1
|
Latina V, De Introna M, Malerba F, Florio R, Balzamino BO, Di Natale G, Sciacca MFM, Pappalardo G, Micera A, Pignataro A, Calissano P, Amadoro G. Acute targeting of N-terminal tau protein has long-lasting beneficial effects in Tg2576 APP/Aβ mouse model by reducing cognitive impairment, cerebral Aβ-amyloidosis, synaptic remodeling and microgliosis later in life. Acta Neuropathol Commun 2025; 13:121. [PMID: 40442822 PMCID: PMC12123992 DOI: 10.1186/s40478-025-02022-y] [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: 03/13/2025] [Accepted: 04/30/2025] [Indexed: 06/02/2025] Open
Abstract
Even though the number of patients suffering from Alzheimer's Disease (AD) is rapidly growing worldwide, only a few symptomatic treatments have been approved for clinical use, pointing out the urgent need for more effective disease-modifying therapies that actually alter the progression of this neurodegenerative disorder which is characterized by co-occurence of both Amyloid beta (Aβ) and tau neuropathologies. Preclinical and clinical evidence suggests that a link between Aβ and tau drives the entire continuum of AD pathobiology. 12A12 is a monoclonal antibody (mAb) which offers neuroprotection into two transgenic lines of AD, including Tg2576 that overexpresses Swedish mutation (KM670/671NL) of Amyloid Precursor Protein (APP, isoform 695) and 3xTg (APP Swedish, MAPT P301L, and PSEN1 M146V), by targeting the 20-22kDa N-terminal tau fragments (NH2htau). In particular, acute (over 14 days with 4 doses), intravenous injection of 12A12mAb leads to significant improvement of cognitive, biochemical and histopathological AD signs in symptomatic 6-month-old Tg2576, a well-established transgenic mouse model that mimics the human amyloidosis with an age-dependent Aβ accumulation/aggregation and plaque deposition. Here, we report that Tg2576 mice, immunized with 12A12mAb at 6 months of age and returned to their home cage for additional 3 months, exhibit preserved spatial memory despite the anticipated interruption of antibody administration (discontinuous treatment). This enduring beneficial effect on memory deficit (up to 90 days after the last injection) is accompanied by normalization in the synaptic imbalance and microgliosis along with decrease of the most toxic A11-positive prefibrillar oligomers and inverse increase in 4kDa monomeric form(s) of Aβ 1-42. These findings reveal that: (i) soluble, pathogenetic tau specie(s) located at the N-terminal domain of protein early synergizes with Aβ in driving the progression of AD neuropathology; (ii) transient immunoneutralization of the NH2htau following short-term treatment with 12A12mAb exerts preventive, long-lasting neuroprotective effects, at least in part by interfering at "pre-plaque" stage with the progressive deposition of insoluble, fibrillar Aβ via a shift of its aggregation pathway into its less harmful, unaggregated monomeric forms. Taken together, these findings represent a strong rationale for the advancement of 12A12mAb to clinical stage aiming at preventing the Aβ-dependent neurodegeneration by lowering the cerebral levels of NH2htau in humans suffering from chronic, slow-progressing AD.
Collapse
Affiliation(s)
- Valentina Latina
- Institute of Translational Pharmacology (IFT)-National Research Council (CNR), Via Fosso del Cavaliere 100, 00133, Rome, Italy
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
| | - Margherita De Introna
- Centro Di Ricerca Europeo Sul Cervello (CERC), IRCCS Santa Lucia Foundation (FSL), Via Fosso del Fiorano 43-44, 00143, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Via Montpellier, 1, 00133, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
- Institute of Nanotechnology Campus Ecotekne- National Research Council (CNR), Via Monteroni, 73100, Lecce, Italy
| | - Rita Florio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
| | - Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184, Rome, Italy
| | - Giuseppe Di Natale
- Institute of Crystallography (IC)-National Research Council (CNR), Via Paolo Gaifami 18, 95126, Catania, Italy
| | | | - Giuseppe Pappalardo
- Institute of Crystallography (IC)-National Research Council (CNR), Via Paolo Gaifami 18, 95126, Catania, Italy
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184, Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT)-National Research Council (CNR), Via Fosso del Cavaliere 100, 00133, Rome, Italy
- Centro Di Ricerca Europeo Sul Cervello (CERC), IRCCS Santa Lucia Foundation (FSL), Via Fosso del Fiorano 43-44, 00143, Rome, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology (IFT)-National Research Council (CNR), Via Fosso del Cavaliere 100, 00133, Rome, Italy.
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy.
| |
Collapse
|
2
|
Matlyuba Jakhonkulovna S, Bahodirova Kamolovna G, Zokirov M, Umida Tajimuratovna B, Yumashev A, Shichiyakh R, Safarova NI, Nargiza Nusratovna A, Esanmuradova N, Muyassar Karimbaevna T, Lazizakhon A, Ishankulov A. Electrochemical biosensors for early detection of Alzheimer's disease. Clin Chim Acta 2025; 572:120278. [PMID: 40185381 DOI: 10.1016/j.cca.2025.120278] [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: 03/15/2025] [Revised: 03/29/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
In recent years, electrochemical biosensors have shown great promise as innovative tools for the early identification of Alzheimer's disease (AD), a neurodegenerative disorder that severely affects cognitive ability and overall quality of life. This comprehensive review aims to consolidate the latest research on the creation and implementation of electrochemical biosensors designed to detect AD-related biomarkers. We examine cutting-edge approaches to surface modification that enhance the attachment of biorecognition molecules, thus enabling the simultaneous identification of multiple biomarkers. This review emphasizes the crucial role that electrochemical biosensors play in the early diagnosis of Alzheimer's disease, highlighting their potential to revolutionize clinical practices by facilitating timely interventions. In the future, research efforts should concentrate on refining these technologies for widespread clinical adoption, ensuring that they meet the needs of both healthcare professionals and patients.
Collapse
Affiliation(s)
| | - Gulnoz Bahodirova Kamolovna
- Department of Scientific Research, Innovations and Scientific and Pedagogical Personnel Training International School of Finance Technology and Science (ISFT Institute), Uzbekistan
| | | | | | - Alexey Yumashev
- Department of Prosthetic Dentistry, Doctor of Medicine, Professor Sechenov First Moscow State Medical University, Russia
| | - Rustem Shichiyakh
- Department of Management, Candidate of Economic Sciences, Associate Professor. Kuban State Agrarian University named after I.T. Trubilin, Krasnodar, Russia
| | - Nasiba I Safarova
- Department of Otorhinolaryngology, Faculty of Postgraduate Education, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | - Nilufar Esanmuradova
- "Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan; Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Tadjibaeva Muyassar Karimbaevna
- Department of Zoology, Human Morphophysiology and Nutrition (PhD), Nukus State Pedagogical Institute Named After Ajiniyaz, Uzbekistan
| | - Alidjanova Lazizakhon
- International Islamic Academy of Uzbekistan, Senior Lecturer of "UNESCO Chair on Religious Studies and the Comparative Study of World Religions", Kadiri st. 11, Tashkent, Uzbekistan
| | - Alisher Ishankulov
- Samarkand State University named after Sharof Rashidov, Uzbekistan; Kimyo International University in Tashkent, Branch Samarkand, Uzbekistan
| |
Collapse
|
3
|
Marongiu R, Platholi J, Park L, Yu F, Sommer G, Woods C, Milner TA, Glass MJ. Promotion of neuroinflammation in select hippocampal regions in a mouse model of perimenopausal Alzheimer's disease. Front Mol Biosci 2025; 12:1597130. [PMID: 40438709 PMCID: PMC12116374 DOI: 10.3389/fmolb.2025.1597130] [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: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Alzheimer's disease, the most common form of dementia, is characterized by age-dependent amyloid beta (Ab) aggregation and accumulation, neuroinflammation, and cognitive deficits. Significantly, there are prominent sex differences in the risk, onset, progression, and severity of AD, as well as response to therapies, with disease burden disproportionately affecting women. Although menopause onset (i.e., perimenopause) may be a critical transition stage for AD susceptibility in women, the role of early ovarian decline in initial disease pathology, particularly key neuroinflammatory processes, is not well understood. Methods To study this, we developed a unique mouse model of perimenopausal AD by combining an accelerated ovarian failure (AOF) model of menopause induced by 4-vinylcyclohexene diepoxide (VCD) with the 5xFAD transgenic AD mouse model. To target early stages of disease progression, 5xFAD females were studied at a young age (∼4 months) and at the beginning stage of ovarian failure analogous to human perimenopause (termed "peri-AOF"), and compared to age-matched males. Assessment of neuropathology was performed by immunohistochemical labeling of Ab as well as markers of astrocyte and microglia activity in the hippocampus, a brain region involved in learning and memory that is deleteriously impacted during AD. Results Our results show that genotype, AOF, and sex contributed to AD-like pathology. Aggregation of Ab was heightened in female 5xFAD mice and further increased at peri-AOF, with hippocampal subregion specificity. Further, select increases in glial activation also paralleled Ab pathology in distinct hippocampal subregions. However, cognitive function was not affected by peri-AOF. Discussion These findings align with the hypothesis that perimenopause constitutes a period of susceptibility for AD pathogenesis in women.
Collapse
Affiliation(s)
- Roberta Marongiu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
- Neurological Surgery Department, Weill Cornell Medicine, New York, NY, United States
- Genetic Medicine Department, Weill Cornell Medicine, New York, NY, United States
| | - Jimcy Platholi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
- Anesthesiology Department, Weill Cornell Medicine, New York, NY, United States
| | - Laibak Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Fangmin Yu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Garrett Sommer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Clara Woods
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Michael J. Glass
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
4
|
Zhu G, Zhang H, Xie R, Younis MR, Fu S, Wang X, Liu B, Li K, Lui S, Wu M. Monitoring Acidification Preceding Aβ Deposition in Alzheimer's Disease. Adv Healthc Mater 2025; 14:e2404907. [PMID: 40103521 DOI: 10.1002/adhm.202404907] [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: 12/06/2024] [Revised: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Amyloid beta (Aβ) is the primary early biomarker of Alzheimer's disease (AD), and since an acidic environment promotes Aβ aggregation, acidification plays a crucial role in AD progression. In this study, a novel acid-responsive near-infrared (NIR) fluorescent probe alongside multiple molecular biology techniques to investigate the temporal relationship between acidification and Aβ deposition, as well as the underlying mechanisms of acidification is employed. By monitoring 2- to 11-month-old APP/PS1 mice and wild-type (WT) mice, it is detected significant fluorescence signal in APP/PS1 mice beginning at 3 months preceding Aβ deposition at 5 months, and peaking at 5 months, followed by cognitive deficits at 8 months. Additionally, elevated monocarboxylate transporter 4 (MCT4) protein expression in 3-month-old APP/PS1 mice indicated disruption of astrocyte-neuron lactate shuttle (ANLS) homeostasis. Overall, this findings first demonstrate that acidification precedes Aβ deposition, peaks at the onset of Aβ deposition, and diminishes thereafter, with early acidification likely driven by the disruption of ANLS.
Collapse
Affiliation(s)
- Guannan Zhu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Hong Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Ruoxi Xie
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California, 90095, USA
| | - Shengxiang Fu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Xiaoze Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Beibei Liu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology of Ministry of Education, College of Chemistry, Sichuan University, No. 29, Wangjiang Road, Chengdu, 610064, China
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Xiang, Chengdu, 610041, China
| |
Collapse
|
5
|
Zhang Z, Li R, Zhou Y, Huang S, Hou Y, Pei G. Dietary Flavonoid Chrysin Functions as a Dual Modulator to Attenuate Amyloid-β and Tau Pathology in the Models of Alzheimer's Disease. Mol Neurobiol 2025; 62:4274-4291. [PMID: 39432184 DOI: 10.1007/s12035-024-04557-y] [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: 03/24/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Growing evidence indicates that healthy diets are associated with a slower progression of Alzheimer's disease (AD). Flavonoids are among the most abundant natural products in diets beneficial to AD, such as the Mediterranean diet. However, the effect and mechanism of these dietary flavonoids on AD remains incompletely understood. Here, we found that a representative dietary natural flavonoid, chrysin (Chr), significantly ameliorated cognitive impairment and AD pathology in APP/PS1 mice. Furthermore, mechanistic studies showed that Chr significantly reduced the levels of amyloid-β (Aβ) and phosphorylated tau (p-tau), along with dual inhibitory activity against β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and glycogen synthase kinase 3β (GSK3β). Moreover, the effect of Chr was further confirmed by EW233, a structural analog of Chr that exhibited an improved pharmacokinetic profile. To further verify the role of Chr and EW233, we utilized our previously established chimeric human cerebral organoid (chCO) model for AD, in which astrogenesis was promoted to mimic the neuron-astrocyte ratio in human brain tissue, and similar dual inhibition of Aβ and p-tau was also observed. Altogether, our study not only reveals the molecular mechanisms through which dietary flavonoids, such as Chr, mitigate AD pathology, but also suggests that identifying a specific constituent that mimics some of the benefits of these healthy diets could serve as a promising approach to discover new treatments for AD.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rongyao Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujun Hou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
6
|
Kour D, Khajuria P, Sharma K, Sharma A, Sharma A, Ali SM, Wazir P, Ramajayan P, Sawant SD, Nandi U, Ahmed Z, Kumar A. Isobavachalcone ameliorates Alzheimer disease pathology by autophagy-mediated clearance of amyloid beta and inhibition of NLRP3 inflammasome in primary astrocytes and 5x-FAD mice. Front Pharmacol 2025; 16:1525364. [PMID: 40183098 PMCID: PMC11965660 DOI: 10.3389/fphar.2025.1525364] [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: 11/09/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background and Aim Alzheimer's disease (AD) progresses with Aβ plaque deposition and neuroinflammation. Given the complexity of AD pathology, single-target therapies have frequently failed in clinical trials. We hypothesized that a multitarget approach could yield better therapeutic outcomes. To this end, we identified isobavachalcone (IBC), a natural compound with dual pharmacological activity in reducing Aβ plaques and neuroinflammation. Experimental Procedure Primary astrocytes were isolated from 3 to 4 days old C57BL/6J mice pups for in-vitro assays, while in-vivo studies were conducted on 5x-FAD mice. Protein alterations were evaluated using ELISA, western blotting, immunocytochemistry, and immunohistochemistry. Behavioral analyses included the radial arm maze, open field, and rotarod tests. Data from all in vitro and in vivo experiments were analyzed by using one-way ANOVA and post-hoc Bonferroni tests. Results In-vitro analyses in astrocytes demonstrated that IBC at 5 and 10 μM concentrations induce AMPK phosphorylation through CAMKK2, promoting autophagy and inhibiting the NLRP3 inflammasome in primary astrocytes. IBC-treated astrocytes exhibited significant clearance of extracellular amyloid beta. Mechanistic studies highlighted autophagy as a key factor in reducing both NLRP3 inflammasome activity and Aβ levels. Two months of treatment of 5x-FAD mice with IBC at 25 and 50 mg/kg significantly improved cognitive functions, as evidenced by enhanced memory and motor performance in behavioral tests. Subsequent brain tissue analysis revealed that IBC upregulated autophagic proteins to reduce the brain's amyloid beta levels, resulting in decreased expression of neuroinflammation markers. Conclusion IBC effectively ameliorates AD pathology through autophagy-mediated clearance of Aβ and suppressing neuroinflammation in 5x-FAD mice.
Collapse
Affiliation(s)
- Dilpreet Kour
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Parul Khajuria
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kuhu Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Alpa Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Ankita Sharma
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Syed Mudassir Ali
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Priya Wazir
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - P. Ramajayan
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanghapal D. Sawant
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Organic Chemsitry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Utpal Nandi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ajay Kumar
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
7
|
Marongiu R, Platholi J, Park L, Yu F, Sommer G, Woods C, Milner TA, Glass MJ. Perimenopause promotes neuroinflammation in select hippocampal regions in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643317. [PMID: 40161644 PMCID: PMC11952527 DOI: 10.1101/2025.03.14.643317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by age-dependent amyloid beta (Aβ) aggregation and accumulation, neuroinflammation, and cognitive deficits. Significantly, there are prominent sex differences in the risk, onset, progression, and severity of AD, as well as response to therapies, with disease burden disproportionally affecting women. Although menopause onset (i.e., perimenopause) may be a critical transition stage for AD susceptibility in women, the role of early ovarian decline in initial disease pathology, particularly key neuroinflammatory processes, is not well understood. To study this, we developed a unique mouse model of perimenopausal AD by combining an accelerated ovarian failure (AOF) model of menopause induced by 4-vinylcyclohexene diepoxide (VCD) with the 5xFAD transgenic AD mouse model. To target early stages of disease progression, 5xFAD females were studied at a young age (∼4 months) and at the beginning stage of ovarian failure analogous to human perimenopause (termed "peri-AOF"), and compared to age-matched males. Assessment of neuropathology was performed by immunohistochemical labeling of Aβ as well as markers of astrocyte and microglia activity in the hippocampus, a brain region involved in learning and memory that is deleteriously impacted during AD. Our results show that genotype, AOF, and sex contributed to AD-like pathology. Aggregation of Aβ was heightened in female 5xFAD mice and further increased at peri-AOF, with hippocampal subregion specificity. Further, select increases in glial activation also paralleled Aβ pathology in distinct hippocampal subregions. However, cognitive function was not affected by peri-AOF. These findings align with the hypothesis that perimenopause constitutes a period of susceptibility for AD pathogenesis in women.
Collapse
|
8
|
Chinnathambi S, Malik S, Chandrashekar M. Tau PET probes for Alzheimer's disease detection and their structural characterization. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 145:255-285. [PMID: 40324849 DOI: 10.1016/bs.apcsb.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
There are two hallmarks for the Alzheimer's disease that are currently used to identify the disease- the presence of the proteins Amyloid-β and Tau. Amyloid PET has been studied for a long time and many effective probes have been introduced, some approved by the FDA, including [18F]-florbetaben (Neuraceq), [18F]-florbetapir (Amyvid), [18F]-flutemetamol (Vizamyl). However, it was found that imaging of NFTs could give more accurate results as the accumulation of Tau could directly be correlated with neurodegeneration, which isn't the case for Amyloid-β. Amyloid PET is thereby a diagnostic tool, which can rather be used for confirming the absence of Alzheimer's Disease. Tau PET, which was found to be a potentially useful diagnostic tool was explored further as it can directly be associated with the extent of spread of the disease. This led to the discovery of many probes for Tau PET. The initial ones were non-selective for Tau over Aβ. Further exploration suggested two generations of Tau probes, both with higher selectivity for Tau over Aβ. A second generation was introduced to overcome the shortcomings of the first generation which are examined in this review. Much research on effective Tau PET probes has led to an FDA-approved Tau probe, 18F-flortaucipir. This systematic review discusses the characteristics and effectiveness of the first-generation probes, second-generation probes and other newer probes. It discusses the structural changes made in the probes over time that led to the enhancement of their properties as a Tau probe, that is, increased affinity and selectivity for Tau. It also discusses the shortcomings of probes developed so far and the ideal characteristics for Tau probes.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Hosur Road, Bangalore, Karnataka, India.
| | - Sneha Malik
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Hosur Road, Bangalore, Karnataka, India
| | - Madhura Chandrashekar
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences Hospital (NIMHANS), Institute of National Importance, Hosur Road, Bangalore, Karnataka, India
| |
Collapse
|
9
|
Panza F, Dibello V, Sardone R, Zupo R, Castellana F, Leccisotti I, Moretti MC, Altamura M, Bellomo A, Daniele A, Solfrizzi V, Resta E, Lozupone M. Successes and failures: the latest advances in the clinical development of amyloid-β-targeting monoclonal antibodies for treating Alzheimer's disease. Expert Opin Biol Ther 2025; 25:275-283. [PMID: 39908579 DOI: 10.1080/14712598.2025.2463963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION The amyloid cascade hypothesis postulated that the accumulation of amyloid-β (Aβ) was the first step of the Alzheimer's disease (AD) pathological process. Effective reduction of Aβ plaque load by numerous drug candidates, among which anti-Aβ monoclonal antibodies, has produced discussible clinical successes and several failures. It was questioned whether Aβ may be the principal AD pathogenic factor and a valid therapeutic target and if targeting Aβ different species could make the difference. AREAS COVERED This review article summarized successes and failures of anti-Aβ monoclonal antibody therapy for AD, delineating the latest advances for their clinical development also according to their target engagement and downstream biomarkers. EXPERT OPINION The preliminary success of the recent Phase III randomized clinical trials (RCTs) of lecanemab, donanemab, and remternetug, and lessons learned from the failure of previous anti-Aβ monoclonal antibodies RCTs, provided critical evidence to support the role of Aβ in AD pathogenesis. The loss of free Aβ instead of an Aβ toxicity may promote AD neuropathology. Cerebrospinal fluid analyses (i.e. increases in Aβ1-42) may indicate a potential benefit of anti-Aβ monoclonal antibodies in AD and downstream biomarkers should be considered for providing comprehension in cognitive and clinical efficacy of future AD RCTs.
Collapse
Affiliation(s)
- Francesco Panza
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Vittorio Dibello
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Health Authority of Taranto, Taranto, Italy
| | - Roberta Zupo
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| | - Fabio Castellana
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| | - Ivana Leccisotti
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Claudia Moretti
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro", Bari, Italy
| | - Emanuela Resta
- Translational Medicine and Health System Management, Department of Economy, University of Foggia, Foggia, Italy
| | | |
Collapse
|
10
|
Yao C, Shan Y, Cui B, Chen Z, Bi S, Wang T, Yan S, Lu J. Hyperconnectivity and Connectome Gradient Dysfunction of Cerebello-Thalamo-Cortical Circuitry in Alzheimer's Disease Spectrum Disorders. CEREBELLUM (LONDON, ENGLAND) 2025; 24:43. [PMID: 39913059 DOI: 10.1007/s12311-025-01792-4] [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] [Accepted: 01/17/2025] [Indexed: 02/07/2025]
Abstract
Cerebellar functional connectivity changes have been reported in Alzheimer's disease (AD), but a comprehensive framework integrating these findings is lacking. This retrospective study investigates the cerebello-thalamo-cortical (CTC) circuit in AD, using functional gradient analysis to elucidate deficits and potential biomarkers. We analyzed data from 246 participants enrolled in the Alzheimer's Disease Neuroimaging Initiative (ADNI-3; NCT02854033), including 58 with AD, 103 with mild cognitive impairment (MCI), and 85 cognitively normal (CN) controls, matched for age and sex. All individuals underwent comprehensive neuropsychological assessments (MMSE, MoCA, ADAS-Cog) and MRI scans. We extracted mean time series for 270 brain regions (an extended Power atlas) and computed pairwise functional connectivity, focusing on CTC circuitry. Thalamic and cerebellar connectivity gradients were derived using voxel-wise correlation matrices and the BrainSpace toolbox, defining thalamic and cerebellar masks from the Melbourne subcortical atlas and AAL atlas, respectively. ANCOVA with post hoc analyses, controlling for age and sex, was conducted to assess abnormal CTC connectivity across AD, MCI, and CN groups. LASSO regression identified edges within the CTC circuitry that significantly differed between AD and CN, MCI and CN, AD and MCI, as well as was used to construct Logistic classification model. Pearson correlations were performed to examine relationships between mean CTC connectivity, individual edges, and cognitive scores (MMSE, MoCA, ADAS-Cog). To explore the hierarchical organization of the thalamus and cerebellum, global gradient distributions were compared across groups using two-sample Kolmogorov-Smirnov tests. Additionally, ANCOVA was applied to compare subfield- and functional-level gradients of the thalamus and cerebellum among AD, MCI, and CN. False discovery rate (FDR) corrections were used, setting the statistical significance threshold was set at P < 0.05. AD and MCI individuals exhibited increased CTC connectivity compared to CN (all P < 0.05). Average CTC connectivity did not correlate with cognitive scores (P > 0.05), but specific CTC edges were correlated. LASSO regression identified 20 discriminative edges, achieving high accuracy in AD-CN classification (AUC = 0.92 training, AUC = 0.80 test). Thalamic and cerebellar gradient distributions differed significantly across groups (all P < 0.05), with specific regions showing distinct gradient scores. Five cerebellar functional networks exhibited decreased gradient scores. Significant CTC hyperconnectivity in AD and MCI compared with CN suggests early thalamic and cerebellar dysregulation. Classification analyses effectively distinguished AD vs. CN but were moderate for MCI vs. CN and limited for MCI vs. AD. Gradient analyses revealed global- and subfield-level disruptions in AD, emphasizing the role of thalamic and cerebellar interactions in cognitive decline and offering potential diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Chenyang Yao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Bixiao Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhigeng Chen
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Sheng Bi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Tao Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
| |
Collapse
|
11
|
Bao L, Li X, Tian J, Wang L, Ji Y, Cui Y, Sun W, Zhang J, Xia M, Zhu P, Cui G, Chen H. GGC repeat expansions in NOTCH2NLC cause uN2CpolyG cerebral amyloid angiopathy. Brain 2025; 148:467-479. [PMID: 39167540 DOI: 10.1093/brain/awae274] [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: 04/15/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The expansion of GGC repeats within NOTCH2NLC leads to translation of the uN2CpolyG protein, the primary pathogenic factor in neuronal intranuclear inclusion disease (NIID). The aim of this study was to explore the deposition of uN2CpolyG as an amyloid in the vessel wall, leading to uN2CpolyG cerebral amyloid angiopathy-related cerebral microbleeds (CMBs). A total of 97 patients with genetically confirmed NIID were enrolled in this study. We analysed the presence of CMBs using susceptibility-weighted imaging sequences and compared general clinical information, cerebrovascular risk factors, stroke history, antiplatelet medication use and MRI features between NIID patients with and without CMBs. We also performed haematoxylin and eosin, Perl's, Congo red and Thioflavin S staining, ubiquitin, p62 and uN2CpolyG immunostaining on brain tissue obtained from four NIID patients. A total of 354 CMBs were detected among 41 patients with NIID, with nearly half located in the deep brain, one-third in the lobes and ∼20% in the infratentorial area. No significant differences in cerebrovascular disease risk factors or history of antiplatelet drug use were observed between patients with and without CMBs. However, patients with CMBs had suffered a higher incidence of previous ischaemic and haemorrhagic stroke events. This group also had a higher incidence of recent subcortical infarcts and a higher proportion of white matter lesions in the external capsule and temporal pole. Conversely, patients without CMBs showed higher detection of high signals at the corticomedullary junction on diffusion-weighted imaging and more pronounced brain atrophy. Haematoxylin and eosin staining showed blood vessel leakage and haemosiderin-laden macrophage clusters, and Prussian blue staining revealed iron deposition in brain tissue. CMBs occurred more frequently in small vessels lacking intranuclear inclusions, and extensive degeneration of endothelial cells and smooth muscle fibres was observed mainly in vessels lacking inclusions. Congo red-positive amyloid deposition was observed in the cerebral vessels of NIID patients, with disordered filamentous fibres appearing under an electron microscope. Additionally, the co-localization of Thioflavin S-labelled amyloid and uN2CpolyG protein in the cerebral vascular walls of NIID patients further suggested that uN2CpolyG is the main pathogenic protein in this form of amyloid angiopathy. In conclusion, we reviewed patients with GGC repeat expansion of NOTCH2NLC from a new perspective, providing initial clinical, neuroimaging and pathological evidence suggesting that uN2CpolyG might contribute to a distinct type of cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Lei Bao
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiaowen Li
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jin Tian
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Lulu Wang
- Department of Pathology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Ying Ji
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yingying Cui
- Department of Pathology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Wen Sun
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Jing Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100000, China
| | - Man Xia
- Department of Neurological Intensive Care, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Pinyi Zhu
- Department of Geriatric Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing 210000, China
| | - Guiyun Cui
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hao Chen
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
12
|
Marvi F, Chen YH, Sawan M. Alzheimer's Disease Diagnosis in the Preclinical Stage: Normal Aging or Dementia. IEEE Rev Biomed Eng 2025; 18:74-92. [PMID: 38478432 DOI: 10.1109/rbme.2024.3376835] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) progressively impairs the memory and thinking skills of patients, resulting in a significant global economic and social burden each year. However, diagnosis of this neurodegenerative disorder can be challenging, particularly in the early stages of developing cognitive decline. Current clinical techniques are expensive, laborious, and invasive, which hinders comprehensive studies on Alzheimer's biomarkers and the development of efficient devices for Point-of-Care testing (POCT) applications. To address these limitations, researchers have been investigating various biosensing techniques. Unfortunately, these methods have not been commercialized due to several drawbacks, such as low efficiency, reproducibility, and the lack of accurate identification of AD markers. In this review, we present diverse promising hallmarks of Alzheimer's disease identified in various biofluids and body behaviors. Additionally, we thoroughly discuss different biosensing mechanisms and the associated challenges in disease diagnosis. In each context, we highlight the potential of realizing new biosensors to study various features of the disease, facilitating its early diagnosis in POCT. This comprehensive study, focusing on recent efforts for different aspects of the disease and representing promising opportunities, aims to conduct the future trend toward developing a new generation of compact multipurpose devices that can address the challenges in the early detection of AD.
Collapse
|
13
|
Kolobova EA, Petrushanko IY, Mitkevich VA, Makarov AA, Grigorova IL. Novel method for detection of Aβ and Iso-D7-Aβ N-terminus-specific B cells and Iso-D7-Aβ-specific antibodies. Biol Methods Protoc 2025; 10:bpaf001. [PMID: 39830230 PMCID: PMC11739456 DOI: 10.1093/biomethods/bpaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025] Open
Abstract
Alzheimer's disease (AD) is a multifactorial systemic disease that is triggered, at least in part, by the accumulation of β-amyloid (Aβ) peptides in the brain, but it also depends on immune system-mediated regulation. Recent studies suggest that B cells may play a role in AD development and point to the accumulation of clonally expanded B cells in AD patients. However, the specificity of the clonally expanded B cells is unknown, and the contribution of Aβ-specific B cells to AD pathology development is unclear. In this study, we have developed a novel method to identify Aβ-specific B cells by flow cytometry using fluorescent tetramers. The suggested method also enables the identification of B-cell clones specific to a more pathology-provoking form of Aβ with an isomerized Asp7 residue (Iso-D7-Aβ) that accumulates in elderly people and in AD patients. The method has been verified using mice immunized with antigens containing the isomerized or non-isomerized Aβ N-terminus peptides. In addition, we describe a new method for the detection of Iso-D7-Aβ-specific antibodies, which was tested on mouse serum. These methods are of potential importance in research aimed at studying AD and may be also utilized for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Elizaveta A Kolobova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
- Institute of Translational Medicine, Department of Molecular Technologies, Pirogov Russian National Research Medical University, 117513, Moscow, Russia
| | - Irina Yu Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - Irina L Grigorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
- Institute of Translational Medicine, Department of Molecular Technologies, Pirogov Russian National Research Medical University, 117513, Moscow, Russia
| |
Collapse
|
14
|
Santillán F, Charron CL, Galarreta BC, Luyt LG. Tailored peptide nanomaterials for receptor targeted prostate cancer imaging. NANOSCALE 2024; 16:22001-22010. [PMID: 39514187 DOI: 10.1039/d4nr03273j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
We report the development of a peptide-based optical nanoprobe specifically tailored for prostate cancer imaging. The imaging probe is comprised of cyclic peptide nanotubes, formed via the aqueous co-assembly of four cyclic D,L-alternating octapeptides. The inherent properties of these cyclic building blocks have been carefully selected to enhance their efficacy in imaging applications, through the addition of a cancer targeting peptide and a fluorescent dye. Comprehensive characterization using scanning electron microscopy (FESEM) and low-voltage transmission electron microscopy (LV-TEM) confirms the formation of nanotubes through co-assembly of the cyclic peptides. The resulting nanotubes show an average diameter of 28 nm. Circular dichroism (CD) spectroscopy validates the formation of stable beta-sheet hydrogen bonding structures at both 20 and 37 °C, ensuring their suitability for biomedical applications. Evaluation of PSMA-binding specificity of the resulting peptide nanotubes is assessed using confocal fluorescence microscopy demonstrating receptor-mediated uptake in prostate cancer cells. We anticipate this strategy will provide the basis for the utilization of co-assembled systems for advancing molecular imaging techniques in prostate cancer and other cancers.
Collapse
Affiliation(s)
- Fátima Santillán
- Department of Chemistry, University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5B7, Canada
| | - Carlie L Charron
- Department of Chemistry, Dalhousie University, 6274 Coburg Road, Halifax, Nova Scotia B3H 4R2, Canada
| | - Betty C Galarreta
- Departamento Académico de Ciencias - Sección Química, Pontificia Universidad Católica del Perú, Av. Universitaria 1801, San Miguel, Lima 15088, Perú
| | - Leonard G Luyt
- Departments of Chemistry and Departments of Medical Imaging and Oncology, University of Western Ontario, University of Western Ontario, Centre, 1151 Richmond St, London, ON, N6A 3K7, Canada
- Verspeeten Family Cancer Centre, London Health Sciences, 800 Commissioners Rd. E., London, ON, N6A 5W9, Canada.
| |
Collapse
|
15
|
Dinsmore TC, Liu J, Miao J, Ünsal Ö, Sürmeli D, Beinborn M, Lin YS, Kumar K. Potent and Protease Resistant Azapeptide Agonists of the GLP-1 and GIP Receptors. Angew Chem Int Ed Engl 2024; 63:e202410237. [PMID: 39151024 DOI: 10.1002/anie.202410237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/17/2024] [Accepted: 08/13/2024] [Indexed: 08/18/2024]
Abstract
The gut-derived peptide hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) play important physiological roles including glucose homeostasis and appetite suppression. Stabilized agonists of the GLP-1 receptor (GLP-1R) and dual agonists of GLP-1R and GIP receptor (GIPR) for the management of type 2 diabetes and obesity have generated widespread enthusiasm and have become blockbuster drugs. These therapeutics are refractory to the action of dipeptidyl peptidase-4 (DPP4), that catalyzes rapid removal of the two N-terminal residues of the native peptides, in turn severely diminishing their activity profiles. Here we report that a single atom change from carbon to nitrogen in the backbone of the entire peptide makes them refractory to DPP4 action while still retaining full potency and efficacy at their respective receptors. This was accomplished by use of aza-amino acids, that are bioisosteric replacements for α-amino acids that perturb the structural backbone and local side chain conformations. Molecular dynamics simulations reveal that aza-amino acid can populate the same conformational space that GLP-1 adopts when bound to the GLP-1R. The insertion of an aza-amino acid at the second position from the N-terminus in semaglutide and in a dual agonist of GLP-1R and GIPR further demonstrates its capability as a viable alternative to current DPP4 resistance strategies while offering additional structural variation that may influence downstream signaling.
Collapse
Affiliation(s)
- Tristan C Dinsmore
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Jamie Liu
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Jiayuan Miao
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Özge Ünsal
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Damla Sürmeli
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Martin Beinborn
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
- Molecular Pharmacology Research Center, Tufts Medical Center, 02111, Boston, Massachusetts, USA
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Krishna Kumar
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, 02155, Medford, Massachusetts, USA
- Cancer Center, Tufts Medical Center, 02111, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Guo Q, Wang T, Qian C, Wang X. Redox Oxygen Species-Responsive Nanotheranostics with Dual-Channel Fluorescent Turn-On for Early Diagnosis and Targeted Therapy of Alzheimer's Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403980. [PMID: 39428844 DOI: 10.1002/smll.202403980] [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: 05/16/2024] [Revised: 09/24/2024] [Indexed: 10/22/2024]
Abstract
Current diagnosis and treatment strategies mainly focus on the pathologies of the mid-to-late stage of AD (Alzheimer's disease), with clinical outcomes that are far from ideal. Herein, we developed the ROS (reactive oxygen species)-responsive brain neuronal targeting nanotheranostic platforms that possess the dual-channel fluorescent "turn-on" properties and release drugs in AD neurons in response to ROS, thereby simultaneously facilitating the diagnosis and therapy of early AD. Through the modification of acetylcholine receptor targeting RVG29 peptide, the nanotheranostics penetrated BBB and accumulated into diseased neurons in an intact form, consequently maximizing the diagnostic and therapeutic performance. The anti-oxidative drug baicalein conjugated onto the surface of nanotheranostics via ROS-cleavable boronate ester linkage rapidly released for ROS scavenging, while the encapsulated fluorophores turned on their fluorescence for AD diagnosis upon microenvironment stimuli. This nanotheranostic strategy exhibited highly sensitivity with a ROS detection limit of up to 100 µm and accurately early detection of ROS in 3×Tg AD mice at 6 months of age in vivo. In addition, it could also rescue memory defects, scavenge oxidative stress, attenuate neuroinflammation and enhance neuroprotective effect in 3×Tg AD mice. This work opens up a promising and smart strategy for early diagnosis and therapy in neurodegenerative disease.
Collapse
Affiliation(s)
- Qian Guo
- Shanghai 411 Hospital, China RongTong Medical Healthcare Group Co.Ltd. / 411 Hospital, Shanghai University, Shanghai, 200081, China
- Laboratory of Drug Delivery, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Tianying Wang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Christopher Qian
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, NT, Shatin, Hong Kong
| | - Xinyu Wang
- Laboratory of Drug Delivery, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| |
Collapse
|
17
|
Wu X, Shirani H, Vidal R, Ghetti B, Ingelsson M, Klingstedt T, Nilsson KPR. Distinct Chemical Determinants are Essential for Achieving Ligands for Superior Optical Detection of Specific Amyloid-β Deposits in Alzheimer's Disease. ChemistryOpen 2024; 13:e202400186. [PMID: 39508558 PMCID: PMC11625938 DOI: 10.1002/open.202400186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Aggregated forms of different proteins are common hallmarks for several neurodegenerative diseases, including Alzheimer's disease, and ligands that selectively detect specific protein aggregates are vital. Herein, we investigate the molecular requirements of thiophene-vinyl-benzothiazole based ligands to detect a specific type of Aβ deposits found in individuals with dominantly inherited Alzheimer's disease caused by the Arctic APP E693G mutation. The staining of these Aβ deposits was alternated when switching the terminal heterocyclic moiety attached to the thiophene-vinyl-benzothiazole scaffold. The most prevalent staining was observed for ligands having a terminal 3-methyl-1H-indazole moiety or a terminal 1,2-dimethoxybenzene moiety, verifying that specific molecular interactions between these ligands and the aggregates were necessary. The synthesis of additional thiophene-vinyl-benzothiazole ligands aided in pinpointing additional crucial chemical determinants, such as positioning of nitrogen atoms and methyl substituents, for achieving optimal staining of Aβ aggregates. When combining the optimized thiophene-vinyl-benzothiazole based ligands with a conventional ligand, CN-PiB, distinct staining patterns were observed for sporadic Alzheimer's disease versus dominantly inherited Alzheimer's disease caused by the Arctic APP E693G mutation. Our findings provide chemical insights for developing novel ligands that allow for a more precise assignment of Aβ deposits, and might also aid in creating novel agents for clinical imaging of distinct Aβ aggregates in AD.
Collapse
Affiliation(s)
- Xiongyu Wu
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - Hamid Shirani
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - Ruben Vidal
- Department of Pathology and Laboratory MedicineIndiana University School of Medicine46202Indianapolis, IndianaUSA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory MedicineIndiana University School of Medicine46202Indianapolis, IndianaUSA
| | - Martin Ingelsson
- Krembil Brain InstituteUniversity Health NetworkM5T 1 M8Toronto, OntarioCanada
- Tanz Centre for Research in Neurodegenerative DiseasesDepartments of Medicine and Laboratory Medicine & PathobiologyUniversity of TorontoM5T 0S8Toronto, OntarioCanada
- Molecular GeriatricsDepartment of Public Health and Caring SciencesUppsala UniversitySE-751 85UppsalaSweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - K. Peter R. Nilsson
- Department of Physics, Chemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| |
Collapse
|
18
|
Tong J, Li C, Zhao J, Wang K, Zhao Z, Liu Y, Qing T, Liu X. Poly-Adenine Assisted Signaling Displaced Probe Ratiometric Electrochemical Aptasensor for Accurate Detection of Alzheimer's Disease Aβ Biomarkers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64297-64306. [PMID: 39514840 DOI: 10.1021/acsami.4c14877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Alzheimer's disease (AD) is a widely prevalent neurodegenerative condition globally, arousing significant interest in the noninvasive early detection of the disease. The concentration of amyloid β (Aβ) biomarkers in the blood is closely linked to the progression of AD, emphasizing the importance of developing a precise method for detecting these biomarkers in blood samples for early diagnosis. In this study, we developed a ratiometric electrochemical aptamer-based (EAB) biosensor for accurate detection of Aβ42 and Aβ40. The ratiometric biosensor utilizes a gold nanoparticle (AuNPs) modified electrode through an electrochemical deposition assay and a self-assembled poly-A based U-shaped structure to achieve signal changes through competitive binding of target molecules. Importantly, using the calibration of the clipping reference probe, the proposed biosensor displayed accurate detection ability for Aβ42 and Aβ40, with detection limits of 59 and 21 pM, respectively. It also has great stability and anti-interference ability in complex biological samples. Furthermore, it demonstrated satisfactory performance in detecting the Aβ peptides in a 20% FBS sample, with recoveries ranging from 96.7% to 102.2% in serum samples, and relative standard deviations (RSD) ranging from 3.3% to 8.9%. Moreover, the proposed method is expected to directly quantify the Aβ42/Aβ40 ratio by introducing a suitable reference probe, providing a potential and effective tool for the early diagnosis of AD.
Collapse
Affiliation(s)
- Jiajun Tong
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Chengxiang Li
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Jing Zhao
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Kemin Wang
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Zijun Zhao
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Yiwei Liu
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Taiping Qing
- College of Environment and Resources, Hunan Provincial University Key Laboratory for Environmental Behavior and Control Principle of New Pollutants, Xiangtan University, Xiangtan 411105, Hunan, China
| | - Xiaofeng Liu
- Hunan Institute of Advanced Sensing and Information Technology, Hunan Provincial Key Laboratory of Smart Carbon Materials and Advanced Sensing, Xiangtan University, Xiangtan 411105, Hunan, China
| |
Collapse
|
19
|
Mir A, Zhu A, Lau R, Barr N, Sheikh Z, Acuna D, Dayal A, Hibino N. Applications, Limitations, and Considerations of Clinical Trials in a Dish. Bioengineering (Basel) 2024; 11:1096. [PMID: 39593756 PMCID: PMC11591410 DOI: 10.3390/bioengineering11111096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Recent advancements in biotechnology forged the path for clinical trials in dish (CTiDs) to advance as a popular method of experimentation in biomedicine. CTiDs play a fundamental role in translational research through technologies such as induced pluripotent stem cells, whole genome sequencing, and organs-on-a-chip. In this review, we explore advancements that enable these CTiD biotechnologies and their applications in animal testing, disease modeling, and space radiation technologies. Furthermore, this review dissects the advantages and disadvantages of CTiDs, as well as their regulatory considerations. Lastly, we evaluate the challenges that CTiDs pose and the role of CTiDs in future experimentation.
Collapse
Affiliation(s)
- Amatullah Mir
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Angie Zhu
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Rico Lau
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Nicolás Barr
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Zyva Sheikh
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Diana Acuna
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Anuhya Dayal
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
| | - Narutoshi Hibino
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA; (A.M.); (A.Z.); (R.L.); (N.B.); (Z.S.); (D.A.); (A.D.)
- Pediatric Cardiac Surgery, Advocate Children’s Hospital, 4440 W 95th St., Oak Lawn, IL 60453, USA
| |
Collapse
|
20
|
Wat JH, Pizzala NJ, Reppert M. Isotope Reverse-Labeled Infrared Spectroscopy as a Probe of In-Cell Protein Structure. J Phys Chem B 2024; 128:9923-9934. [PMID: 39358675 DOI: 10.1021/acs.jpcb.4c03068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
While recent years have seen great progress in determining the three-dimensional structure of isolated proteins, monitoring protein structure inside live cells remains extremely difficult. Here, we examine the utility of Fourier transform infrared (FTIR) spectroscopy as a probe of protein structure in live bacterial cells. Selective isotope enrichment is used both to distinguish recombinantly expressed NuG2b protein from the cellular background and to examine the conformation of specific residues in the protein. To maximize labeling flexibility and to improve spectral resolution between label and main-band peaks, we carry out isotope-labeling experiments in "reverse-labeling" mode: cells are initially grown in 13C-enriched media, with specific 12C-labeled amino acids added when protein expression is induced. 1 Because FTIR measurements require only around 20 μL of sample and each measurement takes only a few minutes to complete, isotope-labeling costs are minimal, allowing us to label multiple different residues in parallel in simultaneously grown cultures. For the stable NuG2b protein, isotope difference spectra from live bacterial cultures are nearly identical to spectra from isolated proteins, confirming that the structure of the protein is unperturbed by the cellular environment. By combining such measurements with site-directed mutagenesis, we further demonstrate that the local conformation of individual amino acids can be monitored, allowing us to determine, for example, whether a specific site in the protein contributes to α-helix or β-sheet structures.
Collapse
Affiliation(s)
- Jacob H Wat
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907-2084, United States
| | - Nicolas J Pizzala
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907-2084, United States
| | - Mike Reppert
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907-2084, United States
| |
Collapse
|
21
|
Lin C, Du X, Wang X. A perspective on Alzheimer's disease: exploring the potential of terminal/paradoxical lucidity and psychedelics. Mol Neurodegener 2024; 19:72. [PMID: 39396013 PMCID: PMC11475160 DOI: 10.1186/s13024-024-00761-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024] Open
Affiliation(s)
- Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xiubo Du
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.
| |
Collapse
|
22
|
Zhou Y, Zhu J, Gao F, Hu M, Qian C, Wang X, Wang X. Fighting fire with fire: remodeling Aβ aggregation with H-aggregates of a europium(III) complex. Dalton Trans 2024; 53:14966-14970. [PMID: 39189405 DOI: 10.1039/d4dt02188f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
We herein report a "Fight Aggregation with Aggregation" (FAA) approach for redirection of amyloid-β peptide (Aβ) aggregation using a europium(III) complex (EuL3) that can undergo H-aggregation in aqueous solution under physiological conditions. The H-aggregates of EuL3 may serve as scaffolds that can facilitate the accumulation of Aβ to form non-fibrillar co-assemblies. As a result, the Aβ aggregation-induced cytotoxicity was inhibited.
Collapse
Affiliation(s)
- Yuancun Zhou
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China.
| | - Jiacheng Zhu
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China.
| | - Furong Gao
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China.
| | - Ming Hu
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China.
| | - Chengyuan Qian
- School of Life Sciences, Nanjing University, Nanjing 210093, P. R. China
| | - Xin Wang
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China.
| | - Xiaohui Wang
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China.
| |
Collapse
|
23
|
Li Y, Awasthi S, Bryan L, Ehrlich RS, Tonali N, Balog S, Yang J, Sewald N, Mayer M. Fluorescence-Based Monitoring of Early-Stage Aggregation of Amyloid-β, Amylin Peptide, Tau, and α-Synuclein Proteins. ACS Chem Neurosci 2024; 15:3113-3123. [PMID: 39150403 PMCID: PMC11378287 DOI: 10.1021/acschemneuro.4c00097] [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: 02/12/2024] [Revised: 07/02/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024] Open
Abstract
Early-stage aggregates of amyloid-forming proteins, specifically soluble oligomers, are implicated in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Protein aggregation is typically monitored by fluorescence using the amyloid-binding fluorophore thioflavin T (ThT). Thioflavin T interacts, however, preferentially with fibrillar amyloid structures rather than with soluble, early-stage aggregates. In contrast, the two fluorophores, aminonaphthalene 2-cyanoacrylate-spiropyran (AN-SP) and triazole-containing boron-dipyrromethene (taBODIPY), were reported to bind preferentially to early-stage aggregates of amyloidogenic proteins. The present study compares ThT with AN-SP and taBODIPY with regard to their ability to monitor early stages of aggregation of four different amyloid-forming proteins, including amyloid-β (Aβ), tau protein, amylin, and α-synuclein. The results show that the three fluorophores vary in their suitability to monitor the early aggregation of different amyloid-forming proteins. For instance, in the presence of Aβ and amylin, the fluorescence intensity of AN-SP increased at an earlier stage of aggregation than the fluorescence of ThT, albeit with only a small fluorescence increase in the case of AN-SP. In contrast, in the presence of tau and amylin, the fluorescence intensity of taBODIPY increased at an earlier stage of aggregation than the fluorescence of ThT. Finally, α-synuclein aggregation could only be monitored by ThT fluorescence; neither AN-SP nor taBODIPY showed a significant increase in fluorescence over the course of aggregation of α-synuclein. These results demonstrate the ability of AN-SP and taBODIPY to monitor the formation of early-stage aggregates from specific amyloid-forming proteins at an early stage of aggregation, although moderate increases in fluorescence intensity, relatively large uncertainties in fluorescence values, and limited solubility of both fluorophores limit their usefulness for some amyloid proteins. The capability to monitor early aggregation of some amyloid proteins, such as amylin, might accelerate the discovery of aggregation inhibitors to minimize the formation of toxic oligomeric species for potential therapeutic use.
Collapse
Affiliation(s)
- Yuanjie Li
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Saurabh Awasthi
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
- Department
of Biotechnology, National Institute of
Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, Uttar Pradesh 226002, India
| | - Louise Bryan
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Rachel S. Ehrlich
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093-0358, United States
| | - Nicolo Tonali
- CNRS,
BioCIS, Bâtiment Henri Moissan, Université
Paris-Saclay, 17 Av. des Sciences, Orsay 91400, France
| | - Sandor Balog
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Jerry Yang
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California 92093-0358, United States
| | - Norbert Sewald
- Bielefeld
University, Department of Chemistry P.O. Box 100131, Bielefeld 33501, Germany
| | - Michael Mayer
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| |
Collapse
|
24
|
Senapati S, Tripathi K, Awad K, Rahimipour S. Multifunctional Liposomes Targeting Amyloid-β Oligomers for Early Diagnosis and Therapy of Alzheimer's Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311670. [PMID: 38461531 DOI: 10.1002/smll.202311670] [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: 12/14/2023] [Revised: 02/14/2024] [Indexed: 03/12/2024]
Abstract
Early detection and treatment are crucial for Alzheimer's disease (AD) management. Current diagnostic and therapeutic methods focus on late-stage amyloid fibrils and plaques, overlooking toxic soluble amyloid β oligomers (AβOs) accumulating early in AD. A multifunctional liposome-based platform is designed for early diagnosis and therapy of AD, leveraging a novel self-assembled cyclic d,l-α-peptide (CP-2) that selectively targets AβOs. Biocompatible CP-2 conjugated liposomes (CP-2-LPs) effectively disrupt Aβ aggregation and mitigate Aβ-mediated toxicity in human neuroblastoma cells. In transgenic Caenorhabditis elegans AD models, CP-2-LPs significantly outperformed free CP-2 by improving cognitive and behavioral functions, extending lifespan, and reducing toxic AβO levels. Intravenous injection of fluorescently labeled CP-2-LPs reveals effective blood-brain barrier penetration, with significantly higher brain fluorescence in transgenic mice than WT, enabling precise diagnosis. These findings underscore CP-2-LPs as a valuable tool for early detection and targeted therapy in AD.
Collapse
Affiliation(s)
- Sudipta Senapati
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Kuldeep Tripathi
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Khadeja Awad
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shai Rahimipour
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| |
Collapse
|
25
|
Yang Y, Jia X, Yang X, Wang J, Fang Y, Ying X, Zhang M, Wei J, Pan Y. Targeting VDAC: A potential therapeutic approach for mitochondrial dysfunction in Alzheimer's disease. Brain Res 2024; 1835:148920. [PMID: 38599511 DOI: 10.1016/j.brainres.2024.148920] [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: 12/15/2023] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Mitochondrial dysfunction has been implicated in the pathogenesis of Alzheimer's disease, a neurodegenerative disorder characterized by progressive cognitive decline. Voltage-dependent anion channel (VDAC), a protein located in the outer mitochondrial membrane, plays a critical role in regulating mitochondrial function and cellular energy metabolism. Recent studies have identified VDAC as a potential therapeutic target for Alzheimer's disease. This article aims to provide an overview of the role of VDAC in mitochondrial dysfunction, its association with Alzheimer's disease, and the potential of targeting VDAC for developing novel therapeutic interventions. Understanding the involvement of VDAC in Alzheimer's disease may pave the way for the development of effective treatments that can restore mitochondrial function and halt disease progression.
Collapse
Affiliation(s)
- Yaqian Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiaotao Jia
- Department of Neurology, The Affifiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, China
| | - Xinmao Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Jie Wang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yan Fang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiaoping Ying
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Meiqian Zhang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Jing Wei
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yanfang Pan
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| |
Collapse
|
26
|
Haynes JR, Whitmore CA, Behof WJ, Landman CA, Ong HH, Feld AP, Suero IC, Greer CB, Gore JC, Wijesinghe P, Matsubara JA, Wadzinski BE, Spiller BW, Pham W. Targeting soluble amyloid-beta oligomers with a novel nanobody. Sci Rep 2024; 14:16086. [PMID: 38992064 PMCID: PMC11239946 DOI: 10.1038/s41598-024-66970-6] [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: 02/09/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
The classical amyloid cascade hypothesis postulates that the aggregation of amyloid plaques and the accumulation of intracellular hyperphosphorylated Tau tangles, together, lead to profound neuronal death. However, emerging research has demonstrated that soluble amyloid-β oligomers (SAβOs) accumulate early, prior to amyloid plaque formation. SAβOs induce memory impairment and disrupt cognitive function independent of amyloid-β plaques, and even in the absence of plaque formation. This work describes the development and characterization of a novel anti-SAβO (E3) nanobody generated from an alpaca immunized with SAβO. In-vitro assays and in-vivo studies using 5XFAD mice indicate that the fluorescein (FAM)-labeled E3 nanobody recognizes both SAβOs and amyloid-β plaques. The E3 nanobody traverses across the blood-brain barrier and binds to amyloid species in the brain of 5XFAD mice. Imaging of mouse brains reveals that SAβO and amyloid-β plaques are not only different in size, shape, and morphology, but also have a distinct spatial distribution in the brain. SAβOs are associated with neurons, while amyloid plaques reside in the extracellular matrix. The results of this study demonstrate that the SAβO nanobody can serve as a diagnostic agent with potential theragnostic applications in Alzheimer's disease.
Collapse
Affiliation(s)
- Justin R Haynes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Clayton A Whitmore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - William J Behof
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Charlotte A Landman
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Henry H Ong
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrew P Feld
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Isabelle C Suero
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Celeste B Greer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, V5Z3N9, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, V5Z3N9, Canada
| | - Brian E Wadzinski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
| | - Benjamin W Spiller
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Center for Structural Biology, Vanderbilt University, Nashville, TN, 37235, USA.
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, 37212, USA.
| |
Collapse
|
27
|
Suri K, Ramesh M, Bhandari M, Gupta V, Kumar V, Govindaraju T, Murugan NA. Role of Amyloidogenic and Non-Amyloidogenic Protein Spaces in Neurodegenerative Diseases and their Mitigation Using Theranostic Agents. Chembiochem 2024; 25:e202400224. [PMID: 38668376 DOI: 10.1002/cbic.202400224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Indexed: 06/15/2024]
Abstract
Neurodegenerative diseases (NDDs) refer to a complex heterogeneous group of diseases which are associated with the accumulation of amyloid fibrils or plaques in the brain leading to progressive loss of neuronal functions. Alzheimer's disease is one of the major NDD responsible for 60-80 % of all dementia cases. Currently, there are no curative or disease-reversing/modifying molecules for many of the NDDs except a few such as donepezil, rivastigmine, galantamine, carbidopa and levodopa which treat the disease-associated symptoms. Similarly, there are very few FDA-approved tracers such as flortaucipir (Tauvid) for tau fibril imaging and florbetaben (Neuraceq), flutemetamol (Vizamyl), and florbetapir (Amyvid) for amyloid imaging available for diagnosis. Recent advances in the cryogenic electron microscopy reported distinctly different microstructures for tau fibrils associated with different tauopathies highlighting the possibility to develop tauopathy-specific imaging agents and therapeutics. In addition, it is important to identify the proteins that are associated with disease development and progression to know about their 3D structure to develop various diagnostics, therapeutics and theranostic agents. The current article discusses in detail the disease-associated amyloid and non-amyloid proteins along with their structural insights. We comprehensively discussed various novel proteins associated with NDDs and their implications in disease pathology. In addition, we document various emerging chemical compounds developed for diagnosis and therapy of different NDDs with special emphasis on theranostic agents for better management of NDDs.
Collapse
Affiliation(s)
- Kapali Suri
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - Mansi Bhandari
- Department of computer science and engineering, Jamia Hamdard University, Hamdard Nagar, New Delhi, Delhi, 110062
| | - Vishakha Gupta
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| | - Virendra Kumar
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - N Arul Murugan
- Department of Computational Biology, Indraprastha Institute of Information Technology (IIIT-Delhi) Okhla, Phase III, New Delhi, 110020, India
| |
Collapse
|
28
|
Waugh ML, Wolf LM, Moore KA, Servoss SL, Moss MA. Rationally Designed Peptoid Inhibitors of Amyloid-β Oligomerization. Chembiochem 2024; 25:e202400060. [PMID: 38715149 PMCID: PMC11219258 DOI: 10.1002/cbic.202400060] [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: 01/22/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
While plaques comprised of fibrillar Aβ aggregates are hallmarks of Alzheimer's disease, soluble Aβ oligomers present higher neurotoxicity. Thus, one therapeutic approach is to prevent the formation of Aβ oligomers and reduce their associated harmful effects. We have proposed a peptoid mimic of the Aβ hydrophobic KLVFF core as an ideal candidate aggregation inhibitor due to its ability to evade proteolytic degradation via repositioning of the side chain from the α-carbon to the amide nitrogen. This peptoid, JPT1, utilizes chiral sidechains to achieve a helical structure, while C-terminal addition of two phenylalanine residues places aromatic groups on two sides of the helix with spacing designed to facilitate interaction with amyloid β-sheet structure. We have previously shown that JPT1 modulates Aβ fibril formation. Here, we demonstrate that JPT1 also modulates Aβ oligomerization, and we explore the role of the charge on the linker between the KLVFF mimic and the extended aromatic residues. Additionally, we demonstrate that peptoid-induced changes in Aβ oligomerization correlate with attenuation of oligomer-induced nuclear factor-κB activation in SH-SY5Y human neuroblastoma cells. These findings support the therapeutic potential of peptoids to target early stages of Aβ aggregation and impact the associated Aβ-induced cellular response.
Collapse
Affiliation(s)
- Mihyun Lim Waugh
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
| | - Lauren M Wolf
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
| | - Kelly A Moore
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
| | - Shannon L Servoss
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR 72701, USA
| | - Melissa A Moss
- Department of Biomedical Engineering, University of South Carolina, 3A46 Swearingen Engineering Center, Columbia, SC 29208, USA
- Department of Chemical Engineering, University of South Carolina, 2C02 Swearingen Engineering Center, Columbia, SC 29208, USA
| |
Collapse
|
29
|
Halder B, Ghosh S, Khan T, Pal S, Das N, Sen P. Tracking heterogenous protein aggregation at nanoscale through fluorescence correlation spectroscopy. Photochem Photobiol 2024; 100:989-999. [PMID: 39032082 DOI: 10.1111/php.14004] [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: 02/21/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/22/2024]
Abstract
Various biophysical techniques have been extensively employed to study protein aggregation due to its significance. Traditionally, these methods detect aggregation at micrometer length scales and micromolar concentrations. However, unlike in vitro, protein aggregation typically occurs at nanomolar concentrations in vivo. Here, using fluorescence correlation spectroscopy (FCS), we captured bromelain aggregation at concentrations as low as ~20 nM, surpassing the detection limit of traditional methods like thioflavin T fluorescence, scattering, and fluorescence microscopy by more than one order of magnitude. Moreover, using thioflavin T fluorescence-based FCS, we have detected larger aggregates at higher bromelain concentrations, which is undetectable in FCS otherwise. Importantly, our study reveals inherent heterogeneity in bromelain aggregation, inaccessible to ensemble-averaged techniques. The presented report may provide a platform for the characterization of premature aggregates at very low protein concentrations, which are thought to be functionally significant species in protein aggregation-induced diseases.
Collapse
Affiliation(s)
- Bisal Halder
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Shreya Ghosh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Tanmoy Khan
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Subhendu Pal
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Nilimesh Das
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Pratik Sen
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
30
|
Liu B, Li X, Liu Z, He B, Xu H, Cao J, Zeng F, Feng H, Ren Y, Li H, Wang T, Li J, Ye Y, Zhao L, Ran C, Li Y. Iterative Design of Near-Infrared Fluorescent Probes for Early Diagnosis of Alzheimer's Disease by Targeting Aβ Oligomers. J Med Chem 2024; 67:9104-9123. [PMID: 38829030 DOI: 10.1021/acs.jmedchem.4c00252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Amyloid-β oligomers (AβOs), crucial toxic proteins in early Alzheimer's disease (AD), precede the formation of Aβ plaques and cognitive impairment. In this context, we present our iterative process for developing novel near-infrared fluorescent (NIRF) probes specifically targeting AβOs, aimed at early AD diagnosis. An initial screening identified compound 18 as being highly selective for AβOs. Subsequent analysis revealed that compound 20 improved serum stability while retaining affinity for AβOs. The most promising iteration, compound 37, demonstrated exceptional qualities: a high affinity for AβOs, emission in the near-infrared region, and good biocompatibility. Significantly, ex vivo double staining indicated that compound 37 detected AβOs in AD mouse brain and in vivo imaging experiments showed that compound 37 could differentiate between 4-month-old AD mice and age-matched wild-type mice. Therefore, compound 37 has emerged as a valuable NIRF probe for early detection of AD and a useful tool in exploring AD's pathological mechanisms.
Collapse
Affiliation(s)
- Bing Liu
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Xiaofang Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Zhengyang Liu
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Bing He
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Hanyue Xu
- Nanjing Foreign Language School, Nanjing 210008, Jiangsu, China
| | - Jianqin Cao
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Fantian Zeng
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Haiwei Feng
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Yanwei Ren
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Haoyu Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Tianyu Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Jia Li
- Pathology and PDX Efficacy Center, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Yuting Ye
- Pathology and PDX Efficacy Center, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Li Zhao
- School of Basic Medicine and Clinical Pharmacology, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Yuyan Li
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211100, Jiangsu, China
| |
Collapse
|
31
|
Nguyen NK, Poduska B, Franks M, Bera M, MacCormack I, Lin G, Petroff AP, Das S, Nag A. A Copper-Selective Sensor and Its Inhibition of Copper-Amyloid Beta Aggregation. BIOSENSORS 2024; 14:247. [PMID: 38785721 PMCID: PMC11117483 DOI: 10.3390/bios14050247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/29/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Copper is an essential trace metal for biological processes in humans and animals. A low level of copper detection at physiological pH using fluorescent probes is very important for in vitro applications, such as the detection of copper in water or urine, and in vivo applications, such as tracking the dynamic copper concentrations inside cells. Copper homeostasis is disrupted in neurological diseases like Alzheimer's disease, and copper forms aggregates with amyloid beta (Ab42) peptide, resulting in senile plaques in Alzheimer's brains. Therefore, a selective copper detector probe that can detect amyloid beta peptide-copper aggregates and decrease the aggregate size has potential uses in medicine. We have developed a series of Cu2+-selective low fluorescent to high fluorescent tri and tetradentate dentate ligands and conjugated them with a peptide ligand to amyloid-beta binding peptide to increase the solubility of the compounds and make the resultant compounds bind to Cu2+-amyloid aggregates. The copper selective compounds were developed using chemical scaffolds known to have high affinity and selectivity for Cu2+, and their conjugates with peptides were tested for affinity and selectivity towards Cu2+. The test results were used to inform further improvement of the next compound. The final Cu2+ chelator-peptide conjugate we developed showed high selectivity for Cu2+ and high fluorescence properties. The compound bound 1:1 to Cu2+ ion, as determined from its Job's plot. Fluorescence of the ligand could be detected at nanomolar concentrations. The effect of this ligand on controlling Cu2+-Ab42 aggregation was studied using fluorescence assays and microscopy. It was found that the Cu2+-chelator-peptide conjugate efficiently reduced aggregate size and, therefore, acted as an inhibitor of Ab42-Cu2+ aggregation. Since high micromolar concentrations of Cu2+ are present in senile plaques, and Cu2+ accelerates the formation of toxic soluble aggregates of Ab42, which are precursors of insoluble plaques, the developed hybrid molecule can potentially serve as a therapeutic for Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Arundhati Nag
- Carlson School of Chemistry and Biochemistry, Clark University, 950 Main Street, Worcester, MA 01610, USA; (N.K.N.); (B.P.); (M.F.); (M.B.); (I.M.); (G.L.); (A.P.P.); (S.D.)
| |
Collapse
|
32
|
Haynes JR, Whitmore CA, Behof WJ, Landman CA, Ong HH, Feld AP, Suero IC, Greer CB, Gore JC, Wijesinghe P, Matsubara JA, Wadzinski BE, Spiller BW, Pham W. TARGETING SOLUBLE AMYLOID-BETA OLIGOMERS WITH A NOVEL NANOBODY. RESEARCH SQUARE 2024:rs.3.rs-3944211. [PMID: 38559050 PMCID: PMC10980145 DOI: 10.21203/rs.3.rs-3944211/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The classical amyloid cascade hypothesis postulates that the aggregation of amyloid plaques and the accumulation of intracellular hyperphosphorylated Tau tangles, together, lead to profound neuronal death. However, emerging research has demonstrated that soluble amyloid-β oligomers (SAβOs) accumulate early, prior to amyloid plaque formation. SAβOs induce memory impairment and disrupt cognitive function independent of amyloid-β plaques, and even in the absence of plaque formation. This work describes the development and characterization of a novel anti-SAβO (E3) nanobody generated from an alpaca immunized with SAβO. In-vitro assays and in-vivo studies using 5XFAD mice indicate that the fluorescein (FAM)-labeled E3 nanobody recognizes both SAβOs and amyloid-β plaques. The E3 nanobody traverses across the blood-brain barrier and binds to amyloid species in the brain of 5XFAD mice. Imaging of mouse brains reveals that SAβO and amyloid-β plaques are not only different in size, shape, and morphology, but also have a distinct spatial distribution in the brain. SAβOs are associated with neurons, while amyloid plaques reside in the extracellular matrix. The results of this study demonstrate that the SAβO nanobody can serve as a diagnostic agent with potential theragnostic applications in Alzheimer's disease.
Collapse
Affiliation(s)
- Justin R. Haynes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Clayton A. Whitmore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - William J. Behof
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Charlotte A. Landman
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Henry H. Ong
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew P. Feld
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Isabelle C. Suero
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Celeste B. Greer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - John C. Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, V5Z3N9, Canada
| | - Joanne A. Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, V5Z3N9, Canada
| | - Brian E. Wadzinski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Benjamin W. Spiller
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
33
|
Andrikopoulos N, Tang H, Wang Y, Liang X, Li Y, Davis TP, Ke PC. Exploring Peptido-Nanocomposites in the Context of Amyloid Diseases. Angew Chem Int Ed Engl 2024; 63:e202309958. [PMID: 37943171 DOI: 10.1002/anie.202309958] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 11/10/2023]
Abstract
Therapeutic peptides are a major class of pharmaceutical drugs owing to their target-binding specificity as well as their versatility in inhibiting aberrant protein-protein interactions associated with human pathologies. Within the realm of amyloid diseases, the use of peptides and peptidomimetics tailor-designed to overcome amyloidogenesis has been an active research endeavor since the late 90s. In more recent years, incorporating nanoparticles for enhancing the biocirculation and delivery of peptide drugs has emerged as a frontier in nanomedicine, and nanoparticles have further demonstrated a potency against amyloid aggregation and cellular inflammation to rival strategies employing small molecules, peptides, and antibodies. Despite these efforts, however, a fundamental understanding of the chemistry, characteristics and function of peptido-nanocomposites is lacking, and a systematic analysis of such strategy for combating a range of amyloid pathogeneses is missing. Here we review the history, principles and evolving chemistry of constructing peptido-nanocomposites from bottom up and discuss their future application against amyloid diseases that debilitate a significant portion of the global population.
Collapse
Affiliation(s)
- Nicholas Andrikopoulos
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Huayuan Tang
- College of Mechanics and Materials, Hohai University, Nanjing, 211100, China
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Yue Wang
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, China
| | - Xiufang Liang
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 510006, China
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Thomas P Davis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Pu Chun Ke
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld 4072, Australia
| |
Collapse
|
34
|
Chen PJ, Yao CA, Chien PC, Tsai HJ, Chen YR, Chuang JH, Chou PL, Lee GC, Lin W, Lin Y. Paeonol Derivative, 6'-Methyl Paeonol, Attenuates Aβ-Induced Pathophysiology in Cortical Neurons and in an Alzheimer's Disease Mice Model. ACS Chem Neurosci 2024; 15:724-734. [PMID: 38290213 DOI: 10.1021/acschemneuro.3c00633] [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] [Indexed: 02/01/2024] Open
Abstract
Herbs themselves and various herbal medicines are great resources for discovering therapeutic drugs for various diseases, including Alzheimer's disease (AD), one of the common neurodegenerative diseases. Utilizing mouse primary cortical neurons and DiBAC4(3), a voltage-sensitive indicator, we have set up a drug screening system and identified an herbal extraction compound, paeonol, obtained from Paeonia lactiflora; this compound is able to ameliorate the abnormal depolarization induced by Aβ42 oligomers. Our aim was to further find effective paeonol derivatives since paeonol has been previously studied. 6'-Methyl paeonol, one of the six paeonol derivatives surveyed, is able to inhibit the abnormal depolarization induced by Aβ oligomers. Furthermore, 6'-methyl paeonol is able to alleviate the NMDA- and AMPA-induced depolarization. When a molecular mechanism was investigated, 6'-methyl paeonol was found to reverse the Aβ-induced increase in ERK phosphorylation. At the animal level, mice injected with 6'-methyl paeonol showed little change in their basic physical parameters compared to the control mice. 6'-Methyl paeonol was able to ameliorate the impairment of memory and learning behavior in J20 mice, an AD mouse model, as measured by the Morris water maze. Thus, paeonol derivatives could provide a structural foundation for developing and designing an effective compound with promising clinical benefits.
Collapse
Affiliation(s)
| | - Chien-An Yao
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | | | | | | | | | - Pei-Li Chou
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | | | | | | |
Collapse
|
35
|
Gao H, Chen J, Huang Y, Zhao R. Advances in targeted tracking and detection of soluble amyloid-β aggregates as a biomarker of Alzheimer's disease. Talanta 2024; 268:125311. [PMID: 37857110 DOI: 10.1016/j.talanta.2023.125311] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/21/2023]
Abstract
Misfolding and aggregation of amyloid-β (Aβ) peptides are key hallmarks of Alzheimer's disease (AD). With accumulating evidence suggesting that different Aβ species have varied neurotoxicity and implications in AD development, the discovery of affinity ligands and analytical approaches to selective distinguish, detect, and monitor Aβ becomes an active research area. Remarkable advances have been achieved, which not only promote our understanding of the biophysical chemistry of the protein aggregation during neurodegeneration, but also provide promising tools for early detection of the disease. In view of this, we summarize the recent progress in selective and sensitive approaches for tracking and detection of Aβ species. Specific attentions are given to soluble Aβ oligomers, due to their crucial roles in AD development and occurrence at early stages. The design principle, performance of targeting units, and their cooperative effects with signal reporters for Aβ analysis are discussed. The applications of the novel targeting probes and sensing systems for dynamic monitoring oligomerization, measuring Aβ in biosamples and in vivo imaging in brain are summarized. Finally, the perspective and challenges are discussed regarding the future development of Aβ-targeting analytical tools to explore the unknown field to contribute to the early diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Han Gao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jian Chen
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China; School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
36
|
Ehlbeck JT, Grimard DM, Hacker RM, Garcia JA, Wall BJ, Bothwell PJ, Jones MA, Webb MI. Finding the best location: Improving the anti-amyloid ability of ruthenium(III) complexes with pyridine ligands. J Inorg Biochem 2024; 250:112424. [PMID: 37952508 DOI: 10.1016/j.jinorgbio.2023.112424] [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: 08/30/2023] [Revised: 10/12/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023]
Abstract
Alzheimer's disease (AD) is a devastating neurological disorder where one of the primary pathological hallmarks are aggregate deposits of the peptide amyloid-beta (Aβ). Although the Food and Drug Administration (FDA) has recently approved therapeutics that specifically target Aβ, resulting in the removal of these deposits, the associated costs of such treatments create a need for effective, yet cheaper, alternatives. Metal-based compounds are propitious therapeutic candidates as they exploit the metal-binding properties of Aβ, forming stable interactions with the peptide, thereby limiting its aggregation and toxicity. Previously, ruthenium-based complexes have shown a strong ability to modulate the aggregation and cytotoxicity of Aβ, where the incorporation of a primary amine on the coordinated heterocyclic ligand gave the greatest activity. To determine the importance of the location of the primary amine on the pyridine ligand, thereby establishing structure-activity relationships (SAR), four complexes (RuP1-4) were prepared and evaluated for their ability to coordinate and subsequently modulate the aggregation and cytotoxicity of Aβ. Coordination to Aβ was determined using three complementary spectroscopic methods: UV-Vis, 1H NMR, and circular dichroism (CD). Similarly, the impact of the complexes on Aβ aggregation was evaluated using three sequential methods of turbidity, dynamic light scattering (DLS), and transmission electron microscopy (TEM). Overall, the location of the primary amine on the pyridine ligand did affect the resultant anti-Aβ performance, with the 2-aminopyridine complex (RuP2) being the most active. This SAR will provide another guiding principle in the design of future metal-based anti-Aβ complexes.
Collapse
Affiliation(s)
- Johanna T Ehlbeck
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States of America
| | - Daniela M Grimard
- Department of Chemistry, SUNY Geneseo, Geneseo, NY 14454, United States of America
| | - Ryan M Hacker
- Department of Chemistry, SUNY Geneseo, Geneseo, NY 14454, United States of America
| | - Jimmy A Garcia
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States of America
| | - Brendan J Wall
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States of America
| | - Paige J Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115, United States of America
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States of America
| | - Michael I Webb
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States of America; Department of Chemistry, SUNY Geneseo, Geneseo, NY 14454, United States of America.
| |
Collapse
|
37
|
Senapati S, Secchi V, Cova F, Richman M, Villa I, Yehuda R, Shenberger Y, Campione M, Rahimipour S, Monguzzi A. Noninvasive Treatment of Alzheimer's Disease with Scintillating Nanotubes. Adv Healthc Mater 2023; 12:e2301527. [PMID: 37826854 PMCID: PMC11469333 DOI: 10.1002/adhm.202301527] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/28/2023] [Indexed: 10/14/2023]
Abstract
Effective and accessible treatments for Alzheimer's disease (AD) are urgently needed. Soluble Aβ oligomers are identified as neurotoxic species in AD and targeted in antibody-based drug development to mitigate cognitive decline. However, controversy exists concerning their efficacy and safety. In this study, an alternative strategy is proposed to inhibit the formation of Aβ oligomers by selectively oxidizing specific amino acids in the Aβ sequence, thereby preventing its aggregation. Targeted oxidation is achieved using biocompatible and blood-brain barrier-permeable multicomponent nanoscintillators that generate singlet oxygen upon X-ray interaction. Surface-modified scintillators interact selectively with Aβ and, upon X-ray irradiation, inhibit the formation of neurotoxic aggregates both in vitro and in vivo. Feeding transgenic Caenorhabditis elegans expressing human Aβ with the nanoscintillators and subsequent irradiation with soft X-ray reduces Aβ oligomer levels, extends lifespan, and restores memory and behavioral deficits. These findings support the potential of X-ray-based therapy for AD and warrant further development.
Collapse
Affiliation(s)
- Sudipta Senapati
- Department of ChemistryBar‐Ilan UniversityRamat‐Gan5290002Israel
| | - Valeria Secchi
- Department of Materials ScienceUniversity of Milano‐BicoccaVia R. Cozzi 55Milan20125Italy
| | - Francesca Cova
- Department of Materials ScienceUniversity of Milano‐BicoccaVia R. Cozzi 55Milan20125Italy
| | - Michal Richman
- Department of ChemistryBar‐Ilan UniversityRamat‐Gan5290002Israel
| | - Irene Villa
- Department of Materials ScienceUniversity of Milano‐BicoccaVia R. Cozzi 55Milan20125Italy
| | - Ronen Yehuda
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat Gan5290002Israel
| | - Yulia Shenberger
- Department of ChemistryBar‐Ilan UniversityRamat‐Gan5290002Israel
| | - Marcello Campione
- Department of Earth and Environmental SciencesUniversity of Milano‐BicoccaPiazza della Scienza 4Milan20126Italy
| | - Shai Rahimipour
- Department of ChemistryBar‐Ilan UniversityRamat‐Gan5290002Israel
| | - Angelo Monguzzi
- Department of Materials ScienceUniversity of Milano‐BicoccaVia R. Cozzi 55Milan20125Italy
| |
Collapse
|
38
|
Gao F, Chen J, Zhou Y, Cheng L, Hu M, Wang X. Recent progress of small-molecule-based theranostic agents in Alzheimer's disease. RSC Med Chem 2023; 14:2231-2245. [PMID: 37974955 PMCID: PMC10650505 DOI: 10.1039/d3md00330b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/11/2023] [Indexed: 11/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative dementia. As a multifactorial disease, AD involves several etiopathogenic mechanisms, in which multiple pathological factors are interconnected with each other. This complicated and unclear pathogenesis makes AD lack effective diagnosis and treatment. Theranostics, exerting the synergistic effect of diagnostic and therapeutic functions, would provide a promising strategy for exploring AD pathogenesis and developing drugs for combating AD. With the efforts in small drug-like molecules for both diagnosis and treatment of AD, small-molecule-based theranostic agents have attracted significant attention owing to their facile synthesis, high biocompatibility and reproducibility, and easy clearance from the body through the excretion systems. In this review, the small-molecule-based theranostic agents reported in the literature for anti-AD are classified into four groups according to their diagnostic modalities. Their design rationales, chemical structures, and working mechanisms for theranostics are summarized. Finally, the opportunities for small-molecule-based theranostic agents in AD are also proposed.
Collapse
Affiliation(s)
- Furong Gao
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University Nanjing 211816 P. R. China
| | - Jiefang Chen
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University Nanjing 211816 P. R. China
| | - Yuancun Zhou
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University Nanjing 211816 P. R. China
| | - Letong Cheng
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University Nanjing 211816 P. R. China
| | - Ming Hu
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University Nanjing 211816 P. R. China
| | - Xiaohui Wang
- Institute of Chemical Biology and Functional Molecules, State Key Laboratory of Materials-Oriented Chemical Engineering, School of Chemistry and Molecular Engineering, Nanjing Tech University Nanjing 211816 P. R. China
| |
Collapse
|
39
|
Atlante A, Valenti D. Mitochondrial Complex I and β-Amyloid Peptide Interplay in Alzheimer's Disease: A Critical Review of New and Old Little Regarded Findings. Int J Mol Sci 2023; 24:15951. [PMID: 37958934 PMCID: PMC10650435 DOI: 10.3390/ijms242115951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and the main cause of dementia which is characterized by a progressive cognitive decline that severely interferes with daily activities of personal life. At a pathological level, it is characterized by the accumulation of abnormal protein structures in the brain-β-amyloid (Aβ) plaques and Tau tangles-which interfere with communication between neurons and lead to their dysfunction and death. In recent years, research on AD has highlighted the critical involvement of mitochondria-the primary energy suppliers for our cells-in the onset and progression of the disease, since mitochondrial bioenergetic deficits precede the beginning of the disease and mitochondria are very sensitive to Aβ toxicity. On the other hand, if it is true that the accumulation of Aβ in the mitochondria leads to mitochondrial malfunctions, it is otherwise proven that mitochondrial dysfunction, through the generation of reactive oxygen species, causes an increase in Aβ production, by initiating a vicious cycle: there is therefore a bidirectional relationship between Aβ aggregation and mitochondrial dysfunction. Here, we focus on the latest news-but also on neglected evidence from the past-concerning the interplay between dysfunctional mitochondrial complex I, oxidative stress, and Aβ, in order to understand how their interplay is implicated in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy;
| | | |
Collapse
|
40
|
Gandy S. Systemically administered alcadein peptide p3-Alcβ neutralizes brain Alzheimer's Aβ oligomers. Trends Mol Med 2023; 29:487-488. [PMID: 37169660 DOI: 10.1016/j.molmed.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Alcadeins are among the 90 known substrates for γ-secretase, the aspartyl proteinase complex that liberates the amyloid β (Aβ) peptide by intramembranous cleavage of the ß-carboxy terminal fragment of the Alzheimer's amyloid precursor protein (APP). A new study by Hata et al. provides some surprising results regarding a potential role for alcadeins and their fragments in the prevention and/or treatment of the dementia of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; James J Peters VA Medical Center, Bronx, NY 10468, USA.
| |
Collapse
|
41
|
Penke B, Szűcs M, Bogár F. New Pathways Identify Novel Drug Targets for the Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:5383. [PMID: 36982456 PMCID: PMC10049476 DOI: 10.3390/ijms24065383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable, progressive neurodegenerative disorder. AD is a complex and multifactorial disease that is responsible for 60-80% of dementia cases. Aging, genetic factors, and epigenetic changes are the main risk factors for AD. Two aggregation-prone proteins play a decisive role in AD pathogenesis: β-amyloid (Aβ) and hyperphosphorylated tau (pTau). Both of them form deposits and diffusible toxic aggregates in the brain. These proteins are the biomarkers of AD. Different hypotheses have tried to explain AD pathogenesis and served as platforms for AD drug research. Experiments demonstrated that both Aβ and pTau might start neurodegenerative processes and are necessary for cognitive decline. The two pathologies act in synergy. Inhibition of the formation of toxic Aβ and pTau aggregates has been an old drug target. Recently, successful Aβ clearance by monoclonal antibodies has raised new hopes for AD treatments if the disease is detected at early stages. More recently, novel targets, e.g., improvements in amyloid clearance from the brain, application of small heat shock proteins (Hsps), modulation of chronic neuroinflammation by different receptor ligands, modulation of microglial phagocytosis, and increase in myelination have been revealed in AD research.
Collapse
Affiliation(s)
- Botond Penke
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary
| | - Mária Szűcs
- Department of Medical Chemistry, University of Szeged, Dóm Square 8, H-6720 Szeged, Hungary
| | - Ferenc Bogár
- ELKH-SZTE Biomimetic Systems Research Group, Eötvös Loránd Research Network (ELKH), Dóm Square 8, H-6720 Szeged, Hungary
| |
Collapse
|
42
|
Ghosh S, Ali R, Verma S. Aβ-oligomers: A potential therapeutic target for Alzheimer's disease. Int J Biol Macromol 2023; 239:124231. [PMID: 36996958 DOI: 10.1016/j.ijbiomac.2023.124231] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023]
Abstract
The cascade of amyloid formation relates to multiple complex events at the molecular level. Previous research has established amyloid plaque deposition as the leading cause of Alzheimer's disease (AD) pathogenesis, detected mainly in aged population. The primary components of the plaques are two alloforms of amyloid-beta (Aβ), Aβ1-42 and Aβ1-40 peptides. Recent studies have provided considerable evidence contrary to the previous claim indicating that amyloid-beta oligomers (AβOs) as the main culprit responsible for AD-associated neurotoxicity and pathogenesis. In this review, we have discussed the primary features of AβOs, such as assembly formation, the kinetics of oligomer formation, interactions with various membranes/membrane receptors, the origin of toxicity, and oligomer-specific detection methods. Recently, the discovery of rationally designed antibodies has opened a gateway for using synthesized peptides as a grafting component in the complementarity determining region (CDR) of antibodies. Thus, the Aβ sequence motif or the complementary peptide sequence in the opposite strand of the β-sheet (extracted from the Protein Data Bank: PDB) helps design oligomer-specific inhibitors. The microscopic event responsible for oligomer formation can be targeted, and thus prevention of the overall macroscopic behaviour of the aggregation or the associated toxicity can be achieved. We have carefully reviewed the oligomer formation kinetics and associated parameters. Besides, we have depicted a thorough understanding of how the synthesized peptide inhibitors can impede the early aggregates (oligomers), mature fibrils, monomers, or a mixture of the species. The oligomer-specific inhibitors (peptides or peptide fragments) lack in-depth chemical kinetics and optimization control-based screening. In the present review, we have proposed a hypothesis for effectively screening oligomer-specific inhibitors using the chemical kinetics (determining the kinetic parameters) and optimization control strategy (cost-dependent analysis). Further, it may be possible to implement the structure-kinetic-activity-relationship (SKAR) strategy instead of structure-activity-relationship (SAR) to improve the inhibitor's activity. The controlled optimization of the kinetic parameters and dose usage will be beneficial for narrowing the search window for the inhibitors.
Collapse
|
43
|
Habashi M, Chauhan PS, Vutla S, Senapati S, Diachkov M, El-Husseini A, Guérin B, Lubell WD, Rahimipour S. Aza-Residue Modulation of Cyclic d,l-α-Peptide Nanotube Assembly with Enhanced Anti-Amyloidogenic Activity. J Med Chem 2023; 66:3058-3072. [PMID: 36763536 DOI: 10.1021/acs.jmedchem.2c02049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Transient soluble oligomers of amyloid-β (Aβ) are considered among the most toxic species in Alzheimer's disease (AD). Soluble Aβ oligomers accumulate early prior to insoluble plaque formation and cognitive impairment. The cyclic d,l-α-peptide CP-2 (1) self-assembles into nanotubes and demonstrates promising anti-amyloidogenic activity likely by a mechanism involving engagement of soluble oligomers. Systematic replacement of the residues in peptide 1 with aza-amino acid counterparts was performed to explore the effects of hydrogen bonding on propensity to mitigate Aβ aggregation and toxicity. Certain azapeptides exhibited improved ability to engage, alter the secondary structure, and inhibit aggregation of Aβ. Moreover, certain azapeptides disassembled preformed Aβ fibrils and protected cells from Aβ-mediated toxicity. Substitution of the l-norleucine3 and d-serine6 residues in peptide 1 with aza-norleucine and aza-homoserine provided, respectively, nontoxic [azaNle3]-1 (4) and [azaHse6]-1 (7), that significantly abated symptoms in a transgenic Caenorhabditis elegans AD model by decreasing Aβ oligomer levels.
Collapse
Affiliation(s)
- Maram Habashi
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Pradeep S Chauhan
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Suresh Vutla
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Sudipta Senapati
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Mykhailo Diachkov
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Ali El-Husseini
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke 3001, 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
- Sherbrooke Molecular Imaging Center (CIMS), Research centre of the CHUS (CRCHUS) 3001, 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - William D Lubell
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Shai Rahimipour
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|