highlights the application prospects of these interventions in treating neurodegenerative diseases. Finally, the review introduces current techniques used to study SGs dynamics, discussing their advantages, limitations, and future development possibilities. This review aims to provide researchers with a comprehensive perspective to advance the understanding and clinical application of SGs dynamics in the field of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zixuan Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Chenyi Yang
- Nankai University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Xinyi Wang
- Nankai University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Wenyuan Lyu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Qilu Hospital of Shandong University (Qingdao), Qingdao 266000, China
| | - Huihui Liao
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Xing Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Huan Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Jingwei Zhang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Huai Shen
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Lin Zhang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China
| | - Haiyun Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Nankai University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, Tianjin 300170, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin 300170, China.
| |
Collapse
|
3
|
Chen J, Chen H, Wei Q, Lu Y, Wang T, Pang X, Xing G, Chen Z, Cao X, Yao J. APOE4 impairs macrophage lipophagy and promotes demyelination of spiral ganglion neurons in mouse cochleae. Cell Death Discov 2025; 11:190. [PMID: 40258814 PMCID: PMC12012174 DOI: 10.1038/s41420-025-02454-4] [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: 09/23/2024] [Revised: 03/09/2025] [Accepted: 03/27/2025] [Indexed: 04/23/2025] Open
Abstract
The ApoE-ε4 gene is a well-established genetic risk factor for neurodegenerative diseases, such as Alzheimer's disease and multiple sclerosis, which are characterized by axonal demyelination in the central nervous system. Recent studies have implicated ApoE-ε4 in age-related hearing loss (ARHL), suggesting a potential role of APOE4 isoform in peripheral nervous system degeneration. However, the role of APOE4 in ARHL are still unclear. In this study, we explored the potential role of APOE4 in axonal demyelination of spiral ganglion neurons (SGNs). ApoE-ε4/ε4 (APOE4) and ApoE-ε3/ε3 (APOE3) mice were used to characterize SGNs. The effect of APOE4 on phagocytosis and autophagy as well as intracellular cholesterol level was evaluated in resident cochlear macrophages (RCMs) and mouse bone marrow-derived macrophages (BMDMs). The results showed that significant axonal demyelination was observed in SGNs of 10-month-old APOE4 mice, accompanied by the presence of myelin debris engulfed by RCMs. Meanwhile, inhibited phagocytosis of myelin debris and impaired lipophagy were detected in APOE4 RCMs and APOE4 BMDMs with an aberrant accumulation of lipid droplets (LDs), which could be reversed by trehalose treatment. This study provided a deep insight into the pathogenesis of APOE4-induced axonal demyelination of SGNs associated with the impaired lipophagy in RCMs, which helped to elucidate the underlying mechanism of ApoE-ε4 in ARHL.
Collapse
Affiliation(s)
- Junru Chen
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Haibing Chen
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Department of Otolaryngology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Qinjun Wei
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Yajie Lu
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Tianming Wang
- Central Laboratory, Translational Medicine Research Center, the affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuhong Pang
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Guangqian Xing
- Department of Otolaryngology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Zhibin Chen
- Department of Otolaryngology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China.
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.
| | - Jun Yao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China.
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| |
Collapse
|
4
|
Xu Y, McRae DM, Leonenko Z. Quantitative Analysis of the Influence of Trehalose on Amyloid-β Binding to Membranes by Localized Surface Plasmon Resonance Spectroscopy. ACS OMEGA 2025; 10:12872-12879. [PMID: 40224471 PMCID: PMC11983167 DOI: 10.1021/acsomega.4c07038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
The damaging effect of amyloid-β (Aβ) on cellular membranes is an essential factor that contributes to Aβ's neurotoxicity in Alzheimer's disease. In this work, we explore the role of trehalose sugar in protecting model lipid membranes composed of DPPC-POPC-Cholesterol against Aβ toxicity. We used localized surface plasmon resonance (LSPR) spectroscopy and conducted a quantitative analysis to study the influence of trehalose on Aβ-membrane interactions. The LSPR data indicate that trehalose can effectively reduce the level of binding of Aβ to the lipid membrane, indicating its protective role against amyloid toxicity. Additionally, atomic force microscopy (AFM) was used to visualize the lipid membranes supported on the LSPR sensors and to elucidate the effect of trehalose on membrane morphology. The ability of trehalose to alter the physical properties of model membranes is discussed in relation to its protective role against Aβ during dehydration.
Collapse
Affiliation(s)
- Yue Xu
- Department
of Physics & Astronomy, University of
Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Danielle M. McRae
- Department
of Physics & Astronomy, University of
Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Zoya Leonenko
- Department
of Physics & Astronomy, University of
Waterloo, Waterloo, Ontario N2L 3G1, Canada
- Department
of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
- Waterloo
Institute for Nanotechnology, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
5
|
Lemos G, Fernandes CMADS, Watanabe IKM, Delbin MA, Silva FH, Calmasini FB. Trehalose induces bladder smooth muscle hypercontractility in mice: involvement of oxidative stress and cellular senescence. Front Physiol 2025; 16:1572139. [PMID: 40255637 PMCID: PMC12006093 DOI: 10.3389/fphys.2025.1572139] [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: 02/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Autophagy, a conserved catabolic process, is critical for cellular homeostasis and its dysregulation has been implicated in a number of conditions including hypertension, obesity and bladder dysfunctions. The autophagy inducer trehalose has shown promise in treating diseases; however, some studies have reported detrimental effects in vascular tissue under health conditions. In the bladder, the effects of trehalose remain unclear. Therefore, in the present study, male C57BL6/JUnib mice (8 weeks old) were divided into control and trehalose-treated groups (120 mg/mouse/day via gavage) for 4 weeks. After treatment, bladders were harvested for functional, biochemical, and molecular analyses. The trehalose treatment increased the bladder smooth muscle (BSM) contractility to carbachol (CCh), without altering relaxation response to isoproterenol. The CCh-induced BSM hypercontractility was completely abolished by the in vitro incubation of apocynin and diphenyleneiodonium (DPI), implicating NADPH oxidase-derived reactive oxygen species (ROS) on this process. Accordingly, increased levels of superoxide anion (O2-) were found in the urothelial layer, but not in BSM, of trehalose-treated mice. Trehalose also increased senescence-associated β-galactosidase activity in the bladder but failed to upregulate autophagy-related proteins LAMP1 and Beclin-1 in the bladder. Collectively, we show for the first time that trehalose induces BSM hypercontractility in mice, linked to increased levels of O2- and senescent cell, independently of autophagy activation. Therefore, trehalose administration is an effective model for studying BSM hypercontractility in mice, particularly associated with oxidative stress and cellular senescence.
Collapse
Affiliation(s)
- Guilherme Lemos
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Maria Andreia Delbin
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Fábio Henrique Silva
- Laboratory of Multidisciplinary Research, São Francisco University (USF), Sao Paulo, Brazil
| | - Fabiano Beraldi Calmasini
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Rashed HR, Nath SR, Milone M. The Spectrum of Small Heat Shock Protein B8 ( HSPB8)-Associated Neuromuscular Disorders. Int J Mol Sci 2025; 26:2905. [PMID: 40243504 PMCID: PMC11989117 DOI: 10.3390/ijms26072905] [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: 03/03/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The heat shock protein B8 (HSPB8) is one of the small heat shock proteins (sHSP or HSPB) and is a ubiquitous protein in various organisms, including humans. It is highly expressed in skeletal muscle, heart, and neurons. It plays a crucial role in identifying misfolding proteins and participating in chaperone-assisted selective autophagy (CASA) for the removal of misfolded and damaged, potentially cytotoxic proteins. Mutations in HSPB8 can cause distal hereditary motor neuropathy (dHMN), Charcot-Marie-Tooth (CMT) disease type 2L, or myopathy. The disease can manifest from childhood to mid-adulthood. Most missense mutations in the N-terminal and α-crystallin domains of HSPB8 lead to dHMN or CMT2L. Frameshift mutations in the C-terminal domain (CTD), resulting in elongation of the HSPB8 C-terminal, cause myopathy with myofibrillar pathology and rimmed vacuoles. Myopathy and motor neuropathy can coexist. HSPB8 frameshift mutations in the CTD result in HSPB8 mutant aggregation, which weakens the CASA ability to direct misfolded proteins to autophagic degradation. Cellular and animal models indicate that HSPB8 mutations drive pathogenesis through a toxic gain-of-function mechanism. Currently, no cure is available for HSPB8-associated neuromuscular disorders, but numerous therapeutic strategies are under investigation spanning from small molecules to RNA interference to exogenous HSPB8 delivery.
Collapse
Affiliation(s)
- Hebatallah R. Rashed
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (H.R.R.); (S.R.N.)
- Department of Neurology, Ain Shams University, Cairo 11588, Egypt
| | - Samir R. Nath
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (H.R.R.); (S.R.N.)
| | - Margherita Milone
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; (H.R.R.); (S.R.N.)
| |
Collapse
|
7
|
Gu X, Li Y, Li Y, Duan X, Hu Y, Chen J, Du H, Bai J, He C, Bai C, Guo J, Yang J, Hu K. Integrating 2D NMR-based metabolomics and in vitro assays to explore the potential viability of cultivated Ophiocordyceps sinensis as an alternative to the wild counterpart. J Pharm Biomed Anal 2025; 253:116551. [PMID: 39488908 DOI: 10.1016/j.jpba.2024.116551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Ophiocordyceps sinensis is widely used to treat various diseases and as a health supplement. The present study comprehensively compared the metabolic differences between wild and cultivated O. sinensis through 2D 1H-13C HSQC-based metabolomics, and assessed their anti-lung cancer activity on A549 cells. To characterize the global metabolic profile, sample preparation was scrutinously optimized, and both polar (1:4 methanol-water) and non-polar (1:4 methanol-chloroform) extracts of O. sinensis were investigated. A total of 47 and 10 metabolites were identified in the polar and non-polar extracts, respectively. Principal Component Analysis (PCA) revealed greater differences between the two types of O. sinensis in the polar extracts than in the non-polar extracts. Orthogonal Partial Least Squares-Discriminant Analysis (OPLS-DA) together with univariate tests captured 23 and 19 differential spectral features (with 22 and 11 of them assigned) between wild and cultivated O. sinensis in the polar and non-polar extracts, respectively. Meanwhile, the anti-lung cancer activities of both polar and non-polar extracts of wild and cultivated O. sinensis were assessed by MTS assay on A549 cells, and the sterols found in non-polar extracts, such as ergosterol, ergosterol peroxide, and 9,11-dehydroergosterol peroxide, and β-sitosterol, are the active ingredients with potential anti-lung cancer properties. In this study, we introduced a comprehensive strategy integrating 2D NMR-based metabolomics with in vitro assays for comparing the chemical composition and assessing the pharmacological activity of wild and cultivated O. sinensis. Our results provided a scientific basis for the potential viability of cultivated O. sinensis as an alternative to the wild counterpart.
Collapse
Affiliation(s)
- Xiu Gu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Yanping Li
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Yang Li
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, By Sichuan Province and MOST, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Xiaohui Duan
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Youfan Hu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Jialuo Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Huan Du
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Jing Bai
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, By Sichuan Province and MOST, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Chengyan He
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, By Sichuan Province and MOST, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Caihong Bai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Jinlin Guo
- Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, By Sichuan Province and MOST, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Jiahui Yang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Kaifeng Hu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| |
Collapse
|
8
|
Mu Z, Sun Z, Wu S, Yang J, Wang P, Zhao X. Trehalose Inhibits ferroptosis Through Activating SIRT3/SOD2 Signaling Axis and Alleviates Brain Injury After Traumatic Brain Injury. Neurochem Res 2025; 50:78. [PMID: 39798057 DOI: 10.1007/s11064-024-04330-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Trehalose has neuroprotective effects in neurodegenerative diseases. This study aimed to explore the impact of trehalose on traumatic brain injury (TBI) by investigating its role in neuroprotection. The TBI mice model was established utilizing the cortical impact technique followed by trehalose treatment. Traumatic neuronal injury induced by scratch followed by trehalose treatment was performed to mimic TBI in vitro. Memory function was assessed using the Water maze test. Brain damage was evaluated through various methods including brain water content analysis, Nissl staining, Evans blue exudation, and TUNEL staining. Biochemical and morphological changes related to ferroptosis post-TBI were also examined. The results showed that trehalose was found to enhance spatial memory, reduce brain injury, and inhibit ferroptosis in TBI mice, similar to ferroptosis inhibitors. The influence of trehalose on TBI was reversed by the SIRT3 inhibitor. Trehalose upregulated SIRT3 to increase SOD activity in TBI, which could also be counteracted by the SIRT3 inhibitor. Combining trehalose with a ferroptosis inhibitor had a more significant effect on reducing brain injury and inhibiting ferroptosis. Furthermore, in TBI mice treated with trehalose and SIRT3 inhibitors, the effect of trehalose was reversed by SIRT3 inhibitors, but the addition of ferroptosis inhibitors reversed the effect of SIRT3 inhibitors, as shown by decreased ferroptosis and neuronal apoptosis in damaged brain tissue. In summary, this study provides initial evidence that trehalose plays a crucial role in neuroprotection post-TBI through the SIRT3/SOD2 pathway-mediated ferroptosis.
Collapse
Affiliation(s)
- Zhenqian Mu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, China
| | - Zhenlie Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, China
| | - Shuai Wu
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong, 226019, China
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, China
| | - Jieqiong Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, China
| | - Peng Wang
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong, 226019, China
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, China
| | - Xudong Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Neurosurgery, Medical School of Nantong University, Nantong University, Nantong, 226019, China.
- Department of Neurosurgery, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, China.
- Wuxi Neurosurgical Institute, Wuxi, 214002, China.
- , 68 Zhongshan Road, Wuxi, 214000, China.
| |
Collapse
|
9
|
Amabebe E, Huang Z, Jash S, Krishnan B, Cheng S, Nakashima A, Li Y, Li Z, Wang R, Menon R, Zhou XZ, Lu KP, Sharma S. Novel Role of Pin1-Cis P-Tau-ApoE Axis in the Pathogenesis of Preeclampsia and Its Connection with Dementia. Biomedicines 2024; 13:29. [PMID: 39857613 PMCID: PMC11763151 DOI: 10.3390/biomedicines13010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Preeclampsia (preE) is a severe multisystem hypertensive syndrome of pregnancy associated with ischemia/hypoxia, angiogenic imbalance, apolipoprotein E (ApoE)-mediated dyslipidemia, placental insufficiency, and inflammation at the maternal-fetal interface. Our recent data further suggest that preE is associated with impaired autophagy, vascular dysfunction, and proteinopathy/tauopathy disorder, similar to neurodegenerative diseases such as Alzheimer's disease (AD), including the presence of the cis stereo-isoform of phosphorylated tau (cis P-tau), amyloid-β, and transthyretin in the placenta and circulation. This review provides an overview of the factors that may lead to the induction and accumulation of cis P-tau-like proteins by focusing on the inactivation of peptidyl-prolyl cis-trans isomerase (Pin1) that catalyzes the cis to trans isomerization of P-tau. We also highlighted the novel role of the Pin1-cis P-tau-ApoE axis in the development of preE, and propagation of cis P-tau-mediated abnormal protein aggregation (tauopathy) from the placenta to cerebral tissues later in life, leading to neurodegenerative conditions. In the case of preE, proteinopathy/tauopathy may interrupt trophoblast differentiation and induce cell death, similar to the events occurring in neurons. These events may eventually damage the endothelium and cause systemic features of disorders such as preE. Despite impressive research and therapeutic advances in both fields of preE and neurodegenerative diseases, further investigation of Pin1-cis P-tau and ApoE-related mechanistic underpinnings may unravel novel therapeutic options, and new transcriptional and proteomic markers. This review will also cover genetic polymorphisms in the ApoE alleles leading to dyslipidemia induction that may regulate the pathways causing preE or dementia-like features in the reproductive age or later in life, respectively.
Collapse
Affiliation(s)
- Emmanuel Amabebe
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| | - Zheping Huang
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| | - Sukanta Jash
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA;
| | - Shibin Cheng
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama 930-8555, Japan;
| | - Yitong Li
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Zhixong Li
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Ruizhi Wang
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
- Departments of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Lawson Health Research Institute, Western University, London, ON N6A 3K7, Canada
| | - Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Surendra Sharma
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| |
Collapse
|
10
|
Zandi R, Razani HO, Mehrvar A, Jowshan MR, Sahebkar A, Nikooyeh B, Zahedi H, Talebi S. Effects of trehalose on bone healing, physical function, and pain in patients with pertrochanteric fractures: a randomized controlled trial protocol. Trials 2024; 25:823. [PMID: 39695828 DOI: 10.1186/s13063-024-08667-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Appropriate management of fractures is crucial for restoring natural bone function and preventing long-term complications. Previous research on animal models indicates that trehalose can improve bone fracture healing by inhibiting the inflammatory cascade. We hope that trehalose can accelerate bone fracture healing in humans, alleviate pain, and ultimately enhance the individual's quality of life. METHODS This randomized, double-blind clinical trial will be conducted at Taleghani Hospital in Tehran, Iran. Sixty-four patients admitted to the orthopedic ward will be enrolled based on eligibility criteria. The participants will be randomly allocated based on the permuted block randomization into two groups: those receiving trehalose (32 patients) or placebo (32 patients). The patients in the trehalose and placebo groups will receive 3.3 g of trehalose or placebo for 12 weeks, respectively. A consent form, general questionnaire, as well as the Visual Analog Scale (VAS), Harris Hip Score (HHS), and radiological analyses will be used to assess fracture healing quality. The intention-to-treat principle will form the basis of the statistical analysis. DISCUSSION The trial results may provide a convenient and safe adjuvant treatment option for the Pertrochanteric Fractures population. TRIAL REGISTRATION Iranian Registry of Clinical Trials. IRCT20240605062013N1. URL of the trial registry record: https://irct.behdasht.gov.ir/trial/77212 . Registration date: 16 June 2024.
Collapse
Affiliation(s)
- Reza Zandi
- Department of Orthopedics, School of Medicine, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Musculoskeletal Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hosna Omidi Razani
- Student Research Committee, Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Mehrvar
- Department of Orthopedics, School of Medicine, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Jowshan
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Bahar Nikooyeh
- Department of Nutrition Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hoda Zahedi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Shahin Talebi
- Department of Orthopedics, School of Medicine, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Qian S, Long Y, Tan G, Li X, Xiang B, Tao Y, Xie Z, Zhang X. Programmed cell death: molecular mechanisms, biological functions, diseases, and therapeutic targets. MedComm (Beijing) 2024; 5:e70024. [PMID: 39619229 PMCID: PMC11604731 DOI: 10.1002/mco2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Programmed cell death represents a precisely regulated and active cellular demise, governed by a complex network of specific genes and proteins. The identification of multiple forms of programmed cell death has significantly advanced the understanding of its intricate mechanisms, as demonstrated in recent studies. A thorough grasp of these processes is essential across various biological disciplines and in the study of diseases. Nonetheless, despite notable progress, the exploration of the relationship between programmed cell death and disease, as well as its clinical application, are still in a nascent stage. Therefore, further exploration of programmed cell death and the development of corresponding therapeutic methods and strategies holds substantial potential. Our review provides a detailed examination of the primary mechanisms behind apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. Following this, the discussion delves into biological functions and diseases associated dysregulated programmed cell death. Finally, we highlight existing and potential therapeutic targets and strategies focused on cancers and neurodegenerative diseases. This review aims to summarize the latest insights on programmed cell death from mechanisms to diseases and provides a more reliable approach for clinical transformation.
Collapse
Affiliation(s)
- Shen'er Qian
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Yao Long
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Guolin Tan
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Xiaoguang Li
- Department of Otolaryngology Head and Neck SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear InstituteShanghai Jiao Tong University School of Medicine, Shanghai Key LabShanghaiChina
| | - Bo Xiang
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Furong LaboratoryCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaowei Zhang
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
12
|
Zhou Y, Nan F, Zhang Q, Xu W, Fang S, Liu K, Zhao B, Han H, Xie X, Qin C, Pang X. Natural products that alleviate depression: The putative role of autophagy. Pharmacol Ther 2024; 264:108731. [PMID: 39426604 DOI: 10.1016/j.pharmthera.2024.108731] [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: 02/29/2024] [Revised: 08/04/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Major depressive disorder (MDD) is a common mental disorder that severely disrupts psychosocial function and decreases the quality of life. Although the pathophysiological mechanism underlying MDD is complex and remains unclear, emerging evidence suggests that autophagy dysfunction plays a role in MDD occurrence and progression. Natural products serve as a major source of drug discovery and exert tremendous potential in developing antidepressants. Recently published reports are paying more attention on the autophagy regulatory effect of antidepressant natural products. In this review, we comprehensively discuss the abnormal changes occurred in multiple autophagy stages in MDD patients, and animal and cell models of depression. Importantly, we emphasize the regulatory mechanism of antidepressant natural products on disturbed autophagy, including monomeric compounds, bioactive components, crude extracts, and traditional Chinese medicine formulae. Our comprehensive review suggests that enhancing autophagy might be a novel approach for MDD treatment, and natural products restore autophagy homeostasis to facilitate the renovation of mitochondria, impede neuroinflammation, and enhance neuroplasticity, thereby alleviating depression.
Collapse
Affiliation(s)
- Yunfeng Zhou
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Fengwei Nan
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Qianwen Zhang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Wangjun Xu
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Shaojie Fang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Ke Liu
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Bingxin Zhao
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Hao Han
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xinmei Xie
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| | - Changjiang Qin
- Huaihe Hospital of Henan University, Kaifeng 475000, China.
| | - Xiaobin Pang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
13
|
Zheng Y, Zhou Z, Liu M, Chen Z. Targeting selective autophagy in CNS disorders by small-molecule compounds. Pharmacol Ther 2024; 263:108729. [PMID: 39401531 DOI: 10.1016/j.pharmthera.2024.108729] [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: 02/22/2024] [Revised: 09/25/2024] [Accepted: 10/04/2024] [Indexed: 10/27/2024]
Abstract
Autophagy functions as the primary cellular mechanism for clearing unwanted intracellular contents. Emerging evidence suggests that the selective elimination of intracellular organelles through autophagy, compared to the increased bulk autophagic flux, is crucial for the pathological progression of central nervous system (CNS) disorders. Notably, autophagic removal of mitochondria, known as mitophagy, is well-understood in an unhealthy brain. Accumulated data indicate that selective autophagy of other substrates, including protein aggregates, liposomes, and endoplasmic reticulum, plays distinctive roles in various pathological stages. Despite variations in substrates, the molecular mechanisms governing selective autophagy can be broadly categorized into two types: ubiquitin-dependent and -independent pathways, both of which can be subjected to regulation by small-molecule compounds. Notably, natural products provide the remarkable possibility for future structural optimization to regulate the highly selective autophagic clearance of diverse substrates. In this context, we emphasize the selectivity of autophagy in regulating CNS disorders and provide an overview of chemical compounds capable of modulating selective autophagy in these disorders, along with the underlying mechanisms. Further exploration of the functions of these compounds will in turn advance our understanding of autophagic contributions to brain disorders and illuminate precise therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Zhuchen Zhou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Mengting Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
14
|
Stevanovic D, Vucicevic L, Misirkic-Marjanovic M, Martinovic T, Mandic M, Harhaji-Trajkovic L, Trajkovic V. Trehalose Attenuates In Vitro Neurotoxicity of 6-Hydroxydopamine by Reducing Oxidative Stress and Activation of MAPK/AMPK Signaling Pathways. Int J Mol Sci 2024; 25:10659. [PMID: 39408988 PMCID: PMC11476739 DOI: 10.3390/ijms251910659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
The effects of trehalose, an autophagy-inducing disaccharide with neuroprotective properties, on the neurotoxicity of parkinsonian mimetics 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenylpiridinium (MPP+) are poorly understood. In our study, trehalose suppressed 6-OHDA-induced caspase-3/PARP1 cleavage (detected by immunoblotting), apoptotic DNA fragmentation/phosphatidylserine externalization, oxidative stress, mitochondrial depolarization (flow cytometry), and mitochondrial damage (electron microscopy) in SH-SY5Y neuroblastoma cells. The protection was not mediated by autophagy, autophagic receptor p62, or antioxidant enzymes superoxide dismutase and catalase. Trehalose suppressed 6-OHDA-induced activation of c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), and AMP-activated protein kinase (AMPK), as revealed by immunoblotting. Pharmacological/genetic inhibition of JNK, p38 MAPK, or AMPK mimicked the trehalose-mediated cytoprotection. Trehalose did not affect the extracellular signal-regulated kinase (ERK) and mechanistic target of rapamycin complex 1 (mTORC1)/4EBP1 pathways, while it reduced the prosurvival mTORC2/AKT signaling. Finally, trehalose enhanced oxidative stress, mitochondrial damage, and apoptosis without decreasing JNK, p38 MAPK, AMPK, or AKT activation in SH-SY5Y cells exposed to MPP+. In conclusion, trehalose protects SH-SY5Y cells from 6-OHDA-induced oxidative stress, mitochondrial damage, and apoptosis through autophagy/p62-independent inhibition of JNK, p38 MAPK, and AMPK. The opposite effects of trehalose on the neurotoxicity of 6-OHDA and MPP+ suggest caution in its potential development as a neuroprotective agent.
Collapse
Affiliation(s)
- Danijela Stevanovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia; (D.S.)
| | - Ljubica Vucicevic
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (M.M.-M.)
| | - Maja Misirkic-Marjanovic
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (M.M.-M.)
| | - Tamara Martinovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia; (D.S.)
| | - Milos Mandic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia; (D.S.)
| | - Ljubica Harhaji-Trajkovic
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia; (M.M.-M.)
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotica 1, 11000 Belgrade, Serbia; (D.S.)
| |
Collapse
|
15
|
Kaur A, Singh S, Sharma SC. Unlocking Trehalose's versatility: A comprehensive Journey from biosynthesis to therapeutic applications. Exp Cell Res 2024; 442:114250. [PMID: 39260672 DOI: 10.1016/j.yexcr.2024.114250] [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: 07/20/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
For over forty years, a sugar of rare configuration known as trehalose (two molecules of glucose linked at their 1-carbons), has been recognised for more than just its roles as a storage compound. The ability of trehalose to protect an extensive range of biological materials, for instance cell lines, tissues, proteins and DNA, has sparked considerable interest in the biotechnology and pharmaceutical industries. Currently, trehalose is now being investigated as a promising therapeutic candidate for human use, as it has shown potential to reduce disease severity in various experimental models. Despite its diverse biological effects, the precise mechanism underlying this observation remain unclear. Therefore, this review delves into the significance of trehalose biosynthesis pathway in the development of novel drug, investigates the inhibitors of trehalose synthesis and evaluates the binding efficiency of T6P with TPS1. Additionally, it also emphasizes the knowledge about the protective effect of trehalose on modulation of autophagy, combating viral infections, addressing the conditions like cancer and neurodegenerative diseases based on the recent advancement. Furthermore, review also highlight the trehalose's emerging role as a surfactant in delivering monoclonal antibodies that will further broadening its potential application in biomedicines.
Collapse
Affiliation(s)
- Amandeep Kaur
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India.
| | - Sukhwinder Singh
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India.
| | | |
Collapse
|
16
|
Barmaki H, Nourazarian A, Shademan B, Khaki-Khatibi F. The autophagy paradox: A new hypothesis in neurodegenerative disorders. Neurochem Int 2024; 179:105827. [PMID: 39111406 DOI: 10.1016/j.neuint.2024.105827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
A recent study showed that while autophagy is usually tied to protein and organelle turnover, it can also play dual roles in neurodegenerative diseases. Traditionally, autophagy was seen as protective since it removes damaged proteins and organelles. but new data suggests autophagy can sometimes promote neuron death. and This review tackles autophagy's seemingly contradictory effects in neurodegeneration, or the "autophagy paradox. " It offers a framework for understanding autophagy in neurodegenerative research and the cellular processes involved. In short, our data uncovers a harmful autophagy role in certain situations, conflicting the view that it's always beneficial. We describe the distinct, disease-specific autophagy pathways functioning in various neurodegenerative diseases. Part two concerns potential therapeutic implications of manipulating autophagy and current strategies targeting the autophagic system, suggesting interesting areas for future research into tailored modulators. This could eventually enable activating or controlling specific autophagy pathways and aid in developing more effective treatments. Researchers believe more molecular-level research is needed so patient-tailored autophagy-modulating therapeutics can be developed given this dilemma. Moreover, research must translate faster into effective neurodegenerative disease treatment options. This article aims to provide a wholly new perspective on autophagy's classically described role in these severe diseases, challenging current dogma and opening new therapeutic avenue options.
Collapse
Affiliation(s)
- Haleh Barmaki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran; Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran.
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
17
|
Wang Z, Wang M, Huang Y, Ma Z, Gao W, Zhang T, Deng J, Cheng X, Liu Y, Wang B, Qi Y, Yang M, He F. Trehalose prevents the formation of aggregates of mutant ataxin-3 and reduces soluble ataxin-3 protein levels in an SCA3 cell model. Neuroscience 2024; 555:76-82. [PMID: 38964453 DOI: 10.1016/j.neuroscience.2024.06.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is a neurodegenerative disorder caused by mutant ataxin-3 with an abnormally expanded polyQ tract and is the most common dominantly inherited ataxia worldwide. There are no suitable therapeutic options for this disease. Autophagy, a defense mechanism against the toxic effects of aggregation-prone misfolded proteins, has been shown to have beneficial effects on neurodegenerative diseases. Thus, trehalose, which is an autophagy inducer, may have beneficial effects on SCA3. In the present study, we examined the effects of trehalose on an SCA3 cell model. After trehalose treatment, aggregate formation, soluble ataxin-3 protein levels and cell viability were evaluated in HEK293T cells overexpressing ataxin-3-15Q or ataxin-3-77Q. We also explored the mechanism by which trehalose affects autophagy and stress pathways. A filter trap assay showed that trehalose decreased the number of aggregates formed by mutant ataxin-3 containing an expanded polyQ tract. Western blot and Cell Counting Kit-8 (CCK-8) results demonstrated that trehalose also reduced the ataxin-3 protein levels and was safe for ataxin-3-expressing cells, respectively. Western blot and total antioxidant capacity assays suggested that trehalose had great therapeutic potential for treating SCA3, likely through its antioxidant activity. Our data indicate that trehalose plays a neuroprotective role in SCA3 by inhibiting the aggregation and reducing the protein level of ataxin-3, which is also known to protect against oxidative stress. These findings provide a new insight into the possibility of treating SCA3 with trehalose and highlight the importance of inducing autophagy in SCA3.
Collapse
Affiliation(s)
- Zijian Wang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China.
| | - Min Wang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Yuhang Huang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Zhiwei Ma
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Wenjing Gao
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Tian Zhang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Jiexin Deng
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Xiaoxia Cheng
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Yingxun Liu
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Bo Wang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Ying Qi
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Min Yang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| | - Fengqin He
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an University, Xi'an 710065, Shaanxi, China
| |
Collapse
|
18
|
Cui Q, Liu Z, Bai G. Friend or foe: The role of stress granule in neurodegenerative disease. Neuron 2024; 112:2464-2485. [PMID: 38744273 DOI: 10.1016/j.neuron.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/12/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024]
Abstract
Stress granules (SGs) are dynamic membraneless organelles that form in response to cellular stress. SGs are predominantly composed of RNA and RNA-binding proteins that assemble through liquid-liquid phase separation. Although the formation of SGs is considered a transient and protective response to cellular stress, their dysregulation or persistence may contribute to various neurodegenerative diseases. This review aims to provide a comprehensive overview of SG physiology and pathology. It covers the formation, composition, regulation, and functions of SGs, along with their crosstalk with other membrane-bound and membraneless organelles. Furthermore, this review discusses the dual roles of SGs as both friends and foes in neurodegenerative diseases and explores potential therapeutic approaches targeting SGs. The challenges and future perspectives in this field are also highlighted. A more profound comprehension of the intricate relationship between SGs and neurodegenerative diseases could inspire the development of innovative therapeutic interventions against these devastating diseases.
Collapse
Affiliation(s)
- Qinqin Cui
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China.
| | - Zongyu Liu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ge Bai
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
19
|
Sahranavard M, Hosseinjani H, Emadzadeh M, Jamialahmadi T, Sahebkar A. Effect of trehalose on mortality and disease severity in ICU-admitted patients: Protocol for a triple-blind, randomized, placebo-controlled clinical trial. Contemp Clin Trials Commun 2024; 40:101324. [PMID: 39021672 PMCID: PMC11252791 DOI: 10.1016/j.conctc.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/04/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Background Improvement in organ failure in intensive care unit (ICU) patients is accompanied by lower mortality rate. A disaccharide, trehalose is a candidate to improve organ failure and survival by autophagy induction and enhancing oxidative stress defense. The aim of this study is to assess the effectiveness of trehalose in improving clinical outcome and reducing mortality in ICU patients. Methods a triple-blind, randomized, placebo-controlled, two arm, parallel-group, superiority clinical trial will enroll 200 ICU-admitted patients at Imam Reza hospital, Mashhad, Iran. The patients will be randomly allocated to receive either a 100 ml solution of 15 % trehalose or normal saline intravenously. Primary outcomes include ICU mortality and 60-day mortality, while secondary outcomes focus on blood parameters on day 5 and length of hospital/ICU stay. Conclusion Trehalose has demonstrated beneficial effects in diverse patients; however, no study has evaluated its effect in all ICU-admitted patients. Consequently, this study provides an opportunity to investigate whether trehalose's anti-inflammatory effects, mediated by inducing autophagy and enhancing oxidative stress defense, can play a role in reducing mortality and improving clinical outcomes in the critically ill patients. If successful, trehalose could offer a potential therapeutic approach in the ICU setting.
Collapse
Affiliation(s)
- Mehrdad Sahranavard
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hesamoddin Hosseinjani
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Emadzadeh
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Ghosh S, Sharma R, Bammidi S, Koontz V, Nemani M, Yazdankhah M, Kedziora KM, Stolz DB, Wallace CT, Yu-Wei C, Franks J, Bose D, Shang P, Ambrosino HM, Dutton JR, Geng Z, Montford J, Ryu J, Rajasundaram D, Hose S, Sahel JA, Puertollano R, Finkel T, Zigler JS, Sergeev Y, Watkins SC, Goetzman ES, Ferrington DA, Flores-Bellver M, Kaarniranta K, Sodhi A, Bharti K, Handa JT, Sinha D. The AKT2/SIRT5/TFEB pathway as a potential therapeutic target in non-neovascular AMD. Nat Commun 2024; 15:6150. [PMID: 39034314 PMCID: PMC11271488 DOI: 10.1038/s41467-024-50500-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 07/10/2024] [Indexed: 07/23/2024] Open
Abstract
Non-neovascular or dry age-related macular degeneration (AMD) is a multi-factorial disease with degeneration of the aging retinal-pigmented epithelium (RPE). Lysosomes play a crucial role in RPE health via phagocytosis and autophagy, which are regulated by transcription factor EB/E3 (TFEB/E3). Here, we find that increased AKT2 inhibits PGC-1α to downregulate SIRT5, which we identify as an AKT2 binding partner. Crosstalk between SIRT5 and AKT2 facilitates TFEB-dependent lysosomal function in the RPE. AKT2/SIRT5/TFEB pathway inhibition in the RPE induced lysosome/autophagy signaling abnormalities, disrupted mitochondrial function and induced release of debris contributing to drusen. Accordingly, AKT2 overexpression in the RPE caused a dry AMD-like phenotype in aging Akt2 KI mice, as evident from decline in retinal function. Importantly, we show that induced pluripotent stem cell-derived RPE encoding the major risk variant associated with AMD (complement factor H; CFH Y402H) express increased AKT2, impairing TFEB/TFE3-dependent lysosomal function. Collectively, these findings suggest that targeting the AKT2/SIRT5/TFEB pathway may be an effective therapy to delay the progression of dry AMD.
Collapse
Affiliation(s)
- Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sridhar Bammidi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victoria Koontz
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mihir Nemani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meysam Yazdankhah
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katarzyna M Kedziora
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Donna Beer Stolz
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Callen T Wallace
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cheng Yu-Wei
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan Franks
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Devika Bose
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - James R Dutton
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Zhaohui Geng
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Jair Montford
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiwon Ryu
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institut De La Vision, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Samuel Zigler
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuri Sergeev
- Protein Biochemistry & Molecular Modeling Group, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Simon C Watkins
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric S Goetzman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deborah A Ferrington
- Doheny Eye Institute, Pasadena, CA, USA
- Department of Ophthalmology, University of California Los Angeles, Los Angeles, CA, USA
| | - Miguel Flores-Bellver
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Akrit Sodhi
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, National Institutes of Health, Bethesda, MD, USA.
| | - James T Handa
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Magalhães RSS, Monteiro Neto JR, Ribeiro GD, Paranhos LH, Eleutherio ECA. Trehalose Protects against Superoxide Dismutase 1 Proteinopathy in an Amyotrophic Lateral Sclerosis Model. Antioxidants (Basel) 2024; 13:807. [PMID: 39061876 PMCID: PMC11274086 DOI: 10.3390/antiox13070807] [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: 06/04/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
This work aimed to study the effect of trehalose in protecting cells against Sod1 proteinopathy associated with amyotrophic lateral sclerosis (ALS). Humanized yeast cells in which native Sod1 was replaced by wild-type human Sod1 or an ALS mutant (WT-A4V Sod1 heterodimer) were used as the experimental model. Cells were treated with 10% trehalose (p/v) before or after the appearance of hSod1 proteinopathy induced by oxidative stress. In both conditions, trehalose reduced the number of cells with Sod1 inclusions, increased Sod1 activity, and decreased the levels of intracellular oxidation, demonstrating that trehalose avoids Sod1 misfolding and loss of function in response to oxidative stress. The survival rates of ALS Sod1 cells stressed in the presence of trehalose were 60% higher than in their absence. Treatment with trehalose after the appearance of Sod1 inclusions in cells expressing WT Sod1 doubled longevity; after 5 days, non-treated cells did not survive, but 15% of cells treated with sugar were still alive. Altogether, our results emphasize the potential of trehalose as a novel therapy, which might be applied preventively in ALS patients with a family history of the disease or after diagnosis in ALS patients who discover the disease following the first symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Elis C. A. Eleutherio
- Institute of Chemistry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (R.S.S.M.); (J.R.M.N.); (G.D.R.); (L.H.P.)
| |
Collapse
|
22
|
Marchese M, Bernardi S, Ogi A, Licitra R, Silvi G, Mero S, Galatolo D, Gammaldi N, Doccini S, Ratto GM, Rapposelli S, Neuhauss SCF, Zang J, Rocchiccioli S, Michelucci E, Ceccherini E, Santorelli FM. Targeting autophagy impairment improves the phenotype of a novel CLN8 zebrafish model. Neurobiol Dis 2024; 197:106536. [PMID: 38763444 PMCID: PMC11163972 DOI: 10.1016/j.nbd.2024.106536] [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: 02/16/2024] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024] Open
Abstract
CLN8 is an endoplasmic reticulum cargo receptor and a regulator of lysosome biogenesis whose loss of function leads to neuronal ceroid lipofuscinosis. CLN8 has been linked to autophagy and lipid metabolism, but much remains to be learned, and there are no therapies acting on the molecular signatures in this disorder. The present study aims to characterize the molecular pathways involved in CLN8 disease and, by pinpointing altered ones, to identify potential therapies. To bridge the gap between cell and mammalian models, we generated a new zebrafish model of CLN8 deficiency, which recapitulates the pathological features of the disease. We observed, for the first time, that CLN8 dysfunction impairs autophagy. Using autophagy modulators, we showed that trehalose and SG2 are able to attenuate the pathological phenotype in mutant larvae, confirming autophagy impairment as a secondary event in disease progression. Overall, our successful modeling of CLN8 defects in zebrafish highlights this novel in vivo model's strong potential as an instrument for exploring the role of CLN8 dysfunction in cellular pathways, with a view to identifying small molecules to treat this rare disease.
Collapse
Affiliation(s)
- Maria Marchese
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy.
| | - Sara Bernardi
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy; Department of Biology, University of Pisa, Pisa, Italy
| | - Asahi Ogi
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Rosario Licitra
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Giada Silvi
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Serena Mero
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Daniele Galatolo
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Nicola Gammaldi
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy; Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Stefano Doccini
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy
| | - Gian Michele Ratto
- National Enterprise for NanoScience and NanoTechnology (NEST), Istituto Nanoscienze, Consiglio Nazionale delle Ricerche (CNR) and Scuola Normale Superiore, Pisa, Italy
| | | | - Stephan C F Neuhauss
- University of Zurich, Department of Molecular Life Sciences, Zurich, Switzerland
| | - Jingjing Zang
- University of Zurich, Department of Molecular Life Sciences, Zurich, Switzerland
| | | | - Elena Michelucci
- Institute of Clinical Physiology, National Research Council, Pisa, Italy; Institute of Chemistry of Organometallic Compounds, National Research Council, Pisa, Italy
| | - Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Filippo M Santorelli
- Department of Neurobiology and Molecular Medicine, IRCCS Fondazione Stella Maris, Calambrone, Pisa, Italy.
| |
Collapse
|
23
|
Xu Y, Filice CT, Leonenko Z. Protective effect of trehalose sugar on amyloid-membrane interactions using BLM electrophysiology. Biophys J 2024; 123:1690-1704. [PMID: 38751113 PMCID: PMC11213996 DOI: 10.1016/j.bpj.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/10/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by dementia and memory loss in the elderly population. The amyloid-β peptide (Aβ) is one of the main pathogenic factors in AD and is known to cause damage to neuronal cellular membranes. There is no cure currently available for AD, and new approaches, including preventive strategies, are highly desirable. In this work, we explore the possibility of protecting neuronal membranes from amyloid-induced damage with naturally existing sugar trehalose. Trehalose has been shown to protect plant cellular membranes in extreme conditions and modify Aβ misfolding. We hypothesize that trehalose can protect the neuronal membrane from amyloid toxicity. In this work, we studied the protective effect of trehalose against Aβ1-42-induced damage in model lipid membranes (DPPC/POPC/cholesterol) using atomic force microscopy and black lipid membrane electrophysiology. Our results demonstrate that Aβ1-42 damaged membranes and led to ionic current leakage across these membranes due to the formation of various defects and pores. The presence of trehalose reduced the ion current across membranes caused by Aβ1-42 peptide damage, thus efficiently protecting the membranes. These findings suggest that the trehalose sugar can potentially be useful in protecting neuronal membranes against amyloid toxicity in AD.
Collapse
Affiliation(s)
- Yue Xu
- Department of Physics & Astronomy, University of Waterloo, Waterloo, ON, Canada
| | - Carina Teresa Filice
- Department of Biology, University of Waterloo, Waterloo, ON, Canada; Waterloo Institute for Nanotechnology, Waterloo, ON, Canada
| | - Zoya Leonenko
- Department of Physics & Astronomy, University of Waterloo, Waterloo, ON, Canada; Department of Biology, University of Waterloo, Waterloo, ON, Canada; Waterloo Institute for Nanotechnology, Waterloo, ON, Canada.
| |
Collapse
|
24
|
Ghorbani M, Abouei Mehrizi M, Tajvidi M, Amin Habibi M, Mohammadi M, Esmaeilian S, Torabi P, Rahmanipour E, Daskareh M, Mohammadi A. Trehalose: A promising new treatment for traumatic brain injury? A systematic review of animal evidence. INTERDISCIPLINARY NEUROSURGERY 2024; 36:101947. [DOI: 10.1016/j.inat.2023.101947] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2024] Open
|
25
|
Zozulya SA, Akopyan AA, Tenditnik MV, Ovsyukova MV, Korolenko TA, Klyushnik TP, Tikhonova MA, Pupyshev AB. Effect of Trehalose Disaccharide on Activation of Microglia and Indices of Systemic Inflammation in an Experimental Model of Alzheimer's Disease. Bull Exp Biol Med 2024; 177:207-211. [PMID: 39090471 DOI: 10.1007/s10517-024-06157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Indexed: 08/04/2024]
Abstract
In an experimental model of Alzheimer's disease in mice, oral administration of trehalose disaccharide reduces neuroinflammation assessed by the expression level of microglia activation marker Iba1 and affects the neutrophil degranulation activity. A potential anti-inflammatory effect of 4% trehalose solution associated with a decrease in the activity of leukocyte elastase in plasma was revealed.
Collapse
Affiliation(s)
- S A Zozulya
- Mental Health Research Center, Moscow, Russia.
| | - A A Akopyan
- Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - M V Tenditnik
- Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - M V Ovsyukova
- Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - T A Korolenko
- Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | | | - M A Tikhonova
- Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - A B Pupyshev
- Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| |
Collapse
|
26
|
Huang Z, Cheng S, Jash S, Fierce J, Agudelo A, Higashiyama T, Hanna N, Nakashima A, Saito S, Padbury J, Schuster J, Sharma S. Exploiting sweet relief for preeclampsia by targeting autophagy-lysosomal machinery and proteinopathy. Exp Mol Med 2024; 56:1206-1220. [PMID: 38760513 PMCID: PMC11148015 DOI: 10.1038/s12276-024-01234-x] [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: 08/25/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 05/19/2024] Open
Abstract
The etiology of preeclampsia (PE), a severe complication of pregnancy with several clinical manifestations and a high incidence of maternal and fetal morbidity and mortality, remains unclear. This issue is a major hurdle for effective treatment strategies. We recently demonstrated that PE exhibits an Alzheimer-like etiology of impaired autophagy and proteinopathy in the placenta. Targeting of these pathological pathways may be a novel therapeutic strategy for PE. Stimulation of autophagy with the natural disaccharide trehalose and its lacto analog lactotrehalose in hypoxia-exposed primary human trophoblasts restored autophagy, inhibited the accumulation of toxic protein aggregates, and restored the ultrastructural features of autophagosomes and autolysosomes. Importantly, trehalose and lactotrehalose inhibited the onset of PE-like features in a humanized mouse model by normalizing autophagy and inhibiting protein aggregation in the placenta. These disaccharides restored the autophagy-lysosomal biogenesis machinery by increasing nuclear translocation of the master transcriptional regulator TFEB. RNA-seq analysis of the placentas of mice with PE indicated the normalization of the PE-associated transcriptome profile in response to trehalose and lactotrehalose. In summary, our results provide a novel molecular rationale for impaired autophagy and proteinopathy in patients with PE and identify treatment with trehalose and its lacto analog as promising therapeutic options for this severe pregnancy complication.
Collapse
Affiliation(s)
- Zheping Huang
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Shibin Cheng
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Sukanta Jash
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Jamie Fierce
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Anthony Agudelo
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | | | - Nazeeh Hanna
- Division of Neonatology, Department of Pediatrics, New York University Long Island School of Medicine, Mineola, New York, NY, USA
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Shigeru Saito
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - James Padbury
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Jessica Schuster
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, 02905, USA.
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
27
|
Zhang S, Yin H, Zhang Y, Zhu Y, Zhu X, Zhu W, Tang L, Liu Y, Wu K, Zhao B, Tian Y, Lu H. Autophagic-lysosomal damage induced by swainsonine is protected by trehalose through activation of TFEB-regulated pathway in renal tubular epithelial cells. Chem Biol Interact 2024; 394:110990. [PMID: 38579922 DOI: 10.1016/j.cbi.2024.110990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Swainsonine (SW) is the main toxic component of locoweed. Previous studies have shown that kidney damage is an early pathologic change in locoweed poisoning in animals. Trehalose induces autophagy and alleviates lysosomal damage, while its protective effect and mechanism against the toxic injury induced by SW is not clear. Based on the published literature, we hypothesize that transcription factor EB(TFEB) -regulated is targeted by SW and activating TFEB by trehalose would reverse the toxic effects. In this study, we investigate the mechanism of protective effects of trehalose using renal tubular epithelial cells. The results showed that SW induced an increase in the expression level of microtubule-associated protein light chain 3-II and p62 proteins and a decrease in the expression level of ATPase H+ transporting V1 Subunit A, Cathepsin B, Cathepsin D, lysosome-associated membrane protein 2 and TFEB proteins in renal tubular epithelial cells in a time and dose-dependent manner suggesting TFEB-regulated lysosomal pathway is adversely affected by SW. Conversely, treatment with trehalose, a known activator of TFEB promote TFEB nuclear translocation suggesting that TFEB plays an important role in protection against SW toxicity. We demonstrated in lysosome staining that SW reduced the number of lysosomes and increased the luminal pH, while trehalose could counteract these SW-induced effects. In summary, our results demonstrated for the first time that trehalose could alleviate the autophagy degradation disorder and lysosomal damage induced by SW. Our results provide an interesting method for reversion of SW-induced toxicity in farm animals and furthermore, activation of TFEB by trehalose suggesting novel mechanism of treating lysosomal storage diseases.
Collapse
Affiliation(s)
- Shuhang Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hai Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yiqingqing Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yanli Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xueyao Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Wenting Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Lihui Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yiling Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Kexin Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Hao Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
28
|
Maycotte P, Illanes M, Moreno DA. Glucosinolates, isothiocyanates, and their role in the regulation of autophagy and cellular function. PHYTOCHEMISTRY REVIEWS 2024. [DOI: 10.1007/s11101-024-09944-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/27/2024] [Indexed: 01/04/2025]
|
29
|
Shi F, Gao YS, Han SM, Huang CS, Hou QS, Wen XW, Wang BS, Zhu ZY, Zou L. Allulose mitigates chronic enteritis by reducing mitochondria dysfunction via regulating cathepsin B production. Int Immunopharmacol 2024; 129:111645. [PMID: 38354512 DOI: 10.1016/j.intimp.2024.111645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
Metabolic changes have been linked to the development of inflammatory bowel disease (IBD), which includes colitis. Allulose, an endogenous bioactive monosaccharide, is vital to the synthesis of numerous compounds and metabolic processes within living organisms. Nevertheless, the precise biochemical mechanism by which allulose inhibits colitis remains unknown. Allulose is an essential and intrinsic protector of the intestinal mucosal barrier, as it maintains the integrity of tight junctions in the intestines, according to the current research. It is also important to know that there is a link between the severity of inflammatory bowel disease (IBD) and colorectal cancer (CRC), chemically-induced colitis in rodents, and lower levels of allulose in the blood. Mice with colitis, either caused by dextran sodium sulphate (DSS) or naturally occurring colitis in IL-10-/- mice, had less damage to their intestinal mucosa after being given allulose. Giving allulose to a colitis model starts a chain of reactions because it stops cathepsin B from ejecting and helps lysosomes stick together. This system effectively stops the activity of myosin light chain kinase (MLCK) when intestinal epithelial damage happens. This stops the breakdown of tight junction integrity and the start of mitochondrial dysfunction. To summarise, the study's findings have presented data that supports the advantageous impact of allulose in reducing the advancement of colitis. Its ability to stop the disruption of the intestinal barrier enables this. Therefore, allulose has potential as a medicinal supplement for treating colitis.
Collapse
Affiliation(s)
- Fang Shi
- Department of Abdominal Radiotherapy, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 250117, China
| | - Yong-Sheng Gao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 250117, China
| | - Shu-Mei Han
- Department of Gastroenterology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Cheng-Suo Huang
- Department of Gastroenterology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Qing-Sheng Hou
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Xiao-Wen Wen
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Ben-Shi Wang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Zhen-Yu Zhu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Lei Zou
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China.
| |
Collapse
|
30
|
So YJ, Lee JU, Yang GS, Yang G, Kim SW, Lee JH, Kim JU. The Potentiality of Natural Products and Herbal Medicine as Novel Medications for Parkinson's Disease: A Promising Therapeutic Approach. Int J Mol Sci 2024; 25:1071. [PMID: 38256144 PMCID: PMC10816678 DOI: 10.3390/ijms25021071] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
As the global population ages, the prevalence of Parkinson's disease (PD) is steadily on the rise. PD demonstrates chronic and progressive characteristics, and many cases can transition into dementia. This increases societal and economic burdens, emphasizing the need to find effective treatments. Among the widely recognized causes of PD is the abnormal accumulation of proteins, and autophagy dysfunction accelerates this accumulation. The resultant Lewy bodies are also commonly found in Alzheimer's disease patients, suggesting an increased potential for the onset of dementia. Additionally, the production of free radicals due to mitochondrial dysfunction contributes to neuronal damage and degeneration. The activation of astrocytes and the M1 phenotype of microglia promote damage to dopamine neurons. The drugs currently used for PD only delay the clinical progression and exacerbation of the disease without targeting its root cause, and come with various side effects. Thus, there is a demand for treatments with fewer side effects, with much potential offered by natural products. In this study, we reviewed a total of 14 articles related to herbal medicines and natural products and investigated their relevance to possible PD treatment. The results showed that the reviewed herbal medicines and natural products are effective against lysosomal disorder, mitochondrial dysfunction, and inflammation, key mechanisms underlying PD. Therefore, natural products and herbal medicines can reduce neurotoxicity and might improve both motor and non-motor symptoms associated with PD. Furthermore, these products, with their multi-target effects, enhance bioavailability, inhibit antibiotic resistance, and might additionally eliminate side effects, making them good alternative therapies for PD treatment.
Collapse
Affiliation(s)
- Yu-Jin So
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
| | - Jae-Ung Lee
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
| | - Ga-Seung Yang
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
| | - Gabsik Yang
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
| | - Sung-Wook Kim
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
| | - Jun-Ho Lee
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
- Da CaPo Co., Ltd., 303 Cheonjam-ro, Wansan-gu, Jeonju-si 55069, Jeollabuk-do, Republic of Korea
| | - Jong-Uk Kim
- College of Korean Medicine, Woosuk University, Jeonju-si 54986, Jeollabuk-do, Republic of Korea; (Y.-J.S.); (J.-U.L.); (G.-S.Y.); (G.Y.); (S.-W.K.)
| |
Collapse
|
31
|
Lv S, Wang Q, Liu W, Zhang X, Cui M, Li X, Xu Y. Comparison of various exercise interventions on cognitive function in Alzheimer's patients: A network meta-analysis. Arch Gerontol Geriatr 2023; 115:105113. [PMID: 37418819 DOI: 10.1016/j.archger.2023.105113] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
OBJECTIVE Exercise helps enhance cognitive function in Alzheimer's patients, although the most effective forms of exercise remain unknown. METHODS This network meta-analysis was registered in INPLASY (INPLASY202330066). According to predetermined criteria, this investigation comprised randomized controlled studies involving exercise therapies in people with Alzheimer's disease. The exercise intervention was ranked using surface under the cumulative ranking curve (SUCRA) and mean ranking, with the critical goal outcomes being overall cognition, executive function, and memory function. RESULTS Resistance exercise is the most likely strategy to be beneficial for slowing down overall cognitive function loss in Alzheimer's patients (72.4%). Additionally, multi-component exercise was the most effective way to improve executive function (30.4%). The only type of exercise that significantly affects memory function is resistance exercise. Memory is the cognitive function that is least responsive to exercise. CONCLUSION Resistance exercise may be an efficient intervention for overall cognitive function decline in patients with Alzheimer's and conjointly for their memory function. Multi-component exercise is more effective in improving executive function in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Shi Lv
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Qian Wang
- Postdoctoral Workstation, Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Wenxin Liu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Xinlei Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Mengmeng Cui
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China
| | - Xiubin Li
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China.
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian 271000, China.
| |
Collapse
|
32
|
Morales-Carrizales DA, Gopar-Cuevas Y, Loera-Arias MDJ, Saucedo-Cardenas O, Montes de Oca-Luna R, Garcia-Garcia A, Rodriguez-Rocha H. A neuroprotective dose of trehalose is harmless to metabolic organs: comprehensive histopathological analysis of liver, pancreas, and kidney. Daru 2023; 31:135-144. [PMID: 37393413 PMCID: PMC10624785 DOI: 10.1007/s40199-023-00468-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/18/2023] [Indexed: 07/03/2023] Open
Abstract
BACKGROUND Trehalose is a non-reducing disaccharide synthesized by lower organisms. It has recently received special attention because of its neuroprotective properties by stimulating autophagy in Parkinson's disease (PD) models. Therefore, evaluating whether trehalose affects metabolic organs is vital to determine its neurotherapeutic safety. METHODS We validated the trehalose neuroprotective dosage in a PD model induced with intraperitoneal paraquat administration twice weekly for 7 weeks. One week before paraquat administration, mice were treated with trehalose in the drinking water and continued along with paraquat treatment. Histological and morphometrical analyses were conducted on the organs involved in trehalose metabolism, including the liver, pancreas, and kidney. RESULTS Paraquat-induced dopaminergic neuronal loss was significantly decreased by trehalose. After trehalose treatment, the liver morphology, the mononucleated/binucleated hepatocytes percentage, and sinusoidal diameter remained unchanged in each liver lobes. Endocrine and exocrine pancreas's histology was not affected, nor was any fibrotic process observed. The islet of Langerhans's structure was preserved when analyzing the area, the largest and smallest diameter, and circularity. Renal morphology remained undamaged, and no changes were identified within the glomerular basement membrane. The renal corpuscle structure did not suffer alterations in the Bowman's space, area, diameter, circularity, perimeter, and cellularity. Besides, the renal tubular structures's luminal area and internal and external diameter were preserved. CONCLUSION Our study demonstrates that systemic trehalose administration preserved the typical histological architecture of the organs involved in its metabolism, supporting its safety as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Diego Armando Morales-Carrizales
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Yareth Gopar-Cuevas
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Maria de Jesus Loera-Arias
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Odila Saucedo-Cardenas
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Roberto Montes de Oca-Luna
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - Aracely Garcia-Garcia
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico.
| | - Humberto Rodriguez-Rocha
- Departamento de Histologia, Universidad Autónoma de Nuevo Leon, Francisco I. Madero S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico.
| |
Collapse
|
33
|
Postaru M, Tucaliuc A, Cascaval D, Galaction AI. Cellular Stress Impact on Yeast Activity in Biotechnological Processes-A Short Overview. Microorganisms 2023; 11:2522. [PMID: 37894181 PMCID: PMC10609598 DOI: 10.3390/microorganisms11102522] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The importance of Saccharomyces cerevisiae yeast cells is known worldwide, as they are the most used microorganisms in biotechnology for bioethanol and biofuel production. Also, they are analyzed and studied for their similar internal biochemical processes to human cells, for a better understanding of cell aging and response to cell stressors. The special ability of S. cerevisiae cells to develop in both aerobic and anaerobic conditions makes this microorganism a viable model to study the transformations and the way in which cellular metabolism is directed to face the stress conditions due to environmental changes. Thus, this review will emphasize the effects of oxidative, ethanol, and osmotic stress and also the physiological and genetic response of stress mitigation in yeast cells.
Collapse
Affiliation(s)
- Madalina Postaru
- Department of Biomedical Science, Faculty of Medical Bioengineering, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, M. Kogălniceanu 9-13, 700454 Iasi, Romania;
| | - Alexandra Tucaliuc
- Department of Organic, Biochemical and Food, “Cristofor Simionescu” Faculty of Chemical, Engineering and Environmental Protection, Engineering, “Gheorghe Asachi” Technical University of Iasi, D. Mangeron 73, 700050 Iasi, Romania; (A.T.); (D.C.)
| | - Dan Cascaval
- Department of Organic, Biochemical and Food, “Cristofor Simionescu” Faculty of Chemical, Engineering and Environmental Protection, Engineering, “Gheorghe Asachi” Technical University of Iasi, D. Mangeron 73, 700050 Iasi, Romania; (A.T.); (D.C.)
| | - Anca-Irina Galaction
- Department of Biomedical Science, Faculty of Medical Bioengineering, “Grigore T. Popa” University of Medicine and Pharmacy of Iasi, M. Kogălniceanu 9-13, 700454 Iasi, Romania;
| |
Collapse
|
34
|
Ghosh S, Sharma R, Bammidi S, Koontz V, Nemani M, Yazdankhah M, Kedziora KM, Wallace CT, Yu-Wei C, Franks J, Bose D, Rajasundaram D, Hose S, Sahel JA, Puertollano R, Finkel T, Zigler JS, Sergeev Y, Watkins SC, Goetzman ES, Flores-Bellver M, Kaarniranta K, Sodhi A, Bharti K, Handa JT, Sinha D. The AKT2/SIRT5/TFEB pathway as a potential therapeutic target in atrophic AMD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552343. [PMID: 37609254 PMCID: PMC10441325 DOI: 10.1101/2023.08.08.552343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Age-related macular degeneration (AMD), the leading cause of geriatric blindness, is a multi-factorial disease with retinal-pigmented epithelial (RPE) cell dysfunction as a central pathogenic driver. With RPE degeneration, lysosomal function is a core process that is disrupted. Transcription factors EB/E3 (TFEB/E3) tightly control lysosomal function; their disruption can cause aging disorders, such as AMD. Here, we show that induced pluripotent stem cells (iPSC)-derived RPE cells with the complement factor H variant [ CFH (Y402H)] have increased AKT2, which impairs TFEB/TFE3 nuclear translocation and lysosomal function. Increased AKT2 can inhibit PGC1α, which downregulates SIRT5, an AKT2 binding partner. SIRT5 and AKT2 co-regulate each other, thereby modulating TFEB-dependent lysosomal function in the RPE. Failure of the AKT2/SIRT5/TFEB pathway in the RPE induced abnormalities in the autophagy-lysosome cellular axis by upregulating secretory autophagy, thereby releasing a plethora of factors that likely contribute to drusen formation, a hallmark of AMD. Finally, overexpressing AKT2 in RPE cells in mice led to an AMD-like phenotype. Thus, targeting the AKT2/SIRT5/TFEB pathway could be a potential therapy for atrophic AMD.
Collapse
|
35
|
Yu S, Park H, Kim W. Trehalose Inhibits Inflammatory Responses through Mitochondrial Reprogramming in RAW 264.7 Macrophages. Antioxidants (Basel) 2023; 12:1166. [PMID: 37371896 DOI: 10.3390/antiox12061166] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Studies reported the beneficial effects of trehalose on metabolic syndromes, hyperlipidemia, and autophagy, but its action mechanisms are still poorly understood. Even though trehalose is digested by disaccharidase and absorbed in the intestine, intact molecules encounter immune cells which form a solid balance between the allowance of nutritive substances and the removal of harmful pathogens. In this regard, the polarization of intestinal macrophages into an anti-inflammatory phenotype through metabolic regulation is emerging as a therapeutic strategy for the prevention of gastrointestinal inflammation. The current study investigated the effects of trehalose on immunological phenotypes, energy metabolism, and LPS-induced macrophage mitochondrial functioning. Results indicate that trehalose reduces prostaglandin E2 and nitric oxide, which are inflammatory mediators of LPS-induced macrophages. In addition, trehalose further significantly suppressed inflammatory cytokines and mediators via energy metabolism reprogramming towards M2-like status in LPS-stimulated macrophages.
Collapse
Affiliation(s)
- Seungmin Yu
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Hyejeong Park
- Department of Food Science and Biotechnology, Graduate School of Kyung Hee University, Yongin 17104, Republic of Korea
| | - Wooki Kim
- Department of Food Science and Biotechnology, Graduate School of Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
36
|
Zhao X, Wang Q, Li X, Xu H, Ren C, Yang Y, Xu S, Wei G, Duan Y, Tan Z, Fang Y. Norgestrel causes digestive gland injury in the clam Mactra veneriformis: An integrated histological, transcriptomics, and metabolomics study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 871:162110. [PMID: 36764532 DOI: 10.1016/j.scitotenv.2023.162110] [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: 10/23/2022] [Revised: 01/13/2023] [Accepted: 02/04/2023] [Indexed: 06/18/2023]
Abstract
The potential adverse effects of progestins on aquatic organisms, especially non-target species, are of increasing concern worldwide. However, the effect and mechanism of progestin toxicity on aquatic invertebrates remain largely unexplored. In the present study, clams Mactra veneriformis were exposed to norgestrel (NGT, 0, 10, and 1000 ng/L), the dominant progestin detected in the aquatic environment, for 21 days. NGT accumulation, histology, transcriptome, and metabolome were assessed in the digestive gland. The bioconcentration factor (BCF) was 386 and 268 in the 10 ng/L NGT group and 1000 ng/L NGT group, respectively, indicating efficient accumulation of NGT in the clams. Histological analysis showed that NGT led to the swelling of epithelial cells and blurring of the basement membrane in the digestive gland. Differentially-expressed genes and KEGG pathway enrichment analysis using a transcriptomic approach suggested that NGT primarily disturbed the detoxification system, antioxidant defense, carbohydrate and amino acid metabolism, and steroid hormone metabolism, which was consistent with the metabolites analyzed using a metabolomic approach. Furthermore, we speculated that the oxidative stress caused by NGT resulted in histological damage to the digestive gland. This study showed that NGT caused adverse effects in the clams and sheds light on the mechanisms of progestin interference in aquatic invertebrates.
Collapse
Affiliation(s)
- Xiaoran Zhao
- School of Agriculture, Ludong University, Yantai 264025, PR China
| | - Qing Wang
- Research and Development Center for Efficient Utilization of Coastal Bioresources, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China
| | - Xiangfei Li
- School of Agriculture, Ludong University, Yantai 264025, PR China
| | - Hua Xu
- Yantai Ecological Environment Monitoring Center, Shandong Province, Yantai 264010, PR China
| | - Chuanbo Ren
- Shandong Provincial Key Laboratory of Restoration for Marine Ecology, Shandong Marine Resource and Environment Research Institute, Yantai 264006, PR China
| | - Yanyan Yang
- Shandong Provincial Key Laboratory of Restoration for Marine Ecology, Shandong Marine Resource and Environment Research Institute, Yantai 264006, PR China
| | - Shuhao Xu
- School of Agriculture, Ludong University, Yantai 264025, PR China
| | - Guoxing Wei
- School of Agriculture, Ludong University, Yantai 264025, PR China
| | - Yujun Duan
- School of Agriculture, Ludong University, Yantai 264025, PR China
| | - Zhitao Tan
- School of Agriculture, Ludong University, Yantai 264025, PR China
| | - Yan Fang
- School of Agriculture, Ludong University, Yantai 264025, PR China.
| |
Collapse
|
37
|
Yuan S, Jiang SC, Zhang ZW, Fu YF, Yang XY, Li ZL, Hu J. Rethinking of Alzheimer's disease: Lysosomal overloading and dietary therapy. Front Aging Neurosci 2023; 15:1130658. [PMID: 36861123 PMCID: PMC9968973 DOI: 10.3389/fnagi.2023.1130658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Affiliation(s)
- Shu Yuan
- College of Resources, Sichuan Agricultural University, Chengdu, China,*Correspondence: Shu Yuan ✉
| | - Si-Cong Jiang
- Haisco Pharmaceutical Group Comp. Ltd., Chengdu, China
| | - Zhong-Wei Zhang
- College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Yu-Fan Fu
- College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Xin-Yue Yang
- College of Resources, Sichuan Agricultural University, Chengdu, China
| | - Zi-Lin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Medical University of the Air Force, Xi'an, China
| | - Jing Hu
- School of Medicine, Northwest University, Xi'an, China
| |
Collapse
|
38
|
Strontium ion attenuates osteoarthritis through inhibiting senescence and enhancing autophagy in fibroblast-like synoviocytes. Mol Biol Rep 2023; 50:1437-1446. [PMID: 36472726 DOI: 10.1007/s11033-022-08112-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) mainly occurs in the elderly population and seriously affects their quality of life (QOL). Strontium (Sr) ions have shown positive effects on bone tissue and are promising for OA treatment. However, the adequate treatment dosage and underlying mechanisms are unclear. This study investigated the effects and underlying mechanisms of different concentrations of Sr ions in a mouse model of OA induced by destabilization of the medial meniscus (DMM) surgery. DMM-induced OA mice received intra-articular injections of different concentrations of Sr ions, and a suitable concentration of Sr ions was found to improve OA. Furthermore, we investigated the mechanism by which Sr ions mediate senescence and autophagy in fibroblast-like synoviocytes (FLSs) in the synovial tissues of DMM-induced OA mice. OA mice treated with 10 µl of 5 mmol/L SrCl2 showed the greatest improvement in pain-related behavior and cartilage damage. In addition, in vivo and in vitro experiments revealed that Sr ions inhibit senescence and improve the autophagic function of FLSs. We also found that enhancement of the autophagic function of FLSs could effectively slow down senescence. Therefore, we show that Sr ions through the AMPK/mTOR/LC3B-II signal axis improve FLSs autophagy function and delay FLSs senescence, and furthermore, improve OA. These results suggest that senescence and autophagy function of FLSs may serve as promising targets for OA treatment, and that Sr ions may inhibit OA progression through these two targets.
Collapse
|
39
|
Rahman MA, Saikat ASM, Rahman MS, Islam M, Parvez MAK, Kim B. Recent Update and Drug Target in Molecular and Pharmacological Insights into Autophagy Modulation in Cancer Treatment and Future Progress. Cells 2023; 12:458. [PMID: 36766800 PMCID: PMC9914570 DOI: 10.3390/cells12030458] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/11/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Recent evidence suggests that autophagy is a governed catabolic framework enabling the recycling of nutrients from injured organelles and other cellular constituents via a lysosomal breakdown. This mechanism has been associated with the development of various pathologic conditions, including cancer and neurological disorders; however, recently updated studies have indicated that autophagy plays a dual role in cancer, acting as a cytoprotective or cytotoxic mechanism. Numerous preclinical and clinical investigations have shown that inhibiting autophagy enhances an anticancer medicine's effectiveness in various malignancies. Autophagy antagonists, including chloroquine and hydroxychloroquine, have previously been authorized in clinical trials, encouraging the development of medication-combination therapies targeting the autophagic processes for cancer. In this review, we provide an update on the recent research examining the anticancer efficacy of combining drugs that activate cytoprotective autophagy with autophagy inhibitors. Additionally, we highlight the difficulties and progress toward using cytoprotective autophagy targeting as a cancer treatment strategy. Importantly, we must enable the use of suitable autophagy inhibitors and coadministration delivery systems in conjunction with anticancer agents. Therefore, this review briefly summarizes the general molecular process behind autophagy and its bifunctional role that is important in cancer suppression and in encouraging tumor growth and resistance to chemotherapy and metastasis regulation. We then emphasize how autophagy and cancer cells interacting with one another is a promising therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Md. Ataur Rahman
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1-5 Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Abu Saim Mohammad Saikat
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md. Saidur Rahman
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Mobinul Islam
- Department of Energy and Materials Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | | | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1-5 Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
40
|
Tikhonova MA, Chang HM, Singh SK. Editorial: Experimental and innovative approaches to multi-target treatment of Parkinson's and Alzheimer's diseases - Volume II. Front Neurosci 2023; 17:1171866. [PMID: 37034172 PMCID: PMC10080043 DOI: 10.3389/fnins.2023.1171866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Affiliation(s)
- Maria A Tikhonova
- Laboratory of the Experimental Models of Neurodegenerative Processes, Scientific Research Institute of Neurosciences and Medicine (SRINM), Novosibirsk, Russia
| | - Hung-Ming Chang
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | |
Collapse
|
41
|
Jiang J, Wang Y, Deng M. New developments and opportunities in drugs being trialed for amyotrophic lateral sclerosis from 2020 to 2022. Front Pharmacol 2022; 13:1054006. [PMID: 36518658 PMCID: PMC9742490 DOI: 10.3389/fphar.2022.1054006] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 08/31/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that primarily affects motor neurons in the brain and spinal cord. In the recent past, there have been just two drugs approved for treatment, riluzole and edaravone, which only prolong survival by a few months. However, there are many novel experimental drugs in development. In this review, we summarize 53 new drugs that have been evaluated in clinical trials from 2020 to 2022, which we have classified into eight mechanistic groups (anti-apoptotic, anti-inflammatory, anti-excitotoxicity, regulated integrated stress response, neurotrophic factors and neuroprotection, anti-aggregation, gene therapy and other). Six were tested in phase 1 studies, 31 were in phase 2 studies, three failed in phase 3 studies and stopped further development, and the remaining 13 drugs were being tested in phase 3 studies, including methylcobalamin, masitinib, MN-166, verdiperstat, memantine, AMX0035, trazodone, CNM-Au8, pridopidine, SLS-005, IONN363, tofersen, and reldesemtiv. Among them, five drugs, including methylcobalamin, masitinib, AMX0035, CNM-Au8, and tofersen, have shown potent therapeutic effects in clinical trials. Recently, AMX0035 has been the third medicine approved by the FDA for the treatment of ALS after riluzole and edaravone.
Collapse
Affiliation(s)
| | | | - Min Deng
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| |
Collapse
|