1
|
Chen X, Zhu M, Shi Q, Huang Z, Zhu J, Sun P, Zhang H, Yang L, Chen X, Zhang Y, Feng L, Horimoto K, Li F, Han F, Chen D. Proof of concept study for developing 1-thienyl-β-carboline derivatives as IDO1 and TDO dual inhibitors to treat Parkinson's disease complicating depression. Eur J Med Chem 2025; 291:117597. [PMID: 40215562 DOI: 10.1016/j.ejmech.2025.117597] [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: 01/10/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025]
Abstract
Depressive symptoms are the most common neuropsychiatric disorders at all stages of Parkinson's disease (PD). Imbalances of the kynurenine pathway of tryptophan metabolism have been closely linked to the pathogenesis of PD and depression. Herein, we designed and synthesized a series of 1-thienyl-β-carboline derivatives as IDO1 and TDO dual inhibitors; among them, compound CZ-17 manifested moderate inhibitory activities to IDO1 and TDO with IC50 values of 0.33 and 1.78 μM, respectively. CZ-17 inhibited the kynurenine pathway of tryptophan degradation at the cellular level, and remarkably reduced the kynurenine/tryptophan ratio. CZ-17 displayed directly neuroprotective effect in corticosterone-induced PC12 neural cell injury model. In vivo experiments demonstrated that CZ-17 significantly increased dopamine and serotonin levels, improved MPTP-induced motor disability and rescued LPS-induced depressive behavior in zebrafish model. Acute toxicity tests of CZ-17 in zebrafish embryos showed no toxicity within the effective dose range. Additionally, CZ-17 displayed the potential to cross the BBB via passive diffusion according to ADMET prediction and Caco-2 permeability assay. Thus, CZ-17 may be a promising drug candidate for PD complicating depression.
Collapse
Affiliation(s)
- Xuan Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; School of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China
| | - Mengxiao Zhu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Qingyuan Shi
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zhenquan Huang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Junjie Zhu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Pingping Sun
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hongzhen Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Lili Yang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xun Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yu Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, Jiangsu, China; National Center of Technology Innovation for Biopharmaceuticals, Suzhou, 215000, Jiangsu, China
| | - Lili Feng
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | | | - Fei Li
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, Jiangsu, China; The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, 223300, China; Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Dongyin Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, Jiangsu, China; National Center of Technology Innovation for Biopharmaceuticals, Suzhou, 215000, Jiangsu, China; The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, 223300, China.
| |
Collapse
|
2
|
Krupa MM, Pienkowski T, Tankiewicz-Kwedlo A, Lyson T. Targeting the kynurenine pathway in gliomas: Insights into pathogenesis, therapeutic targets, and clinical advances. Biochim Biophys Acta Rev Cancer 2025; 1880:189343. [PMID: 40345262 DOI: 10.1016/j.bbcan.2025.189343] [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: 11/26/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Gliomas, the most prevalent primary brain tumors, continue to present significant challenges in oncology due to poor patient prognosis despite advances in treatment such as immunotherapy and cancer vaccines. Recent research highlights the potential of targeting tryptophan metabolism, particularly the kynurenine pathway (KP) and combinatorial approaches with immunotherapies, as a promising strategy in cancer research. The key enzymes of the kynurenine pathway, such as IDO1, IDO2, and TDO, and metabolites like kynurenine, kynurenic acid, and quinolinic acid, are implicated in fostering an immunosuppressive tumor microenvironment and promoting glioma cell survival. In glioblastoma, a highly aggressive glioma subtype, elevated IDO and TDO expression correlates with reduced survival rates. KP metabolites, such as kynurenine (KYN), 3-hydroxykynurenine (3-HK), kynurenic acid (KYNA), and quinolinic acid (QUIN), are involved in modulating immune responses, oxidative stress, neuroprotection, and neurotoxicity. This review synthesizes recent findings on the kynurenine pathway involvement in glioma pathogenesis, examining potential therapeutic targets within this pathway and discussing ongoing clinical trials that draw attention to treatments based on this pathway. Furthermore, it highlights novel findings on the post-translational modifications of kynurenine pathway enzymes and their regulatory roles, presenting their potential as therapeutic targets in gliomas.
Collapse
Affiliation(s)
- Mikolaj Marek Krupa
- Department of Neurosurgery, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Tomasz Pienkowski
- Laboratory of Metabolomics and Proteomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland.
| | - Anna Tankiewicz-Kwedlo
- Department of Pharmacodynamics, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Tomasz Lyson
- Department of Neurosurgery, Medical University of Bialystok, 15-276 Bialystok, Poland; Department of Interventional Neurology, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
3
|
Fu X, Guo W, Cheng Y, Li L. An ultra-high-performance tandem mass spectrometry method to quantify tryptophan metabolites in aqueous humor of primary angle-closure glaucoma patients. J Chromatogr A 2025; 1750:465939. [PMID: 40194501 DOI: 10.1016/j.chroma.2025.465939] [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: 12/04/2024] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
This study presented the development and validation of a robust ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method for the simultaneous quantification of tryptophan (TRP) and its nine metabolites in aqueous humor (AH) to explore the regulation of the TRP metabolic pathway in primary angle-closure glaucoma (PACG). The optimized UPLC-MS/MS method demonstrated good linearity (R² > 0.99), sensitivity (LLMI: 0.11 - 1.31 ng/mL), precision (CVs: 2.18 % and 12.88 %), recovery rates (85.06 % - 105.74 %), bench-top, long-term and on-instrument stabilities (CVs: 2.35 % - 6.88 %). The validated UPLC-MS/MS method was applied to AH samples from PACG patients with cataract and cataract-alone patients. The results showed that kynurenine and 3-hydroxykynurenine concentrations were significantly increased in the AH of the PACG group, indicating up-regulated indoleamine 2,3-dioxygenase activity and a metabolic shift towards the production of the neurotoxic metabolites within the kynurenine pathway. These findings underscore the potential involvement of TRP metabolism in PACG pathogenesis.
Collapse
Affiliation(s)
- Xingang Fu
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Weiwei Guo
- Department of Anesthesiology, The Affiliated Chengdu 363 Hospital of Southwest Medical University, Chengdu, PR China
| | - Yuheng Cheng
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Lin Li
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China.
| |
Collapse
|
4
|
Tian L, Long G, Zhu S, Wang Y, Xu P, Liu L, Yao H, Fang S, Chen S, Li S. Nanoimmunomodulation of the Aβ-STING feedback machinery in microglia for Alzheimer's disease treatment. Proc Natl Acad Sci U S A 2025; 122:e2427257122. [PMID: 40434641 DOI: 10.1073/pnas.2427257122] [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: 12/31/2024] [Accepted: 04/08/2025] [Indexed: 05/29/2025] Open
Abstract
Imbalanced production and clearance of amyloid-β (Aβ) is a hallmark pathological feature of Alzheimer's disease (AD). While several monoclonal antibodies targeting Aβ have shown reductions in amyloid burden, their impact on cognitive function remains controversial, with the added risk of inflammatory side effects. Dysregulated stimulator of interferon genes (STING) signaling is implicated in neurodegenerative disorders, yet the biological interaction between this pathway and Aβ, as well as their combined influence on AD progression, is poorly understood. Here, we show that while microglia play a protective role in clearing extracellular Aβ, excessive Aβ engulfment triggers the cytosolic leakage of mitochondrial DNA for cGAS-STING cascade. This creates a negative feedback loop that not only exacerbates neuroinflammation but also impairs further Aβ clearance. To address this, we present a nanomedicine approach termed "Aβ-STING Synergistic ImmunoSilencing Therapy (ASSIST)". ASSIST comprises STING inhibitors encapsulated within a blood-brain barrier (BBB)-permeable polymeric micelle that also serves as an Aβ scavenger. Through a multivalent interaction mechanism, ASSIST efficiently destabilizes Aβ plaques and prevents monomer aggregation, subsequently promoting the engulfment of the dissociated Aβ by microglia rather than neurocytes. Furthermore, the STING signaling induced by excessive Aβ uptake is blocked, reducing inflammation and restoring microglial homeostatic functions involved in Aβ clearance. Intravenous administration of ASSIST significantly reduces Aβ burden and improves cognition in AD mice, with minimal cerebral amyloid angiopathy or microhemorrhages. We provide a proof-of-concept nanoengineering strategy to target the maladaptive immune feedback loop arising from conventional immunotherapy for AD treatment.
Collapse
Affiliation(s)
- Limin Tian
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guangyu Long
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Siqi Zhu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yuelong Wang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Pengcheng Xu
- Department of Pharmaceutical Engineering, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China
| | - Lifeng Liu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Yao
- Department of Medicinal Chemistry, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Shentong Fang
- School of Biopharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuqing Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Suxin Li
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210009, China
| |
Collapse
|
5
|
Loera-Lopez AL, Lord MN, Noble EE. Astrocytes of the hippocampus and responses to periprandial neuroendocrine hormones. Physiol Behav 2025; 295:114913. [PMID: 40209869 PMCID: PMC12066093 DOI: 10.1016/j.physbeh.2025.114913] [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: 01/22/2025] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/12/2025]
Abstract
Astrocytes have risen as stars in the field of energy homeostasis and neurocognitive function, acting as a bridge of communication between the periphery and the brain, providing metabolic support, signaling via gliotransmitters, and altering synaptic communication. Dietary factors and energy state have a profound influence on hippocampal function, and the hippocampus is critical for appropriate behavioral responses associated with feeding and internal hunger cues (being in the fasted or full state), but how the hippocampus senses periprandial status and is impacted by diet is largely unknown. Periprandial hormones act within the hippocampus to modulate processes involved in hippocampal-dependent learning and memory function and astrocytes likely play an important role in modulating this signaling. In addition to periprandial hormones, astrocytes are positioned to respond to changes in circulating nutrients like glucose. Here, we review literature investigating how astrocytes mediate changes in hippocampal function, highlighting astrocyte location, morphology, and function in the context of integrating glucose metabolism, neuroendocrine hormone action, and/or cognitive function in the hippocampus. Specifically, we discuss research findings on the effects of insulin, ghrelin, leptin, and GLP-1 on glucose homeostasis, neural activity, astrocyte function, and behavior in the hippocampus. Because obesogenic diets impact neuroendocrine hormones, astrocytes, and cognitive function, we also discuss the effects of diet and diet-induced obesity on these parameters.
Collapse
Affiliation(s)
- Ana L Loera-Lopez
- Neuroscience Graduate Program, University of Georgia, Athens, GA, 30606, USA; Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA
| | - Magen N Lord
- Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA
| | - Emily E Noble
- Neuroscience Graduate Program, University of Georgia, Athens, GA, 30606, USA; Department of Nutritional Sciences, University of Georgia, Athens, GA, 30606, USA.
| |
Collapse
|
6
|
Qiu H, Yin Y, Qin Z, Li D, Wang P. A patent review of IDO1 inhibitors for cancer (2023 - present): an update. Expert Opin Ther Pat 2025:1-15. [PMID: 40414695 DOI: 10.1080/13543776.2025.2510641] [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: 12/20/2024] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION Indoleamine 2,3-dioxygenase 1 (IDO1) is a promising target in cancer immunotherapy, yet its application faces significant challenges due to complex mechanisms of action. Recent advancements in IDO1 inhibitors aim to tackle these issues, potentially paving the way for successful therapeutic development. AREAS COVERED This review highlights patent publications (2023-2024) related to IDO1 inhibitors with potential anti-cancer applications, sourced from Espacenet and Google Scholar. EXPERT OPINION IDO1 exhibits complex mechanisms of action and variable expression across different cancer types, presenting both challenges and opportunities. Its intricate mechanisms in tumor development and immune evasion pose significant challenges for translating IDO1 inhibitors into clinical drugs. However, recent advancements in AI-guided drug design, combination therapies, and improved drug delivery methods offer promising insights for enhancing IDO1 inhibitors, although further data is warranted. Despite these challenges, the increasing availability of IDO1 crystal structures and a deeper understanding of its biological roles support ongoing trials that combine IDO1 inhibitors with other therapies. These developments hold potential for improving therapeutic outcomes in cancer treatment. Moreover, the growing interest in applying IDO1 inhibitors to other diseases could stimulate further research and development of new IDO1 inhibitors, potentially benefiting their application in cancer therapy as well.
Collapse
Affiliation(s)
- Hanyue Qiu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Yiheng Yin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Ziyu Qin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Dongdong Li
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, People's Republic of China
| | - Pengfei Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, People's Republic of China
| |
Collapse
|
7
|
Tang Z, Sha T, Wang Y, Xiao Y, Ding Y, Ni R, Qi X. Isoliquiritigenin attenuated cognitive impairment, cerebral tau phosphorylation and oxidative stress in a streptozotocin-induced mouse model of Alzheimer's disease. Life Sci 2025; 376:123759. [PMID: 40414556 DOI: 10.1016/j.lfs.2025.123759] [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: 01/07/2025] [Revised: 05/11/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
AIMS Isoliquiritigenin is a natural flavonoid extracted from the root of the medicinal herb liquorice. Isoliquiritigenin has various biological effects, including antioxidant, neuroprotective, anti-inflammatory, and antidiabetic activities, and improved mitochondrial function in earlier studies. Tau hyperphosphorylation, mitochondrial dysfunction and oxidative stress play important roles in Alzheimer's disease (AD). Here, we assessed the neuroprotective effects of isoliquiritigenin on a streptozotocin-injected mouse model. MATERIALS Molecular docking analysis of isoliquiritigenin with mammalian target of rapamycin (mTOR) and ERK2. The mice (n = 27, male) were intracerebroventricularly injected with streptozotocin, treated with isoliquiritigenin (intraperitoneal, 2 days) and assessed using the Morris water maze. Oxidative stress, tau phosphorylation, mitochondrial dysfunction and synaptic impairment were evaluated in the cortex and hippocampal tissues of the mice by using biochemical assays and immunostaining. RESULTS Isoliquiritigenin treatment mitigated the spatial memory capacity of streptozotocin-injected mice and alleviated tau phosphorylation at Ser396 and Thr231, the production of reactive oxygen species, the intracellular ATP level, the mitochondrial proteins p-DRP1 (S616), Mfn1 and Mfn2, neuronal loss, and synaptic impairment (PSD95, SNAP25). Isoliquiritigenin treatment reduced the levels of mTOR Ser2448 and ERK2 Thr202/Tyr204 and upregulated the level of GSK-3β Ser9 in the cortex and hippocampus of streptozotocin-injected mice. CONCLUSION In conclusion, our findings suggest that isoliquiritigenin ameliorates streptozotocin-induced cognitive impairment, hyperphosphorylated tau, oxidative stress, mitochondrial dysfunction and synaptic impairment by decreasing mTOR and ERK activity and increasing GSK-3β activity.
Collapse
Affiliation(s)
- Zhi Tang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Ting Sha
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Yuanshang Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Yuanting Ding
- The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Department of Nuclear Medicine, Inselspital University of Bern, Bern, Switzerland.
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
8
|
Han M, He W, Zhu W, Guo L. The role of protein lactylation in brain health and disease: current advances and future directions. Cell Death Discov 2025; 11:213. [PMID: 40307243 PMCID: PMC12043837 DOI: 10.1038/s41420-025-02408-w] [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: 11/06/2024] [Revised: 02/19/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Lactate, the end product of glycolysis, plays a crucial role in cellular signaling and metabolism. The discovery of lactylation, a novel post-translational modification, has uncovered the role of lactate in regulating diseases, especially in the brain. Lactylation connects genetic encoding with protein function, thereby influencing key biological processes. Increasing evidence supports lactate-mediated lactylation as a critical modulator in neurological disorders. This review offers an overview of lactate metabolism and lactylation, highlighting recent advances in understanding the regulatory enzymes of lactylation and their role in the central nervous system. We investigate the impact of lactylation on brain dysfunctions, including neurodegenerative diseases, cerebrovascular disorders, neuroinflammation, brain tumors, and psychiatric conditions. Moreover, we highlight the therapeutic potential of targeting lactylation in treating brain disorders and outline key research gaps and future directions needed to advance this promising field.
Collapse
Affiliation(s)
- Mingrui Han
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Queen Mary school, medical department, Nanchang University, Nanchang, Jiangxi, China
| | - Wenfeng He
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Wengen Zhu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Linjuan Guo
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China.
| |
Collapse
|
9
|
Na D, Zhang Z, Meng M, Li M, Gao J, Kong J, Zhang G, Guo Y. Energy Metabolism and Brain Aging: Strategies to Delay Neuronal Degeneration. Cell Mol Neurobiol 2025; 45:38. [PMID: 40259102 PMCID: PMC12011708 DOI: 10.1007/s10571-025-01555-z] [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: 01/12/2025] [Accepted: 04/09/2025] [Indexed: 04/23/2025]
Abstract
Aging is characterized by a gradual decline in physiological functions, with brain aging being a major risk factor for numerous neurodegenerative diseases. Given the brain's high energy demands, maintaining an adequate ATP supply is crucial for its proper function. However, with advancing age, mitochondria dysfunction and a deteriorating energy metabolism lead to reduced overall energy production and impaired mitochondrial quality control (MQC). As a result, promoting healthy aging has become a key focus in contemporary research. This review examines the relationship between energy metabolism and brain aging, highlighting the connection between MQC and energy metabolism, and proposes strategies to delay brain aging by targeting energy metabolism.
Collapse
Affiliation(s)
- Donghui Na
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Zechen Zhang
- Mudi Meng Honors College, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meng Meng
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
- Department of Pathology, Hebei North University, Zhangjiakou, Hebei, China
| | - Junyan Gao
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada.
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China.
| | - Ying Guo
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, Hebei, China.
- Department of Pathology, Hebei North University, Zhangjiakou, Hebei, China.
- Hebei Key Laboratory of Neuropharmacology, Hebei North University, Zhangjiakou, Hebei, China.
| |
Collapse
|
10
|
Huang D, Ovcharenko I. Silencer variants are key drivers of gene upregulation in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.07.25325386. [PMID: 40297423 PMCID: PMC12036408 DOI: 10.1101/2025.04.07.25325386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Alzheimer's disease (AD), particularly late-onset AD, stands as the most prevalent neurodegenerative disorder globally. Owing to its substantial heritability, genetic studies have emerged as indispensable for elucidating genes and biological pathways driving AD onset and progression. However, genetic and molecular mechanisms underlying AD remain poorly defined, largely due to the pronounced heterogeneity of AD and the intricate interactions among AD genetic factors. Notably, approximately 90% of AD-associated genetic variants reside in intronic and intergenic regions, yet their functional significance has remained largely uncharacterized. To address this challenge, we developed a deep learning framework combining bulk and single-cell epigenomic data to evaluate the regulatory potential (i.e., silencing and activating strength) of noncoding AD variants in the dorsolateral prefrontal cortex (DLPFCs) and its major cell types. This model identified 1,457 silencer and 3,084 enhancer AD-associated variants in the DLPFC and binned them into silencer variants only (SL), enhancer variants only (EN), or both variant types (ENSL) classes. Each class exerts distinct cellular and molecular influences on AD pathogenesis. EN loci predominantly regulate housekeeping metabolic processes, whereas SL loci (including the genes MS4A6A , TREM2 , USP6NL , HLA-D ) are selectively linked to immune responses. Notably, 71% of these genes are significantly upregulated in AD and pro-inflammation-stimulated microglia. Furthermore, genes associated with SL loci are, in neuronal cells, often responsive to glutamate receptor antagonists (e.g, NBQX) and anti-inflammatory perturbagens (such as D-64131), the compound classes known for reducing the AD risk. ENSL loci, in contrast, are uniquely implicated in memory maintenance, neurofibrillary tangle assembly, and are also shared by other neurological disorders such as Parkinson's disease and schizophrenia. Key genes in this class of loci, such as MAPT , CR1/2 , and CLU , are frequently upregulated in AD subtypes with hyperphosphorylated tau aggregates. Critically, our model can accurately predict the impact of regulatory variants, with an average Pearson correlation coefficient of 0.54 and a directional concordance rate of 70% between our predictions and experimental outcomes. This model identified rs636317 as a causal AD variant in the MS4A locus, distinguishing it from the 7bp-away allele-neutral variant rs636341. Similarly, rs7922621 was prioritized over its 54-bp-away allele-neutral rs7901634 in the TSPAN14 locus. Additional causal variants include rs6701713 in the CR1 locus, and rs28834970 and rs755951 in the PTK2B locus. Collectively, this work advances our understanding of the regulatory landscape of AD-associated genetic variants, providing a framework to explore their functional roles in the pathogenesis of this complex disease.
Collapse
|
11
|
Waller TJ, Collins CA, Dus M. Pyruvate kinase deficiency links metabolic perturbations to neurodegeneration and axonal protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647282. [PMID: 40235982 PMCID: PMC11996495 DOI: 10.1101/2025.04.04.647282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Neurons rely on tightly regulated metabolic networks to sustain their high-energy demands, particularly through the coupling of glycolysis and oxidative phosphorylation. Here, we investigate the role of pyruvate kinase (PyK), a key glycolytic enzyme, in maintaining axonal and synaptic integrity in the Drosophila melanogaster neuromuscular system. Using genetic deficiencies in PyK, we show that disrupting glycolysis induces progressive synaptic and axonal degeneration and severe locomotor deficits. These effects require the conserved dual leucine zipper kinase (DLK), Jun N-terminal kinase (JNK), and activator protein 1 (AP-1) Fos transcription factor axonal damage signaling pathway and the SARM1 NADase enzyme, a key driver of axonal degeneration. As both DLK and SARM1 regulate degeneration of injured axons (Wallerian degeneration), we probed the effect of PyK loss on this process. Consistent with the idea that metabolic shifts may influence neuronal resilience in context-dependent ways, we find that pyk knockdown delays Wallerian degeneration following nerve injury, suggesting that reducing glycolytic flux can promote axon survival under stress conditions. This protective effect is partially blocked by DLK knockdown and fully abolished by SARM1 overexpression. Together, our findings help bridge metabolism and neurodegenerative signaling by demonstrating that glycolytic perturbations causally activate stress response pathways that dictate the balance between protection and degeneration depending on the system's state. These results provide a mechanistic framework for understanding metabolic contributions to neurodegeneration and highlight the potential of metabolism as a target for therapeutic strategies. Abstract Figure
Collapse
|
12
|
Marshall TJ, Watne LO, Sanders RD. Mechanisms of perioperative neuronal injury and the search for therapies. Br J Anaesth 2025; 134:906-908. [PMID: 39893064 PMCID: PMC11947568 DOI: 10.1016/j.bja.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Perioperative neuronal injury includes both delirium and postoperative cognitive decline, and has profound potentially long-term effects on surgical patients and an economic cost. Recent advances have been made in the underlying biological causes of these injuries, including validation of biomarkers of neuronal damage such as neurofilament light, further understanding of the inflammatory pathways and mediators responsible for neuronal injuries, metabolic triggers, and the role of ischaemia. Several novel approaches to perioperative protection of brain health are also being trialled. We summarise the current evidence regarding the causes of neuronal injury, and work taking place related to its prevention and treatment.
Collapse
Affiliation(s)
- Timothy J Marshall
- Department of Anaesthetics, Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Central Clinical School, University of Sydney, Camperdown, NSW, Australia.
| | - Leiv O Watne
- Oslo Delirium Research Group, Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway; Department of Geriatric Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Robert D Sanders
- Department of Anaesthetics, Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Central Clinical School, University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
13
|
Liu J, Gao WJ, Chen YX, Jia DX, Fang CH, Li Y, Lv MJ, Yue JM, Yu JH. Triterpenoids with bioactivities from Oenothera biennis. Fitoterapia 2025; 182:106408. [PMID: 39909355 DOI: 10.1016/j.fitote.2025.106408] [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: 12/18/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Phytochemical investigation into the whole plants of Oenothera biennis led to the isolation and identification of 28 triterpenoids, of which oenothebienoids A-G (1-7) are reported for the first time. Oenothebienoids A-D (1-4) have an unnormal 20-epi-ursane type triterpenoid framework, which is rarely documented. To swiftly and precisely differentiate the C-20 configurations, a rule was summarized by analysis of the NMR data from the two pairs of simultaneously obtained C-20 epimers, namely 1/8 and 3/9. A series of bioactive assessments revealed that compounds 13 and 14 show moderate inhibitory activity against α-glucosidase with IC50 values of 5.16 ± 1.08 and 4.24 ± 0.48 μM, respectively. Additionally, compounds 14 and 26 display significant inhibitory activity against IDO1 with IC50 values of 8.47 ± 0.57 and 8.83 ± 0.72 μM, respectively.
Collapse
Affiliation(s)
- Jing Liu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Wen-Jing Gao
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Yi-Xiao Chen
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Dong-Xue Jia
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Chu-Hong Fang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Ying Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Ming-Jun Lv
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Jian-Min Yue
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.
| | - Jin-Hai Yu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| |
Collapse
|
14
|
Guzmán‐Hernández R, Fossati S. Fibrillar tau alters cerebral endothelial cell metabolism, vascular inflammatory activation, and barrier function in vitro and in vivo. Alzheimers Dement 2025; 21:e70077. [PMID: 40110691 PMCID: PMC11923556 DOI: 10.1002/alz.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION The presence of tau aggregates in and around the brain vasculature in Alzheimer's disease (AD) and tauopathies suggests its possible pathogenicity to cerebral endothelial cells (ECs). METHODS We used an in vitro model of the blood-brain barrier (BBB) to understand the mechanisms of fibrillar tau-mediated cerebral EC and BBB pathology, confirming our findings in 3-month-old P301S mice brains and extracted microvessels. RESULTS Protofibrillar and fibrillar tau species induce endothelial barrier permeability through an increase in glycolysis, which activates ECs toward a pro-inflammatory phenotype, inducing loss of junction protein expression and localization. The Warburg-like metabolic shift toward glycolysis and increased vascular pathological phenotypes are also present in young P301S mice. DISCUSSION In sum, our work reveals that fibrillar tau species, by enhancing endothelial glycolytic metabolism, promote vascular inflammatory phenotypes and loss of BBB function, highlighting the importance of addressing and targeting early tau-mediated neurovascular damage in AD and tauopathies. HIGHLIGHTS We improve the understanding of the mechanisms of vascular pathology in tauopathies. Fibrillar tau mediates vascular metabolic changes, inflammation, and blood-brain barrier (BBB) dysfunction. These events are replicated at early stages in a tauopathy mouse model. Inhibiting altered glycolysis reduces BBB permeability and endothelial activation.
Collapse
Affiliation(s)
- Roberto Guzmán‐Hernández
- Alzheimer's Center at TempleDepartment of Neural SciencesTemple University Lewis Katz School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Silvia Fossati
- Alzheimer's Center at TempleDepartment of Neural SciencesTemple University Lewis Katz School of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
15
|
Ko EA, Zhou T, Ko JH, Jung SC. Transcriptomic Alteration in the Brain and Gut of Offspring Following Prenatal Exposure to Corticosterone. Exp Neurobiol 2025; 34:9-19. [PMID: 40091635 PMCID: PMC11919639 DOI: 10.5607/en24029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Maternal stress during pregnancy can profoundly affect offspring health, increasing the risk of psychiatric disorders, metabolic diseases, and gastrointestinal problems. In this study, the effects of high prenatal corticosterone exposure on gene expression in the brain and small intestine of rat offspring were investigated via RNA-sequencing analysis. Pregnant rats were divided into two groups: Corti.Moms were injected with corticosterone daily, while Nor.Moms were given saline injections. Their offspring were labeled as Corti.Pups and Nor.Pups, respectively. The brain tissue analysis of Corti.Pups showed that the expression levels of the genes linked to neurodegenerative conditions increased and enhanced mitochondrial biogenesis, possibly due to higher ATP demands. The genes associated with calcium signaling pathways, neuroactive ligand-receptor interactions, and IgA production were also upregulated in the small intestine of Corti.pups. Conversely, the genes related to protein digestion, absorption, and serotonergic and dopaminergic synaptic activities were downregulated. These findings revealed that gene expression patterns in both the brain and intestinal smooth muscle of offspring prenatally exposed to corticosterone were substantially altered. Thus, this study provided valuable insights into the effects of prenatal stress on neurodevelopment and gut function.
Collapse
Affiliation(s)
- Eun-A Ko
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Sung-Cherl Jung
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
16
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-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: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Pérez-Oliveira S, Álvarez I, Menéndez-González M, Duarte-Herrera ID, Blázquez-Estrada M, Castilla-Silgado J, Suárez E, García-Fernández C, Siso-García P, García-González P, Rosende-Roca M, Boada M, Ruiz A, Infante J, De la Casa-Fages B, González-Aramburu I, Álvarez V, Pastor P. HTT, ATXN1 and ATXN2 CAG triplet repeat sizes: exploring their role in the disease risk and cancer comorbidity in Parkinson's disease. Brain Commun 2025; 7:fcaf060. [PMID: 39974178 PMCID: PMC11837329 DOI: 10.1093/braincomms/fcaf060] [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: 08/26/2024] [Revised: 01/07/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease genetic embraces genetic and non-genetic factors. It has been suggested a link between CAG repeat number in the HTT, ATXN1 and ATXN2 genes and different neurodegenerative diseases. Several genetic factors involved in Parkinson's disease development are indeed associated with cancer pathways. Moreover, several studies found a low prevalence of cancer in neurodegenerative diseases that can be associated with a low CAG repeat size in several genes. This study aimed to investigate the influence of CAG repeat sizes in ATXN1, ATXN2 and HTT genes on the risk for developing cancer and Parkinson's disease in a large cohort of patients with idiopathic Parkinson's disease and healthy controls. The work included 1052 patients with idiopathic Parkinson's disease and 1070 controls of European ancestry. CAG repeat sizes in HTT, ATXN1 and ATXN2 genes were analysed. Dunn's multiple comparison test for quantitative variables and logistic and linear regression were used. The long ATXN1 and HTT alleles and CAG size and both the ATXN2 short and long alleles were predictors for the Parkinson's disease risk. The long CAG ATXN1 allele gene was associated with the risk of cancer. No association was observed between CAG size in the HTT and ATXN2 genes and risk of cancer in patients with Parkinson's disease. We described an association of HTT, ATXN1 and ATXN2 with the risk of Parkinson's disease, which reinforce the hypothesis of the common pathway of neurodegeneration. Besides, ATXN1 could be a predictor of cancer risk among patients with Parkinson's disease, and these results suggest that cancer and neurodegeneration processes can share common pathways.
Collapse
Affiliation(s)
- Sergio Pérez-Oliveira
- Laboratorio de Genética, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ignacio Álvarez
- Department of Neurology, Movement Disorders Unit, University Hospital Mútua de Terrassa and Fundació Docència i Recerca Mútua de Terrassa, 08221 Terrassa, Barcelona, Spain
| | - Manuel Menéndez-González
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Neurología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Israel David Duarte-Herrera
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Networked Biomedical Research Center (CIBER)—Respiratory Diseases, 28029 Madrid, Spain
| | - Marta Blázquez-Estrada
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Neurología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Departamento de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain
| | | | - Esther Suárez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Neurología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Ciara García-Fernández
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Neurología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | - Pablo Siso-García
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Servicio de Neurología, Hospital Universitario de Cabueñes, 33394 Gijón, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08028 Barcelona, Spain
| | - Maitee Rosende-Roca
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08028 Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08028 Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, 08028 Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jon Infante
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Neurology, Marqués de Valdecilla University Hospital, Universidad de Cantabria, 39008 Santander, Spain
| | - Beatriz De la Casa-Fages
- Department of Neurology, Movement Disorders Unit, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Instituto Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
| | - Isabel González-Aramburu
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Neurology, Marqués de Valdecilla University Hospital, Universidad de Cantabria, 39008 Santander, Spain
| | - Victoria Álvarez
- Laboratorio de Genética, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Pau Pastor
- Department of Neurology, Unit of Neurodegenerative diseases, University Hospital Germans Trias i Pujol and The Germans Trias i Pujol Research Institute (IGTP) Badalona, 08916 Barcelona, Spain
| |
Collapse
|
18
|
Qian W, Liu D, Liu J, Liu M, Ji Q, Zhang B, Yang Z, Cheng Y, Zhou S. The Mitochondria-Targeted Micelle Inhibits Alzheimer's Disease Progression by Alleviating Neuronal Mitochondrial Dysfunction and Neuroinflammation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408581. [PMID: 39713820 DOI: 10.1002/smll.202408581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/07/2024] [Indexed: 12/24/2024]
Abstract
Mitochondrial dysfunction plays an important role in neuroinflammation and cognitive impairment in Alzheimer's disease (AD). Herein, this work designs a mitochondria-targeted micelle CsA-TK-SS-31 (CTS) to block the progression of AD by simultaneously alleviating mitochondrial dysfunction in microglia and neurons. The mitochondria-targeted peptide SS-31 drives cyclosporin A (CsA) to penetrate the blood-brain barrier (BBB) and delivers CsA to mitochondria of microglia and neurons in the brains of 5 × FAD mice. Under the high level of reactive oxygen species (ROS) environment in damaged mitochondria of microglia and neurons, the linker (thioketal, TK) between CsA and SS-31 is broken and CsA and SS-31 are released while consuming ROS in the microenvironment. The released CsA and SS-31 synergistically restore the mitochondrial membrane potential and the balance between the fission and fusion of mitochondria, which subsequently protect neurons from apoptosis and reduce the activation of microglia in the brains of 5 × FAD mice. Ultimately, the neuroinflammation and cognitive impairment of 5 × FAD mice are ameliorated. This research provides a synergistic treatment strategy for AD through alleviating mitochondrial dysfunction to reduce neuroinflammation and restore the function of neurons simultaneously.
Collapse
Affiliation(s)
- Wenqiang Qian
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Jie Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Zhifu Yang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| |
Collapse
|
19
|
Zhang X, Liu Y, Rekowski MJ, Wang N. Lactylation of tau in human Alzheimer's disease brains. Alzheimers Dement 2025; 21:e14481. [PMID: 39740133 PMCID: PMC11851134 DOI: 10.1002/alz.14481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION Aggregation of hyperphosphorylated tau (tauopathy) is associated with cognitive impairment in patients with Alzheimer's disease (AD). In AD, a metabolic shift due to the Warburg effect results in increased lactate production. Lactate can induce a post-translational modification (PTM) on proteins that conjugates lactyl groups to lysine (K) residues, which is known as lactylation. METHODS We analyzed lactylation of tau in control and AD brain tissue and conducted cell-based assays. In addition, we used in vitro assays to determine whether p300 catalyzed tau lactylation. RESULTS Quantitative proteomics detected that tau lactylation was elevated in AD brains, with K residue at position 331 (K331) being a prominent site. Lactate induced tau lactylation, which increased tau phosphorylation and cleavage and reduced ubiquitination. Inhibition of lactate production lowered tau lactylation; p300 catalyzed tau lactylation. DISCUSSION Our findings suggest that tau lactylation links metabolic dysregulation with tauopathy and could serve as a novel diagnostic and therapeutic target. HIGHLIGHTS Elevated tau lactylation, particularly at K331, is evident in in human AD brain samples. Lactate induces tau lactylation, enhancing phosphorylation and cleavage while inhibiting ubiquitination. The acetyl-transferase p300 catalyzes tau lactylation, with K331 being the most prominent site.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Cell Biology and PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- Institute of Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Yan Liu
- Department of Cell Biology and PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- Institute of Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Michaella J. Rekowski
- Mass Spectrometry/Proteomics Core LaboratoryUniversity of Kansas Medical CenterKansas CityKansasUSA
- Department of Cancer BiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Ning Wang
- Department of Cell Biology and PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- Institute of Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
- Landon Center on AgingUniversity of Kansas Medical CenterKansas CityKansasUSA
- University of Kansas Alzheimer's Disease Research CenterFairwayKansasUSA
| |
Collapse
|
20
|
Wang Y, Wu W, Zeng F, Meng X, Peng M, Wang J, Chen Z, Liu W. The role of kynurenine pathway metabolism mediated by exercise in the microbial-gut-brain axis in Alzheimer's disease. Exp Neurol 2025; 384:115070. [PMID: 39603488 DOI: 10.1016/j.expneurol.2024.115070] [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: 08/07/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
In recent years, the role of the microbiome-gut-brain axis in the pathogenesis of Alzheimer's disease (AD) has garnered increasing attention. Specifically, tryptophan metabolism via the kynurenine pathway (KP) plays a crucial regulatory role in this axis. This study reviews how exercise regulates the microbiome-gut-brain axis by influencing kynurenine pathway metabolism, thereby exerting resistance against AD. This paper also discusses how exercise positively impacts AD via the microbiome-gut-brain axis by modulating the endocrine, autonomic nervous, and immune systems. Although the specific mechanisms are not fully understood, research indicates that exercise may optimize tryptophan metabolism by promoting the growth of beneficial microbiota and inhibiting harmful microbiota, producing substances that are beneficial to the nervous system and combating AD. The aim of this review is to provide new perspectives and potential intervention strategies for the prevention and treatment of AD by exploring the links between exercise, KP and the gut-brain axis.
Collapse
Affiliation(s)
- Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Mei Peng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
21
|
Zhu Y, Verkhratsky A, Chen H, Yi C. Understanding glucose metabolism and insulin action at the blood-brain barrier: Implications for brain health and neurodegenerative diseases. Acta Physiol (Oxf) 2025; 241:e14283. [PMID: 39822067 PMCID: PMC11737474 DOI: 10.1111/apha.14283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
The blood-brain barrier (BBB) is a highly selective, semipermeable barrier critical for maintaining brain homeostasis. The BBB regulates the transport of essential nutrients, hormones, and signaling molecules between the bloodstream and the central nervous system (CNS), while simultaneously protecting the brain from potentially harmful substances and pathogens. This selective permeability ensures that the brain is nourished and shielded from toxins. An exception to this are brain regions, such as the hypothalamus and circumventricular organs, which are irrigated by fenestrated capillaries, allowing rapid and direct response to various blood components. We overview the metabolic functions of the BBB, with an emphasis on the impact of altered glucose metabolism and insulin signaling on BBB in the pathogenesis of neurodegenerative diseases. Notably, endothelial cells constituting the BBB exhibit distinct metabolic characteristics, primarily generating ATP through aerobic glycolysis. This occurs despite their direct exposure to the abundant oxygen in the bloodstream, which typically supports oxidative phosphorylation. The effects of insulin on astrocytes, which form the glial limitans component of the BBB, show a marked sexual dimorphism. BBB nutrient sensing in the hypothalamus, along with insulin signaling, regulates systemic metabolism. Insulin modifies BBB permeability by regulating the expression of tight junction proteins, angiogenesis, and vascular remodeling, as well as modulating blood flow in the brain. The disruptions in glucose and insulin signaling are particularly evident in neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, where BBB breakdown accelerates cognitive decline. This review highlights the critical role of normal glucose metabolism and insulin signaling in maintaining BBB functionality and investigates how disruptions in these pathways contribute to the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiyi Zhu
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of NeurosciencesUniversity of the Basque Country, CIBERNEDLeioaBizkaiaSpain
- IKERBASQUE Basque Foundation for ScienceBilbaoSpain
- Department of Forensic Analytical Toxicology, School of Forensic MedicineChina Medical UniversityShenyangChina
| | - Hui Chen
- School of Life Sciences, Faculty of ScienceUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Chenju Yi
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseGuangzhouChina
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational ResearchShenzhenChina
| |
Collapse
|
22
|
Kang I, Theodoropoulos G, Wangpaichitr M. Targeting the kynurenine pathway: another therapeutic opportunity in the metabolic crosstalk between cancer and immune cells. Front Oncol 2025; 14:1524651. [PMID: 39911818 PMCID: PMC11794083 DOI: 10.3389/fonc.2024.1524651] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
The pivotal role of metabolic reprogramming in cancer-related drug resistance, through the tryptophan-catabolized kynurenine pathway (KP), has been particularly underscored in recent research. This pathway, driven by indoleamine 2,3-dioxygenase 1 (IDO1), facilitates immune evasion and promotes tumor progression by fostering an immunosuppressive environment. In Phase III investigation of the combination of IDO1 inhibition with immune checkpoint inhibitors (ICIs), the combination therapy was not efficacious. In this review, we revisit current advances, explore future directions, and emphasize the importance of dual inhibition of the KP rate-limiting enzymes IDO1 and tryptophan 2,3-dioxygenase-2 (TDO2) in appropriate patient populations. We propose that dual inhibition may maximize the therapeutic potential of KP inhibition. Additionally, we delve into the complex cellular interactions in cancer and metabolic dependencies within the tumor microenvironment (TME). Insights from preclinical studies, recent clinical trials, and promising therapeutic combinations will be discussed to elucidate and promote a clear path forward for the direction of KP research into cancer-related outcomes.
Collapse
Affiliation(s)
- Irene Kang
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, United States
- South Florida VA Foundation for Research and Education, Miami, FL, United States
| | - George Theodoropoulos
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, United States
| | - Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, United States
- South Florida VA Foundation for Research and Education, Miami, FL, United States
- Department of Surgery, Division of Thoracic Surgery, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States
| |
Collapse
|
23
|
Ryu T, Kim K, Asiimwe N, Na CH. Proteomic Insight Into Alzheimer's Disease Pathogenesis Pathways. Proteomics 2025:e202400298. [PMID: 39791267 DOI: 10.1002/pmic.202400298] [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: 09/14/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, but the pathogenesis mechanism is still elusive. Advances in proteomics have uncovered key molecular mechanisms underlying AD, revealing a complex network of dysregulated pathways, including amyloid metabolism, tau pathology, apolipoprotein E (APOE), protein degradation, neuroinflammation, RNA splicing, metabolic dysregulation, and cognitive resilience. This review examines recent proteomic findings from AD brain tissues and biological fluids, highlighting potential biomarkers and therapeutic targets. By examining the proteomic landscape of them, we aim to deepen our understanding of the disease and support developing precision medicine strategies for more effective interventions.
Collapse
Affiliation(s)
- Taekyung Ryu
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kyungdo Kim
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicholas Asiimwe
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chan Hyun Na
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Xu C, Liao M, Zhang S, Chen Y, Shulai X, Wang G, Aa J. The Comorbidity of Depression and Diabetes Is Involved in the Decidual Protein Induced by Progesterone 1 (Depp1) Dysfunction in the Medial Prefrontal Cortex. Metabolites 2025; 15:34. [PMID: 39852377 PMCID: PMC11767987 DOI: 10.3390/metabo15010034] [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: 10/04/2024] [Revised: 12/07/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND There is a high rate of depressive symptoms such as irritability, anhedonia, fatigue, and hypersomnia in patients with type 2 diabetes mellitus (T2DM). However, the causes and underlying mechanisms of the comorbidity of depression and diabetes remain unknown. METHODS For the first time, we identified Decidual protein induced by progesterone 1 (Depp1), also known as DEPP autophagy regulator 1, as a hub gene in both depression and T2DM models. Depp1 levels were increased in the mPFC but not in other brain regions, such as the hippocampus or nucleus accumbens, according to Western blot and PCR assays. RESULTS Glucose dysregulation and synaptic loss occur in both depression and T2DM. The typical hyperglycemia in T2DM was observed in two models of depression, namely, chronic social defeat stress (CSDS) and chronic restraint stress (CRS). Hyperglycemia, which occurred in T2DM, was observed, and metabolomics data clearly showed the perturbation of glucose levels and glucose metabolism in the medial prefrontal cortex (mPFC). Decreased protein levels of BDNF and PSD95 suggested significant synaptic loss in depressed and diabetic mice. CONCLUSION These findings suggest that the comorbidity of depression and diabetes is involved in the dysfunction of Depp1 in the mPFC.
Collapse
Affiliation(s)
| | | | | | | | | | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
25
|
Yip JMX, Chiang GSH, Lee ICJ, Lehming-Teo R, Dai K, Dongol L, Wang LYT, Teo D, Seah GT, Lehming N. Mitochondria and the Repurposing of Diabetes Drugs for Off-Label Health Benefits. Int J Mol Sci 2025; 26:364. [PMID: 39796218 PMCID: PMC11719901 DOI: 10.3390/ijms26010364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
This review describes our current understanding of the role of the mitochondria in the repurposing of the anti-diabetes drugs metformin, gliclazide, GLP-1 receptor agonists, and SGLT2 inhibitors for additional clinical benefits regarding unhealthy aging, long COVID, mental neurogenerative disorders, and obesity. Metformin, the most prominent of these diabetes drugs, has been called the "Drug of Miracles and Wonders," as clinical trials have found it to be beneficial for human patients suffering from these maladies. To promote viral replication in all infected human cells, SARS-CoV-2 stimulates the infected liver cells to produce glucose and to export it into the blood stream, which can cause diabetes in long COVID patients, and metformin, which reduces the levels of glucose in the blood, was shown to cut the incidence rate of long COVID in half for all patients recovering from SARS-CoV-2. Metformin leads to the phosphorylation of the AMP-activated protein kinase AMPK, which accelerates the import of glucose into cells via the glucose transporter GLUT4 and switches the cells to the starvation mode, counteracting the virus. Diabetes drugs also stimulate the unfolded protein response and thus mitophagy, which is beneficial for healthy aging and mental health. Diabetes drugs were also found to mimic exercise and help to reduce body weight.
Collapse
Affiliation(s)
- Joyce Mei Xin Yip
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Grace Shu Hui Chiang
- Well Programme, Alexandra Hospital, National University Health System, Singapore 159964, Singapore; (G.S.H.C.)
| | - Ian Chong Jin Lee
- NUS High School of Mathematics and Science, Singapore 129957, Singapore
| | - Rachel Lehming-Teo
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Kexin Dai
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Lokeysh Dongol
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| | - Laureen Yi-Ting Wang
- Well Programme, Alexandra Hospital, National University Health System, Singapore 159964, Singapore; (G.S.H.C.)
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore 119074, Singapore
- Division of Cardiology, Department of Medicine, Alexandra Hospital, National University Health System, Singapore 159964, Singapore
| | - Denise Teo
- Chi Longevity, Camden Medical Centre #10-04, 1 Orchard Blvd, Singapore 248649, Singapore
| | - Geok Teng Seah
- Clifford Dispensary, 77 Robinson Rd #06-02, Singapore 068896, Singapore
| | - Norbert Lehming
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (R.L.-T.)
| |
Collapse
|
26
|
Wu Q, Wang W, Huang Z, Lin X, Yao M, Cai C, Weng G, Gu Y, Li H, Liu J, Fang J, Li W. Unveiling the molecular mechanisms of Danggui-Shaoyao-San against Alzheimer's disease in APP/PS1 mice via integrating proteomic and metabolomic approaches. Alzheimers Res Ther 2024; 16:251. [PMID: 39563386 DOI: 10.1186/s13195-024-01618-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder for which no effective therapy is currently available. Given that various attempts to target beta-amyloid (Aβ) have been unsuccessful in clinical trials, other potential pathogenic factors such as brain energy metabolism (EM) have attracted increasing attention. Traditional Chinese medicines, including danggui-shaoyao-san (DSS), play a notable role in AD. However, it remains unclear whether DSS exerts therapeutic effects on AD through EM regulation. METHODS In this study, we conducted behavioural tests, Nissl staining, haematoxylin and eosin staining, and thioflavin S staining, in APP/PS1 mice to assess the pharmacodynamic effect of DSS on AD. Subsequently, we integrated the drug target network of herbal ingredients in DSS and evaluated their absorption, distribution, metabolism, excretion, and toxicity properties to identify the core ingredients. We used proteomic and metabolomic approaches to explore the potential mechanisms of action of DSS against AD. Consequently, we verified the mechanism underlying EM using qPCR, western blotting, and ELISA. RESULTS In vivo experimental results revealed that DSS ameliorated cognitive impairment in APP/PS1 mice, attenuated neuronal apoptosis, and reduced Aβ burden. Furthermore, the drug-target network comprised 6,514 drug-target interactions involving 1,118 herbal ingredients and 218 AD genes, of which 253 were identified as the core ingredients in DSS. The proteomic results implied that DSS could act on EM to alleviate AD, and targeted energy metabolomics suggested that DSS regulated 47 metabolites associated with EM. Mechanistically, we found that DSS could regulate the GSK3β/PGC1α signalling pathway to improve brain glucose uptake and mitigate mitochondrial dysfunction and oxidative stress, ultimately promoting EM to treat AD. CONCLUSION Our study is the first to integrate multi-omics approaches to reveal that DSS could regulate the GSK3β/PGC1α signalling pathway to exert therapeutic effects in AD through the promotion of EM, thereby providing new insights into the mechanism of action of DSS against AD.
Collapse
Affiliation(s)
- Qihui Wu
- Clinical Research Center, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, 570100, China.
| | - Wei Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Zhuangzi Huang
- Clinical Research Center, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, 570100, China
| | - Xianghao Lin
- School of Clinical Medicine, Hubei University of Science and Technology, Xianning, 437100, China
| | - Maozhong Yao
- Clinical Research Center, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, 570100, China
| | - Chuipu Cai
- Department of Computer Science, Key Laboratory of Intelligent Manufacturing Technology of Ministry of Education, Shantou University, Shantou, 515000, China
| | - Guohu Weng
- Hainan Clinical Center for Encephalopathy of Chinese Medicine, Haikou, 571000, China
| | - Yong Gu
- Clinical Research Center, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, 570100, China
- Hainan Clinical Center for Encephalopathy of Chinese Medicine, Haikou, 571000, China
| | - Hongying Li
- College of Traditional Chinese Medicine, Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, 571000, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, 529099, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
27
|
Crunkhorn S. Targeting the kynurenine pathway. Nat Rev Drug Discov 2024; 23:816. [PMID: 39333710 DOI: 10.1038/d41573-024-00160-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
|
28
|
Patel MD, Lavekar SS, Jaisalmeria R, Oji S, Jayasi J, Cvetkovic C, Krencik R. Human Astrocytes Synchronize Neural Organoid Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618921. [PMID: 39464076 PMCID: PMC11507866 DOI: 10.1101/2024.10.17.618921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Biological neural networks exhibit synchronized activity within and across interconnected regions of the central nervous system. Understanding how these coordinated networks are established and maintained may reveal therapeutic targets for neurodegeneration and neuromodulation. Here, we tested the influence of astrocytes upon synchronous network activity using human pluripotent stem cell-derived bioengineered neural organoids. This study revealed that astrocytes significantly increase activity within individual organoids and across long distances among numerous rapidly merged organoids via influencing synapses and bioenergetics. Treatment of amyloid protein inhibited synchronous activity during neurodegeneration, yet this can be rescued by propagating activity from neighboring networks. Altogether, this study identifies critical contributions of human astrocytes to biological neural networks and delivers a rapid, reproducible, and scalable model to investigate long-range functional communication of the nervous system in healthy and disease states.
Collapse
|
29
|
Pocivavsek A, Schwarcz R, Erhardt S. Neuroactive Kynurenines as Pharmacological Targets: New Experimental Tools and Exciting Therapeutic Opportunities. Pharmacol Rev 2024; 76:978-1008. [PMID: 39304346 PMCID: PMC11549936 DOI: 10.1124/pharmrev.124.000239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Both preclinical and clinical studies implicate functional impairments of several neuroactive metabolites of the kynurenine pathway (KP), the major degradative cascade of the essential amino acid tryptophan in mammals, in the pathophysiology of neurologic and psychiatric diseases. A number of KP enzymes, such as tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenases (IDO1 and IDO2), kynurenine aminotransferases (KATs), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilic acid oxygenase (3-HAO), and quinolinic acid phosphoribosyltransferase (QPRT), control brain KP metabolism in health and disease and are therefore increasingly considered to be promising targets for the treatment of disorders of the nervous system. Understanding the distribution, cellular expression, and regulation of KP enzymes and KP metabolites in the brain is therefore critical for the conceptualization and implementation of successful therapeutic strategies. SIGNIFICANCE STATEMENT: Studies have implicated the kynurenine pathway of tryptophan in the pathophysiology of neurologic and psychiatric diseases. Key enzymes of the kynurenine pathway regulate brain metabolism in both health and disease, making them promising targets for treating these disorders. Therefore, understanding the distribution, cellular expression, and regulation of these enzymes and metabolites in the brain is critical for developing effective therapeutic strategies. This review endeavors to describe these processes in detail.
Collapse
Affiliation(s)
- Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Robert Schwarcz
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| | - Sophie Erhardt
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina (A.P.); Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland (R.S.); and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.E.)
| |
Collapse
|
30
|
Cortés Malagón EM, López Ornelas A, Olvera Gómez I, Bonilla Delgado J. The Kynurenine Pathway, Aryl Hydrocarbon Receptor, and Alzheimer's Disease. Brain Sci 2024; 14:950. [PMID: 39335444 PMCID: PMC11429728 DOI: 10.3390/brainsci14090950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, mainly affecting elderly individuals. AD is characterized by β-amyloid plaques, abnormal tau tangles, neuronal loss, and metabolic disruptions. Recent studies have revealed the involvement of the kynurenine (KP) pathway and the aryl hydrocarbon receptor (AhR) in AD development. The KP pathway metabolizes tryptophan to produce neuroactive substances like kynurenine, kynurenic acid, and quinolinic acid. In AD, high levels of kynurenine and the neurotoxic quinolinic acid are associated with increased neuroinflammation and excitotoxicity; conversely, reduced levels of kynurenic acid, which acts as a glutamate receptor antagonist, compromise neuroprotection. Research has indicated elevated KP metabolites and enzymes in the hippocampus of AD patients and other tissues such as blood, cerebrospinal fluid, and urine. However, the finding that KP metabolites are AD biomarkers in blood, cerebrospinal fluid, and urine has been controversial. This controversy, stemming from the lack of consideration of the specific stage of AD, details of the patient's treatment, cognitive deficits, and psychiatric comorbidities, underscores the need for more comprehensive research. AhR, a ligand-activated transcription factor, regulates immune response, oxidative stress, and xenobiotic metabolism. Various ligands, including tryptophan metabolites, can activate it. Some studies suggest that AhR activation contributes to AD, while others propose that it provides neuroprotection. This discrepancy may be explained by the specific ligands that activate AhR, highlighting the complex relationship between the KP pathway, AhR activation, and AD, where the same pathway can produce both neuroprotective and harmful effects.
Collapse
Affiliation(s)
- Enoc Mariano Cortés Malagón
- Research Division, Hospital Juárez de México, Mexico City 07760, Mexico; (E.M.C.M.); (A.L.O.); (I.O.G.)
- Genetics Laboratory, Hospital Nacional Homeopático, Mexico City 06800, Mexico
| | - Adolfo López Ornelas
- Research Division, Hospital Juárez de México, Mexico City 07760, Mexico; (E.M.C.M.); (A.L.O.); (I.O.G.)
- Genetics Laboratory, Hospital Nacional Homeopático, Mexico City 06800, Mexico
| | - Irlanda Olvera Gómez
- Research Division, Hospital Juárez de México, Mexico City 07760, Mexico; (E.M.C.M.); (A.L.O.); (I.O.G.)
- Facultad Ciencias de la Salud, Universidad Anáhuac Norte, Estado de México 52786, Mexico
| | - José Bonilla Delgado
- Research Unit, Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS-BINESTAR, Ixtapaluca 56530, Mexico
| |
Collapse
|
31
|
Johnson LA, Macauley SL. Alzheimer's and metabolism wed with IDO1. Science 2024; 385:826-827. [PMID: 39172856 PMCID: PMC11661799 DOI: 10.1126/science.adr5836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Kynurenine pathway inhibition reverses deficits in Alzheimer's mouse models.
Collapse
Affiliation(s)
- Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40508 USA
- Sanders Brown Center on Aging,University of Kentucky, Lexington, KY 40508 USA
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508 USA
- Sanders Brown Center on Aging,University of Kentucky, Lexington, KY 40508 USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508 USA
| |
Collapse
|