1
|
Halder T, Acharya N. Sialic Acid-based Glycoconjugation on Myricetin-encapsulated Cationic Nanocarriers for the Treatment of Alzheimer's. Pharm Res 2025:10.1007/s11095-025-03877-5. [PMID: 40490615 DOI: 10.1007/s11095-025-03877-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 05/21/2025] [Indexed: 06/11/2025]
Abstract
PURPOSE The current study was conducted to develop and evaluate sialic acid grafted cationic myricetin (MY) fabricated nanostructured lipid carrier (Sia-Cat-MY-NLC) for Alzheimer's disease (AD) management. METHODS In-vitro amyloid beta aggregation inhibition and mitochondrial membrane potential of prepared NLCs were observed in SH-SY5Y cells. The transendothelial electrical resistance was measured through hCMEC/D3 cells. Pharmacokinetic and pharmacodynamic studies were conducted to evaluate neuropharmacokinetic parameters and levels of AD hallmarks in AD rats. RESULTS The optimized formulations showed particle sizes (142.26 ± 24.16 nm and 236.3 ± 15.26 nm), zeta potentials (36.5 ± 2.43 mv and -2.4 ± 1.30 mv) respectively for Cat-MY-NLC and Sia-Cat-MY-NLC. Prepared NLCs treatments revealed significant neuroprotective effects in SH-SY5Y cells followed by the ability to cross the in-vitro BBB model. Results of pharmacokinetic studies showed 5.3 and 5.88 folds enhanced bioavailability with Cat-MY-NLC and Sia-Cat-MY-NLC administration respectively. CONCLUSIONS The results of enzymatic analysis showed a significant (p < 0.05) restoration of AD hallmark levels in the brain after Sia-Cat-MY-NLC treatment than Cat-MY-NLC.
Collapse
Affiliation(s)
- Tripti Halder
- Department of Pharmacognosy, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
- School of Pharmaceutical and Population Health Informatics, DIT University, Dehradun, Uttarakhand, 248009, India
| | - Niyati Acharya
- Department of Pharmacognosy, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
2
|
Zhang YY, Hu ZX, Zhang SY, Liu L, Galan MC, Voglmeir J, Ghirardello M. Improved ESI-MS Sensitivity via an Imidazolium Tag (DAPMI-ITag) for Precise Sialic Acid Detection in Human Serum and CMAH-Null Mouse Tissues. Anal Chem 2025. [PMID: 40490447 DOI: 10.1021/acs.analchem.5c00752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
Sialic acids (Sias), consisting primarily of N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), play crucial roles in many biological processes. The detection and quantification of Sias are essential for understanding their roles in health and disease progression. Although numerous techniques have been developed to enhance the specificity and sensitivity of Sias analysis, traditional methods such as derivatization with fluorescent tags coupled with HPLC-MS analysis often suffer from low limits of detection, limiting the quantification of Sias in trace samples. Here, we introduce DAPMI, a novel imidazolium-based ITag for sensitive Sia detection. We demonstrate its utility in the detection and quantification of Sia composition in human serum, and in different tissues from CMAH (cytidine monophosphate-N-acetylneuraminic acid hydroxylase) knockout mice, using ESI-MS analysis and with a limit of detection (LOD) down to the low fmol range. The results showed that both Neu5Ac and Neu5Gc were present in varying proportions in wild-type mice and CMAH heterogeneous mice. Trace amounts of Neu5Gc were also detected in the tissues of CMAH null homogeneous mice (CMAH-/-) and in human blood serum using ESI-ToF-MS, suggesting its presence may be linked to dietary intake of Neu5Gc-containing foods, as Neu5Gc cannot be synthesized endogenously in CMAH-/- mice, and in humans. The DAPMI-ITag and the labeling technology developed in this study significantly improve the sensitivity of Sias detection compared to conventional tags such as o-phenylenediamine (OPD), and provide a new chemical tool for the exploration of Sias' biological roles and their use as biomarkers in different human conditions.
Collapse
Affiliation(s)
- Yao-Yao Zhang
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
- Lipid Technology and Engineering, School of Food Science and Engineering, Henan University of Technology, Lianhua Road 100, 450001 Zhengzhou, China
- School of Chemistry, University of Bristol, Cantock's Close, BS8 1TS Bristol, U.K
| | - Zi-Xuan Hu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - Si-Yu Zhang
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - M Carmen Galan
- School of Chemistry, University of Bristol, Cantock's Close, BS8 1TS Bristol, U.K
| | - Josef Voglmeir
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, 1 Weigang, 210095 Nanjing, China
| | - Mattia Ghirardello
- School of Chemistry, University of Bristol, Cantock's Close, BS8 1TS Bristol, U.K
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Calle Mariano Esquillor, Edificio I+D, 50018 Zaragoza, Spain
| |
Collapse
|
3
|
Henze E, Burkhardt RN, Fox BW, Schwertfeger TJ, Gelsleichter E, Michalski K, Kramer L, Lenfest M, Boesch JM, Lin H, Schroeder FC, Kawate T. ATP-release pannexin channels are gated by lysophospholipids. eLife 2025; 14:RP107067. [PMID: 40309905 PMCID: PMC12045621 DOI: 10.7554/elife.107067] [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] [Indexed: 05/02/2025] Open
Abstract
In addition to its role as cellular energy currency, adenosine triphosphate (ATP) serves as an extracellular messenger that mediates diverse cell-to-cell communication. Compelling evidence supports that ATP is released from cells through pannexins, a family of membrane proteins that form heptameric large-pore channels. However, the activation mechanisms that trigger ATP release by pannexins remain poorly understood. Here, we discover lysophospholipids as endogenous pannexin activators, using activity-guided fractionation of mouse tissue extracts combined with untargeted metabolomics and electrophysiology. We show that lysophospholipids directly and reversibly activate pannexins in the absence of other proteins. Secretomics experiments reveal that lysophospholipid-activated pannexin 1 leads to the release of not only ATP but also other signaling metabolites, such as 5'-methylthioadenosine, which is important for immunomodulation. We also demonstrate that lysophospholipids activate endogenous pannexin 1 in human monocytes, leading to the release of IL-1β through inflammasome activation. Our results provide a connection between lipid metabolism and purinergic signaling, both of which play major roles in immune responses.
Collapse
Affiliation(s)
- Erik Henze
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Russell N Burkhardt
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Bennett William Fox
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Tyler J Schwertfeger
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Eric Gelsleichter
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Kevin Michalski
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Lydia Kramer
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| | - Margret Lenfest
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Jordyn M Boesch
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
- Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell UniversityIthacaUnited States
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Toshimitsu Kawate
- Department of Molecular Medicine, Cornell UniversityIthacaUnited States
| |
Collapse
|
4
|
Zheng L, Xin J, Ye H, Sun N, Gan B, Gong X, Bao S, Xiang M, Wang H, Ni X, Li H, Zhang T. Lactobacillus Johnsonii YH1136 alleviates schizophrenia-like behavior in mice: a gut-microbiota-brain axis hypothesis study. BMC Microbiol 2025; 25:191. [PMID: 40175911 PMCID: PMC11963707 DOI: 10.1186/s12866-025-03893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Based on the microbiota-gut-brain axis (MGBA) hypothesis, probiotics play an increasingly important role in treating various psychiatric disorders. Schizophrenia (SCZ) is a common mental disease with a complex pathogenesis and is challenging to treat. Although studies have elucidated the mechanisms associated with the interactions between the microbiota-gut-brain axis and SCZ, few have specifically used probiotics as a therapeutic intervention for SCZ. Accordingly, the current study determines whether L. johnsonii YH1136 effectively prevents SCZ-like behavior in mice and identifies the associated key microbes and metabolites. An SCZ mouse model was established by intraperitoneal injection of MK-801; L. johnsonii YH1136 was administered via oral gavage. L. johnsonii YH1136 significantly improves abnormal behaviors, including psychomotor hyperactivity and sociability and alleviates aberrant enzyme expression associated with tryptophan metabolism in SCZ mice. Additionally, L. johnsonii YH1136 upregulates hippocampal brain-derived neurotrophic factor (BDNF) levels while downregulating tryptophan 2,3-dioxygenase (TDO2), indoleamine-pyrrole 2,3-dioxygenase 1 (IDO1), kynurenine aminotransferase 1 (KAT1). Subsequent 16S rRNA sequencing of intestinal contents suggests that L. johnsonii YH1136 modulates the gut flora structure and composition by increasing the relative abundance of Lactobacillus and decreasing Dubosiella in SCZ mice. N-acetylneuraminic acid and hypoxanthine are the key serum metabolites mediating the interaction between the MGBA and SCZ. These results partially reveal the mechanism underlying the effects of L. johnsonii YH1136 on SCZ-like behavior in mice, supporting the development of therapeutic L. johnsonii probiotic formulations against SCZ.
Collapse
Affiliation(s)
- Liqin Zheng
- School of Life Science and Technology, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, Sichuan Institute for Brain Science and Brain-Inspired Intelligence, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, West Hi-Tech Zone, Chengdu , Sichuan, 611731, China
| | - Jinge Xin
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huiqian Ye
- The Fourth People's Hospital of Ya'an, 7 Qingxi Road Ya'an 625000, Yucheng ZoneSichuan, China
| | - Ning Sun
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Baoxing Gan
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Gong
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shusheng Bao
- School of Life Science and Technology, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Lab for Neuroinformation, Sichuan Institute for Brain Science and Brain-Inspired Intelligence, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, West Hi-Tech Zone, Chengdu , Sichuan, 611731, China
| | - Min Xiang
- The Fourth People's Hospital of Ya'an, 7 Qingxi Road Ya'an 625000, Yucheng ZoneSichuan, China
| | - Hesong Wang
- Baiyun Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueqin Ni
- Animal Microecology Institute College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hao Li
- The Fourth People's Hospital of Ya'an, 7 Qingxi Road Ya'an 625000, Yucheng ZoneSichuan, China.
| | - Tao Zhang
- School of Life Science and Technology, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China.
- MOE Key Lab for Neuroinformation, Sichuan Institute for Brain Science and Brain-Inspired Intelligence, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, West Hi-Tech Zone, Chengdu , Sichuan, 611731, China.
| |
Collapse
|
5
|
Sharma V, Fernando V, Zheng X, Choi ES, Sweef O, Thomas V, Szpendyk J, Furuta S. Immunogenic shift of arginine metabolism triggers systemic metabolic and immunological reprogramming to suppress HER2 + breast cancer. Cancer Metab 2025; 13:15. [PMID: 40114277 PMCID: PMC11927160 DOI: 10.1186/s40170-025-00384-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Arginine metabolism in tumors is often shunted into the pathway producing pro-tumor and immune suppressive polyamines (PAs), while downmodulating the alternative nitric oxide (NO) synthesis pathway. Aiming to correct arginine metabolism in tumors, arginine deprivation therapy and inhibitors of PA synthesis have been developed. Despite some therapeutic advantages, these approaches have often yielded severe side effects, making it necessary to explore an alternative strategy. We previously reported that supplementing sepiapterin (SEP), the endogenous precursor of tetrahydrobiopterin (BH4, the essential NO synthase cofactor), could correct arginine metabolism in tumor cells and tumor-associated macrophages (TAMs) and induce their metabolic and phenotypic reprogramming. We saw that oral SEP treatment effectively suppressed the growth of HER2-positive mammary tumors in animals. SEP also has no reported dose-dependent toxicity in clinical trials for metabolic disorders. In the present study, we tested our hypothesis that a long-term administration of SEP to individuals susceptible to HER2-positive mammary tumor would protect them against tumor occurrence. METHODS We administered SEP, in comparison to control DMSO, to MMTV-neu mice susceptible to HER2-positive mammary tumors for 8 months starting at their pre-pubertal stage. We monitored tumor onsets to determine the rate of tumor-free survival. After 8 months of treatment, we grouped animals into DMSO treatment with or without tumors and SEP treatment with or without tumors. We analyzed blood metabolites, PBMC, and bone marrow of DMSO vs. SEP treated animals. RESULTS We found that a long-term use of SEP in animals susceptible to HER2-positive mammary tumors effectively suppressed tumor occurrence. These SEP-treated animals had undergone reprogramming of the systemic metabolism and immunity, elevating total T cell counts in the circulation and bone marrow. Given that bone marrow-resident T cells are mostly memory T cells, it is plausible that chronic SEP treatment promoted memory T cell formation, leading to a potent tumor prevention. CONCLUSIONS These findings suggest the possible roles of the SEP/BH4/NO axis in promoting memory T cell formation and its potential therapeutic utility for preventing HER2-positive breast cancer.
Collapse
Affiliation(s)
- Vandana Sharma
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA
- Department of Zoology and Physiology, University of Wyoming, 1000 E. University Ave, Biological Science Building, Room 319F, Laramie, WY, 82071, USA
| | - Veani Fernando
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus Barbara Davis Center, Mail Stop B115, 1775 Aurora Court, Aurora, CO, 80045, USA
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA
| | - Eun-Seok Choi
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Osama Sweef
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Venetia Thomas
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Justin Szpendyk
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH, 43614, USA.
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA.
| |
Collapse
|
6
|
Gillenwater LA, Galbraith MD, Rachubinski AL, Eduthan NP, Sullivan KD, Espinosa JM, Costello JC. Integrated analysis of immunometabolic interactions in Down syndrome. SCIENCE ADVANCES 2024; 10:eadq3073. [PMID: 39671500 PMCID: PMC11641111 DOI: 10.1126/sciadv.adq3073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Down syndrome (DS), caused by trisomy 21 (T21), results in immune and metabolic dysregulation. People with DS experience co-occurring conditions at higher rates than the euploid population. However, the interplay between immune and metabolic alterations and the clinical manifestations of DS are poorly understood. Here, we report an integrated analysis of immunometabolic pathways in DS. Using multi-omics data, we infered cytokine-metabolite relationships mediated by specific transcriptional programs. We observed increased mediation of immunometabolic interactions in those with DS compared to euploid controls by genes in interferon response, heme metabolism, and oxidative phosphorylation. Unsupervised clustering of immunometabolic relationships in people with DS revealed subgroups with different frequencies of co-occurring conditions. Across the subgroups, we observed distinct mediation by DNA repair, Hedgehog signaling, and angiogenesis. The molecular stratification associates with the clinical heterogeneity observed in DS, suggesting that integrating multiple omic profiles reveals axes of coordinated dysregulation specific to DS co-occurring conditions.
Collapse
Affiliation(s)
- Lucas A. Gillenwater
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Matthew D. Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angela L. Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelly D. Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James C. Costello
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Computational Bioscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
7
|
Liu Z, Xiang P, Zeng S, Weng P, Wen Y, Zhang W, Hu H, Zhao D, Ma L, Yu C. N-Acetylneuraminic acid triggers endothelial pyroptosis and promotes atherosclerosis progression via GLS2-mediated glutaminolysis pathway. Cell Death Discov 2024; 10:467. [PMID: 39537619 PMCID: PMC11561128 DOI: 10.1038/s41420-024-02233-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Vascular endothelial injury initiates atherosclerosis (AS) progression. N-Acetylneuraminic acid (Neu5Ac) metabolic disorder was found to intensify endothelial mitochondrial damage. And GLS2-associated glutaminolysis disorder contributed to mitochondrial dysfunction. However, mechanisms underlying Neu5Ac-associated mitochondrial dysfunction as well as its association with GLS2 remains unclear. In this study, we constructed GLS2-/-ApoE-/- mice by using HBLV-GLS2 shRNA injection. And methods like immunofluorescence, western blotting, transmission electron microscopy were applied to detect profiles of endothelial injury and AS progression both in vivo and in vitro. We demonstrated that Neu5Ac accumulation increased GLS2 expression and promoted glutaminolysis disorder, which further induced endothelial mitochondrial dysfunction via a pyroptosis-dependent pathway in vivo and in vitro. Mechanically, Neu5Ac interacted with SIRT3 and led to FOXO3a deacetylation and phosphorylation, further facilitated c-Myc antagonism and ultimately increased GLS2 levels. Inhibition of GLS2 could improve mitochondrial function and mitigate pyroptosis process. In addition, blocking Neu5Ac production using neuraminidases (NEUs) inhibitor could rescue endothelial damage and alleviate AS development in ApoE-/- mice. These findings proposed that Neu5Ac induced GLS2-mediated glutaminolysis disorder and then promoted mitochondrial dysfunction in a pyroptosis-dependent pathway. Targeting GLS2 or inhibiting Neu5Ac production could prevent AS progression.
Collapse
Affiliation(s)
- Zhaohong Liu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Peng Xiang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Shengmei Zeng
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Ping Weng
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Yilin Wen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Hui Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Dezhang Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Research Center for Innovative Pharmaceutical and Experiment Analysis Technology, Chongqing, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.
| |
Collapse
|
8
|
Sharma V, Fernando V, Zheng X, Sweef O, Choi ES, Thomas V, Furuta S. Immunogenic shift of arginine metabolism triggers systemic metabolic and immunological reprogramming to prevent HER2+ breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619827. [PMID: 39484369 PMCID: PMC11527010 DOI: 10.1101/2024.10.23.619827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Arginine metabolism in tumors is often shunted into the pathway producing pro-tumor and immune suppressive polyamines (PAs), while downmodulating the alternative nitric oxide (NO) synthesis pathway. Aiming to correct arginine metabolism in tumors, arginine deprivation therapy and inhibitors of PA synthesis have been developed. Despite some therapeutic advantages, these approaches have often yielded severe side effects, making it necessary to explore an alternative strategy. We previously reported that supplementing SEP, the endogenous precursor of BH4 (the essential NO synthase cofactor), could correct arginine metabolism in tumor cells and tumor-associated macrophages (TAMs) and induce their metabolic and phenotypic reprogramming. We saw that oral SEP treatment effectively suppressed the growth of HER2-positive mammary tumors in animals. SEP also has no reported dose-dependent toxicity in clinical trials for metabolic disorders. In the present study, we report that a long-term use of SEP in animals susceptible to HER2-positive mammary tumors effectively prevented tumor occurrence. These SEP-treated animals had undergone reprogramming of the systemic metabolism and immunity, elevating total T cell counts in the circulation and bone marrow. Given that bone marrow-resident T cells are mostly memory T cells, it is plausible that chronic SEP treatment promoted memory T cell formation, leading to a potent tumor prevention. These findings suggest the possible roles of the SEP/BH4/NO axis in promoting memory T cell formation and its potential therapeutic utility for preventing HER2-positive breast cancer.
Collapse
Affiliation(s)
- Vandana Sharma
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
- Department of Zoology and Physiology, University of Wyoming, 1000 E. University Ave, Biological Science Building, Room 319F, Laramie, WY 82071
| | - Veani Fernando
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus Barbara Davis Center, Mail Stop B115, 1775 Aurora Court, Aurora, Colorado 80045
| | - Xunzhen Zheng
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
| | - Osama Sweef
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH 44109
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Eun-Seok Choi
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH 44109
| | - Venetia Thomas
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH 44109
| | - Saori Furuta
- Department of Cell & Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave. Toledo, OH 43614, USA
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, 2500 MetroHealth Drive, Cleveland, OH 44109
| |
Collapse
|
9
|
Schwartz M, Colaiuta SP. Boosting peripheral immunity to fight neurodegeneration in the brain. Trends Immunol 2024; 45:760-767. [PMID: 39358094 DOI: 10.1016/j.it.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 10/04/2024]
Abstract
Reciprocal communication between the brain and the immune system is essential for maintaining lifelong brain function. This interaction is mediated, at least in part, by immune cells recruited from both the circulation and niches at the borders of the brain. Here, we describe how immune exhaustion and senescence, even if not primary causative factors, can accelerate neurodegenerative diseases. We emphasize the role of a compromised peripheral immune system in driving neurodegeneration and discuss strategies for harnessing peripheral immunity to effectively treat neurodegenerative diseases, including the underlying mechanisms and opportunities for clinical translation. Specifically, we highlight the potential of boosting the immune system by blocking inhibitory checkpoint molecules to harness reparative immune cells in helping the brain to fight against neurodegeneration.
Collapse
Affiliation(s)
- Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | | |
Collapse
|
10
|
Zhang S, Yuan J, Sun Y, Wu F, Liu Z, Zhai F, Zhang Y, Somekh J, Peleg M, Zhu YC, Huang Z, for the Alzheimer’s Disease Neuroimaging Initiative and the Australian Imaging Biomarkers and Lifestyle Study of Aging. Machine learning on longitudinal multi-modal data enables the understanding and prognosis of Alzheimer's disease progression. iScience 2024; 27:110263. [PMID: 39040055 PMCID: PMC11261013 DOI: 10.1016/j.isci.2024.110263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 07/24/2024] Open
Abstract
Alzheimer's disease (AD) is a complex pathophysiological disease. Allowing for heterogeneity, not only in disease manifestations but also in different progression patterns, is critical for developing effective disease models that can be used in clinical and research settings. We introduce a machine learning model for identifying underlying patterns in Alzheimer's disease (AD) trajectory using longitudinal multi-modal data from the ADNI cohort and the AIBL cohort. Ten biologically and clinically meaningful disease-related states were identified from data, which constitute three non-overlapping stages (i.e., neocortical atrophy [NCA], medial temporal atrophy [MTA], and whole brain atrophy [WBA]) and two distinct disease progression patterns (i.e., NCA → WBA and MTA → WBA). The index of disease-related states provided a remarkable performance in predicting the time to conversion to AD dementia (C-Index: 0.923 ± 0.007). Our model shows potential for promoting the understanding of heterogeneous disease progression and early predicting the conversion time to AD dementia.
Collapse
Affiliation(s)
- Suixia Zhang
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, P.R. China
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830017, China
| | - Jing Yuan
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yu Sun
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, P.R. China
| | - Fei Wu
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, P.R. China
| | - Ziyue Liu
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Feifei Zhai
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yaoyun Zhang
- DAMO Academy, Alibaba Group, 969 Wenyixi Rd, Hangzhou 310058, P.R. China
| | - Judith Somekh
- Department of Information Systems, University of Haifa, Haifa 3303220, Israel
| | - Mor Peleg
- Department of Information Systems, University of Haifa, Haifa 3303220, Israel
| | - Yi-Cheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Zhengxing Huang
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, P.R. China
| | - for the Alzheimer’s Disease Neuroimaging Initiative and the Australian Imaging Biomarkers and Lifestyle Study of Aging
- Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, P.R. China
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
- DAMO Academy, Alibaba Group, 969 Wenyixi Rd, Hangzhou 310058, P.R. China
- Department of Information Systems, University of Haifa, Haifa 3303220, Israel
- Department of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830017, China
| |
Collapse
|
11
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
12
|
Teng F, Sun J, Chen Z, Li H. Genetically determined dietary habits and risk of Alzheimer's disease: a Mendelian randomization study. Front Nutr 2024; 11:1415555. [PMID: 38887501 PMCID: PMC11180739 DOI: 10.3389/fnut.2024.1415555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Background Emerging evidence have suggested that dietary habits have potential implication on the development of Alzheimer's disease (AD). However, elucidating the causal relationship between specific dietary factors and AD risk remains a challenge. Therefore, our study endeavors to investigate the causal association between dietary habits and the risk of AD. Materials and methods We analyzed data on 231 dietary habits sourced from the UK Biobank and MRC-IEU, and AD data obtained from the FinnGen database. Employing a framework based on the classic two-sample Mendelian randomization (MR) study, we utilized the inverse-variance weighted (IVW) method as the primary analysis. Additionally, we conducted Steiger filtering and other methods to mitigate horizontal pleiotropy. The robustness of our overall findings was confirmed through multiple sensitivity analysis methods, and forward MR and reverse MR to address potential reverse causality bias. Results Our study evaluated the causal effect between 231 dietary habits involving over 500,000 participants of European ancestry, and 10,520 AD cases. Only oily fish intake demonstrated a significant protective causal relationship with AD following FDR correction (raw p-value = 1.28e-4, FDR p-value = 0.011, OR = 0.60, 95%CI: 0.47-0.78). Additionally, six dietary habits potentially influenced AD risk, with protective causal effects observed for average monthly intake of other alcoholic drinks (raw p-value = 0.024, FDR p-value = 0.574, OR = 0.57, 95%CI: 0.35-0.93) and tea intake (raw p-value = 0.047, FDR p-value = 0.581, OR = 0.78, 95%CI: 0.603-1.00). Conversely, detrimental causal effects were observed for the average weekly champagne plus white wine intake (raw p-value = 0.006, FDR p-value = 0.243, OR = 2.96, 95%CI: 1.37-6.38), Danish pastry intake (raw p-value = 0.036, FDR p-value = 0.574, OR = 13.33, 95%CI: 1.19-149.69), and doughnut intake (raw p-value = 0.039, FDR p-value = 0.574, OR = 7.41, 95%CI: 1.11-49.57). Moreover, the protective effect of goat's cheese intake phenotype exhibited statistical significance only in the IVW method (raw p-value<0.05). Conclusion Our results provide genetic support for a protective causal effect of oily fish intake on AD risk. Additionally, average monthly intake of other alcoholic drinks and tea consumption were also related with a lower risk of AD. Conversely, average weekly champagne plus white wine intake, Danish pastry intake, and doughnut intake were causally associated with increased risk of AD.
Collapse
Affiliation(s)
- Fei Teng
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jiahui Sun
- Wangjing Hospital of China Academy of Chinese Medicine Sciences, Beijing, China
| | - Zheyu Chen
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hao Li
- Wangjing Hospital of China Academy of Chinese Medicine Sciences, Beijing, China
| |
Collapse
|
13
|
Fang Z, Liu R, Xie J, He JC. Molecular mechanism of renal lipid accumulation in diabetic kidney disease. J Cell Mol Med 2024; 28:e18364. [PMID: 38837668 PMCID: PMC11151220 DOI: 10.1111/jcmm.18364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 06/07/2024] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of end stage renal disease with unmet clinical demands for treatment. Lipids are essential for cell survival; however, renal cells have limited capability to metabolize overloaded lipids. Dyslipidaemia is common in DKD patients and renal ectopic lipid accumulation is associated with disease progression. Unveiling the molecular mechanism involved in renal lipid regulation is crucial for exploring potential therapeutic targets. In this review, we focused on the mechanism underlying cholesterol, oxysterol and fatty acid metabolism disorder in the context of DKD. Specific regulators of lipid accumulation in different kidney compartment and TREM2 macrophages, a lipid-related macrophages in DKD, were discussed. The role of sodium-glucose transporter 2 inhibitors in improving renal lipid accumulation was summarized.
Collapse
Affiliation(s)
- Zhengying Fang
- Department of Nephrology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Barbara T. Murphy Division of Nephrology, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ruijie Liu
- Barbara T. Murphy Division of Nephrology, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Jingyuan Xie
- Department of Nephrology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - John Cijiang He
- Barbara T. Murphy Division of Nephrology, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Renal SectionJames J Peters Veterans Affair Medical CenterBronxNew YorkUSA
| |
Collapse
|
14
|
Xiao YL, Gong Y, Qi YJ, Shao ZM, Jiang YZ. Effects of dietary intervention on human diseases: molecular mechanisms and therapeutic potential. Signal Transduct Target Ther 2024; 9:59. [PMID: 38462638 PMCID: PMC10925609 DOI: 10.1038/s41392-024-01771-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Diet, serving as a vital source of nutrients, exerts a profound influence on human health and disease progression. Recently, dietary interventions have emerged as promising adjunctive treatment strategies not only for cancer but also for neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, and metabolic disorders. These interventions have demonstrated substantial potential in modulating metabolism, disease trajectory, and therapeutic responses. Metabolic reprogramming is a hallmark of malignant progression, and a deeper understanding of this phenomenon in tumors and its effects on immune regulation is a significant challenge that impedes cancer eradication. Dietary intake, as a key environmental factor, can influence tumor metabolism. Emerging evidence indicates that dietary interventions might affect the nutrient availability in tumors, thereby increasing the efficacy of cancer treatments. However, the intricate interplay between dietary interventions and the pathogenesis of cancer and other diseases is complex. Despite encouraging results, the mechanisms underlying diet-based therapeutic strategies remain largely unexplored, often resulting in underutilization in disease management. In this review, we aim to illuminate the potential effects of various dietary interventions, including calorie restriction, fasting-mimicking diet, ketogenic diet, protein restriction diet, high-salt diet, high-fat diet, and high-fiber diet, on cancer and the aforementioned diseases. We explore the multifaceted impacts of these dietary interventions, encompassing their immunomodulatory effects, other biological impacts, and underlying molecular mechanisms. This review offers valuable insights into the potential application of these dietary interventions as adjunctive therapies in disease management.
Collapse
Affiliation(s)
- Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yue Gong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
15
|
Seah C, Signer R, Deans M, Bader H, Rusielewicz T, Hicks EM, Young H, Cote A, Townsley K, Xu C, Hunter CJ, McCarthy B, Goldberg J, Dobariya S, Holtzherimer PE, Young KA, Noggle SA, Krystal JH, Paull D, Girgenti MJ, Yehuda R, Brennand KJ, Huckins LM. Common genetic variation impacts stress response in the brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573459. [PMID: 38234801 PMCID: PMC10793429 DOI: 10.1101/2023.12.27.573459] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
To explain why individuals exposed to identical stressors experience divergent clinical outcomes, we determine how molecular encoding of stress modifies genetic risk for brain disorders. Analysis of post-mortem brain (n=304) revealed 8557 stress-interactive expression quantitative trait loci (eQTLs) that dysregulate expression of 915 eGenes in response to stress, and lie in stress-related transcription factor binding sites. Response to stress is robust across experimental paradigms: up to 50% of stress-interactive eGenes validate in glucocorticoid treated hiPSC-derived neurons (n=39 donors). Stress-interactive eGenes show brain region- and cell type-specificity, and, in post-mortem brain, implicate glial and endothelial mechanisms. Stress dysregulates long-term expression of disorder risk genes in a genotype-dependent manner; stress-interactive transcriptomic imputation uncovered 139 novel genes conferring brain disorder risk only in the context of traumatic stress. Molecular stress-encoding explains individualized responses to traumatic stress; incorporating trauma into genomic studies of brain disorders is likely to improve diagnosis, prognosis, and drug discovery.
Collapse
|
16
|
Tsitsou-Kampeli A, Suzzi S, Kenigsbuch M, Satomi A, Strobelt R, Singer O, Feldmesser E, Purnapatre M, Colaiuta SP, David E, Cahalon L, Hahn O, Wyss-Coray T, Shaul Y, Amit I, Schwartz M. Cholesterol 24-hydroxylase at the choroid plexus contributes to brain immune homeostasis. Cell Rep Med 2023; 4:101278. [PMID: 37944529 PMCID: PMC10694665 DOI: 10.1016/j.xcrm.2023.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/26/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
The choroid plexus (CP) plays a key role in remotely controlling brain function in health, aging, and disease. Here, we report that CP epithelial cells express the brain-specific cholesterol 24-hydroxylase (CYP46A1) and that its levels are decreased under different mouse and human brain conditions, including amyloidosis, aging, and SARS-CoV-2 infection. Using primary mouse CP cell cultures, we demonstrate that the enzymatic product of CYP46A1, 24(S)-hydroxycholesterol, downregulates inflammatory transcriptomic signatures within the CP, found here to be elevated across multiple neurological conditions. In vitro, the pro-inflammatory cytokine tumor necrosis factor α (TNF-α) downregulates CYP46A1 expression, while overexpression of CYP46A1 or its pharmacological activation in mouse CP organ cultures increases resilience to TNF-α. In vivo, overexpression of CYP46A1 in the CP in transgenic mice with amyloidosis is associated with better cognitive performance and decreased brain inflammation. Our findings suggest that CYP46A1 expression in the CP impacts the role of this niche as a guardian of brain immune homeostasis.
Collapse
Affiliation(s)
| | - Stefano Suzzi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | - Mor Kenigsbuch
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Akisawa Satomi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Romano Strobelt
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Oded Singer
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ester Feldmesser
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liora Cahalon
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
17
|
Choi JJ, Koscik RL, Jonaitis EM, Panyard DJ, Morrow AR, Johnson SC, Engelman CD, Schmitz LL. Assessing the Biological Mechanisms Linking Smoking Behavior and Cognitive Function: A Mediation Analysis of Untargeted Metabolomics. Metabolites 2023; 13:1154. [PMID: 37999250 PMCID: PMC10673384 DOI: 10.3390/metabo13111154] [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: 10/14/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023] Open
Abstract
(1) Smoking is the most significant preventable health hazard in the modern world. It increases the risk of vascular problems, which are also risk factors for dementia. In addition, toxins in cigarettes increase oxidative stress and inflammation, which have both been linked to the development of Alzheimer's disease and related dementias (ADRD). This study identified potential mechanisms of the smoking-cognitive function relationship using metabolomics data from the longitudinal Wisconsin Registry for Alzheimer's Prevention (WRAP). (2) 1266 WRAP participants were included to assess the association between smoking status and four cognitive composite scores. Next, untargeted metabolomic data were used to assess the relationships between smoking and metabolites. Metabolites significantly associated with smoking were then tested for association with cognitive composite scores. Total effect models and mediation models were used to explore the role of metabolites in smoking-cognitive function pathways. (3) Plasma N-acetylneuraminate was associated with smoking status Preclinical Alzheimer Cognitive Composite 3 (PACC3) and Immediate Learning (IMM). N-acetylneuraminate mediated 12% of the smoking-PACC3 relationship and 13% of the smoking-IMM relationship. (4) These findings provide links between previous studies that can enhance our understanding of potential biological pathways between smoking and cognitive function.
Collapse
Affiliation(s)
- Jerome J. Choi
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA; (J.J.C.); (A.R.M.)
| | - Rebecca L. Koscik
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA; (R.L.K.); (E.M.J.)
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Erin M. Jonaitis
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA; (R.L.K.); (E.M.J.)
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Daniel J. Panyard
- Department of Genetics, School of Medicine, Stanford University, Palo Alto, CA 94305, USA;
| | - Autumn R. Morrow
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA; (J.J.C.); (A.R.M.)
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA; (R.L.K.); (E.M.J.)
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison, Madison, WI 53792, USA
- William S. Middleton Memorial Veterans Hospital, Middleton, WI 53705, USA
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Corinne D. Engelman
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53726, USA; (J.J.C.); (A.R.M.)
| | - Lauren L. Schmitz
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI 53706, USA;
| |
Collapse
|
18
|
Shoob S, Buchbinder N, Shinikamin O, Gold O, Baeloha H, Langberg T, Zarhin D, Shapira I, Braun G, Habib N, Slutsky I. Deep brain stimulation of thalamic nucleus reuniens promotes neuronal and cognitive resilience in an Alzheimer's disease mouse model. Nat Commun 2023; 14:7002. [PMID: 37919286 PMCID: PMC10622498 DOI: 10.1038/s41467-023-42721-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
The mechanisms that confer cognitive resilience to Alzheimer's Disease (AD) are not fully understood. Here, we describe a neural circuit mechanism underlying this resilience in a familial AD mouse model. In the prodromal disease stage, interictal epileptiform spikes (IESs) emerge during anesthesia in the CA1 and mPFC regions, leading to working memory disruptions. These IESs are driven by inputs from the thalamic nucleus reuniens (nRE). Indeed, tonic deep brain stimulation of the nRE (tDBS-nRE) effectively suppresses IESs and restores firing rate homeostasis under anesthesia, preventing further impairments in nRE-CA1 synaptic facilitation and working memory. Notably, applying tDBS-nRE during the prodromal phase in young APP/PS1 mice mitigates age-dependent memory decline. The IES rate during anesthesia in young APP/PS1 mice correlates with later working memory impairments. These findings highlight the nRE as a central hub of functional resilience and underscore the clinical promise of DBS in conferring resilience to AD pathology by restoring circuit-level homeostasis.
Collapse
Affiliation(s)
- Shiri Shoob
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Nadav Buchbinder
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ortal Shinikamin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Or Gold
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Halit Baeloha
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Tomer Langberg
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Daniel Zarhin
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Gabriella Braun
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
19
|
Xiang P, Chen Q, Chen L, Lei J, Yuan Z, Hu H, Lu Y, Wang X, Wang T, Yu R, Zhang W, Zhang J, Yu C, Ma L. Metabolite Neu5Ac triggers SLC3A2 degradation promoting vascular endothelial ferroptosis and aggravates atherosclerosis progression in ApoE -/-mice. Theranostics 2023; 13:4993-5016. [PMID: 37771765 PMCID: PMC10526676 DOI: 10.7150/thno.87968] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Background: Atherosclerosis (AS) is still the major cause of cardiovascular disease (CVD) as well as stroke. Endothelial metabolic disorder has been found to be activated and then promote endothelial cells (ECs) injury, which is regarded to initiate AS progression. N-acetylneuraminic acid (Neu5Ac), a metabolite produced by hexosamine-sialic acid pathway branching from glucose metabolism, was presented as a notable biomarker of CVD and is positively correlated with ECs function. However, few studies explain whether Neu5Ac regulate AS progression by affecting EC function as well as its involved mechanisms are still unknown. Methods: Here, we mimicked an animal model in ApoE-/- mice which displaying similar plasma Neu5Ac levels with AS model to investigate its effect on AS progression. Results: We found that Neu5Ac exacerbated plaques area and increased lipids in plasma in absence of HFD feeding, and ECs inflammatory injury was supposed as the triggering factor upon Neu5Ac treatment with increasing expression of IL-1β, ICAM-1, and promoting ability of monocyte adhesion to ECs. Mechanistic studies showed that Neu5Ac facilitated SLC3A2 binding to ubiquitin and then triggered P62 mediated degradation, further leading to accumulation of lipid peroxidation in ECs. Fer-1 could inhibit ECs injury and reverse AS progression induced by Neu5Ac in ApoE-/- mice. Interestingly, mitochondrial dysfunction was also partly participated in ECs injury after Neu5Ac treatment and been reversed by Fer-1. Conclusions: Together, our study unveils a new mechanism by which evaluated metabolite Neu5Ac could promote SLC3A2 associated endothelial ferroptosis to activate ECs injury and AS plaque progression, thus providing a new insight into the role of Neu5Ac-ferroptosis pathway in AS. Also, our research revealed that pharmacological inhibition of ferroptosis may provide a novel therapeutic strategy for premature AS.
Collapse
Affiliation(s)
- Peng Xiang
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Qingqiu Chen
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Le Chen
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Jin Lei
- Xi'an No.1 Hospital, The First Affiliated Hospital of Northwest University, Xi'an, 710002, Shaanxi, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Hui Hu
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
| | - Yining Lu
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Xianmin Wang
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Tingting Wang
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Ruihong Yu
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Jun Zhang
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| | - Limei Ma
- College of Pharmacy, Chongqing Medical University, 400010, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, 400010, Chongqing, China
| |
Collapse
|
20
|
Kiani L. Obesity-associated immune exhaustion accelerates AD pathology. Nat Rev Neurol 2023; 19:257. [PMID: 37002365 DOI: 10.1038/s41582-023-00805-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|