1
|
Forloni G. Doxycycline: An essential tool for Alzheimer's disease. Biomed Pharmacother 2025; 188:118159. [PMID: 40367557 DOI: 10.1016/j.biopha.2025.118159] [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: 03/07/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025] Open
Abstract
The identification of active interventions in neurodegenerative disorders is a major challenge in neurology; the use of repurposed drugs may represent a valuable strategy. Tetracyclines, a second generation of antibiotic molecules, offer various potential applications. Following an anecdotal observation of the potential anti-amyloidogenic activity of iododoxorubicin, the search for chemical analogs with a better safety profile led to tetracyclines. Their heterocyclic structures with a planar conformation interfere with b-sheet amyloid formation. Thus, doxycycline, a derivative with favorable blood-brain barrier penetration, emerged as a strong candidate to combat peripheral and central amyloidosis. In particular, we tested the anti-prion activity of doxycycline in vitro and in vivo experiments, confirming its capacity to disrupt or inhibit the formation of prion protein aggregates associated with pathological events. Treatment with doxycycline in human subjects with prion - related encephalopathies yielded contradictory results, suggesting that a preventive approach is a more favorable condition to verify efficacy; a clinical trial involving subjects at genetic risk of developing fatal familial insomnia, exposed to doxycycline for ten years, is currently ongoing. The anti-amyloidogenic capacity of doxycycline, combined with its safety profile in long-term treatment, has suggested its use in peripheral amyloidosis, which was tested with positive results. A specific interaction with β-amyloid or α-synuclein oligomers, as well as tau aggregation has also been demonstrated. More recently, the action of doxycycline has been extended to its anti-inflammatory and antioxidant capacities. In particular, the anti-inflammatory activity of doxycycline may explain the drug 's efficacy in numerous experimental models where protein misfolding has been associated with neuroinflammation, including Huntington's and Parkinson' s diseases. Thus, the pleiotropic action of doxycycline appears to be an interesting tool for addressing progressive neuronal dysfunction in multifactorial neurodegenerative diseases. The application of precision medicine principles to doxycycline treatment represents the best strategy to determine its efficacy. These aspects are illustrated here concerning another pleiotropic tetracycline, minocycline.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy.
| |
Collapse
|
2
|
Lin J, Li H, Jiang L, Li J. Novel strategies for targeting tau oligomers in neurodegenerative diseases. J Neurol 2025; 272:383. [PMID: 40335778 DOI: 10.1007/s00415-025-13117-w] [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: 03/10/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025]
Abstract
Tau protein is a soluble microtubule-associated protein enriched in neurons, is mainly distributed in the central nervous system, and is responsible for stabilizing neurons. Tau maintains nerve cell morphology and internal transport by binding to normal microtubules. In neurodegenerative diseases, such as Alzheimer's disease (AD), tau proteins undergo aberrant phosphorylation, resulting in their removal from microtubules and the formation of neurofibrillary tangles (NFTs), which are key pathological features. In contrast to the late formation of non-soluble NFTs, early, smaller, soluble tau oligomers (tauO) with disseminated toxicity are considered necessary in neurodegenerative disorders, such as the primary form of tau toxicity in the AD process. Although an increasing number of studies are focusing on tauO, there are still problems to be solved, mainly concerning the molecular and inhibitory mechanisms of tauO toxicity. In this paper, we summarize the new strategies for the molecular mechanisms of tauO toxicity, detection methods, and interventions in the last five years. An outlook on these new strategies and the challenges that may be foreseen is presented to provide new directions for future applications in the clinical treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Lin
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Tongzipo Road, Changsha, 410000, Hunan, China
| | - Hong Li
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Tongzipo Road, Changsha, 410000, Hunan, China
| | - Lingxia Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road, Changsha, 410000, Hunan, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Tongzipo Road, Changsha, 410000, Hunan, China.
| |
Collapse
|
3
|
Hussein SA, Tolba MF, Michel HE, Albohy A, Azab SS. In silico and In vivo protective effect of biochanin-A mitigating doxorubicin- induced cognitive deficits and neuroinflammation: Insights to the role of p-Tau and miR-132. Neurotoxicology 2025; 107:22-36. [PMID: 39848501 DOI: 10.1016/j.neuro.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/08/2025] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Doxorubicin (DOX)-induced chemobrain has been reported in several studies. Its main culprit is the induction of massive amounts of reactive oxygen species (ROS), hence triggering damage to brain tissues and thus leading to neuroinflammation. Biochanin A (BIO-A) is known to be an antioxidant, anti-inflammatory, and neuroprotective agent. An in silico study was designed to examine the potential neuroprotective effect of BIO-A. An in vivo study was used to evaluate the modulatory effect of BIO-A on cognitive impairment engendered by DOX. The insilico investigation proved the putative neuroprotective effect of BIO-A. In the in vivo study, BIO-A treatment counteracted DOX-induced memory deficits, as evidenced by improved spatial memory in rats compared to the DOX-only group. BIO-A also reversed DOX-triggered hippocampal neurodegeneration and neuroinflammation, supported by a significant decrease in tissue contents of NF-κB (p65) by 32 % and NLRP3 by 36 % versus the DOX-only group. BIO-A also abrogated DOX-induced neurodegneration, as evidenced by increasing SIRT1 content by 2-fold and BDNF content by 2-fold versus the DOX-only group in hippocampal tissues. In addition, BIO-A ameliorated DOX-augmented apoptosis in the hippocampus, as evidenced by lowering caspase-3 content in the hippocampus by 26 % versus the DOX-only group. Regarding tauopathy, BIO-A reversed DOX-increased tauopathy by 35 % versus the DOX-only group. The neuroprotectant miR-132 was increased by BIO-A in hippocampal tissues by 4-fold, contrary to the DOX-only group. Thus, BIO-A treatment modulated DOX-induced behavioral, histological, and molecular changes in the hippocampi of rats. Further studies are recommended to evaluate BIO-A in early clinical trials for the purpose of protection against chemobrain in cancer patients.
Collapse
Affiliation(s)
- Sarah A Hussein
- Center for Drug Discovery Research and Development, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mai F Tolba
- Center for Drug Discovery Research and Development, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Amgad Albohy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo 11837, Egypt; Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo 11837, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| |
Collapse
|
4
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025; 16:2105-2135. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
5
|
Conceição M, Di Filippo LD, Duarte JL, Beserra FP, Gremião MPD, Chorilli M. Repurposing doxycycline for Alzheimer's treatment: Challenges from a nano-based drug delivery perspective. Brain Behav Immun Health 2024; 42:100894. [PMID: 39525305 PMCID: PMC11550769 DOI: 10.1016/j.bbih.2024.100894] [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: 03/29/2024] [Revised: 07/16/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Drug repurposing, also known as drug repositioning, involves identifying new applications for drugs whose effects in a disease are already established. Doxycycline, a broad-spectrum antibiotic belonging to the tetracycline class, has demonstrated potential activity against neurodegenerative diseases like Alzheimer's and Parkinson's. However, despite its promise, the repurposed use of doxycycline encounters challenges in reaching the brain in adequate concentrations to exert its effects. To address this issue, nanostructured systems offer an innovative approach that can enhance brain targeting and the desired therapeutic outcomes. This review discusses the advances in doxycycline repurposing for Alzheimer's disease, presenting physicochemical and biological aspects that permeate doxycycline's repositioning and its application in nano-based delivery systems.
Collapse
Affiliation(s)
- Mariana Conceição
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Leonardo Delello Di Filippo
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Jonatas Lobato Duarte
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Fernando Pereira Beserra
- Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Maria Palmira Daflon Gremião
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
6
|
Li S, Zhao L, Xiao J, Guo Y, Fu R, Zhang Y, Xu S. The gut microbiome: an important role in neurodegenerative diseases and their therapeutic advances. Mol Cell Biochem 2024; 479:2217-2243. [PMID: 37787835 DOI: 10.1007/s11010-023-04853-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/06/2023] [Indexed: 10/04/2023]
Abstract
There are complex interactions between the gut and the brain. With increasing research on the relationship between gut microbiota and brain function, accumulated clinical and preclinical evidence suggests that gut microbiota is intimately involved in the pathogenesis of neurodegenerative diseases (NDs). Increasingly studies are beginning to focus on the association between gut microbiota and central nervous system (CNS) degenerative pathologies to find potential therapies for these refractory diseases. In this review, we summarize the changes in the gut microbiota in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis and contribute to our understanding of the function of the gut microbiota in NDs and its possible involvement in the pathogenesis. We subsequently discuss therapeutic approaches targeting gut microbial abnormalities in these diseases, including antibiotics, diet, probiotics, and fecal microbiota transplantation (FMT). Furthermore, we summarize some completed and ongoing clinical trials of interventions with gut microbes for NDs, which may provide new ideas for studying NDs.
Collapse
Affiliation(s)
- Songlin Li
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Jie Xiao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
7
|
Ploper D, Pernicone AO, Tomas-Grau RH, Manzano VE, Socías SB, Teran MDM, Budeguer Isa V, Sosa-Padilla B, González-Lizárraga F, Avila CL, Guayán ML, Chaves S, Cruz H, Vera Pingitore E, Varela O, Chehín R. Design, Synthesis, and Evaluation of a Novel Conjugate Molecule with Dopaminergic and Neuroprotective Activities for Parkinson's Disease. ACS Chem Neurosci 2024; 15:2795-2810. [PMID: 38991155 DOI: 10.1021/acschemneuro.4c00169] [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: 07/13/2024] Open
Abstract
The escalating prevalence of Parkinson's disease (PD) underscores the need for innovative therapeutic interventions since current palliative measures, including the standard l-Dopa formulations, face challenges of tolerance and side effects while failing to address the underlying neurodegenerative processes. Here, we introduce DAD9, a novel conjugate molecule that aims to combine symptomatic relief with disease-modifying strategies for PD. Crafted through knowledge-guided chemistry, the molecule combines a nonantibiotic doxycycline derivative with dopamine, preserving neuroprotective attributes while maintaining dopaminergic agonism. This compound exhibited no off-target effects on PD-relevant cell functions and sustained antioxidant and anti-inflammatory properties of the tetracycline precursor. Furthermore, it effectively interfered with the formation and seeding of toxic α-synuclein aggregates without producing detrimental oxidative species. In addition, DAD9 was able to activate dopamine receptors, and docking simulations shed light onto the molecular details of this interaction. These findings position DAD9 as a potential neuroprotective dopaminergic agonist, promising advancements in PD therapeutics.
Collapse
Affiliation(s)
- Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Agustín O Pernicone
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Rodrigo H Tomas-Grau
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Verónica E Manzano
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Sergio B Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - María Del Milagro Teran
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Valentina Budeguer Isa
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Bernardo Sosa-Padilla
- Instituto de Química del Noroeste Argentino (INQUINOA) (CONICET), Universidad Nacional de Tucumán (UNT), Ayacucho 471, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - César L Avila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - María Laura Guayán
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Silvina Chaves
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Hernán Cruz
- Instituto de Química Física, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Lorenzo 456, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Esteban Vera Pingitore
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| | - Oscar Varela
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Universidad de Buenos Aires, Pabellón 2, C1428EHA Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigación en Hidratos de Carbono (CIHIDECAR), Ciudad Universitaria, C1428EHA Buenos Aires, Argentina
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET), Universidad Nacional de Tucumán (UNT), Ministerio de Salud Pública de Tucumán - SIPROSA, Pasaje Dorrego 1080, 4000 San Miguel de Tucumán, Tucumán, Argentina
| |
Collapse
|
8
|
Rose C, Tomas-Grau RH, Zabala B, Michel PP, Brunel JM, Chehín R, Raisman-Vozari R, Ferrié L, Figadère B. C9-Functionalized Doxycycline Analogs as Drug Candidates to Prevent Pathological α-Synuclein Aggregation and Neuroinflammation in Parkinson's Disease Degeneration. ChemMedChem 2024; 19:e202300597. [PMID: 38526011 DOI: 10.1002/cmdc.202300597] [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: 10/31/2023] [Revised: 03/05/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Doxycycline, a semi-synthetic tetracycline, is a widely used antibiotic for treating mild-to-moderate infections, including skin problems. However, its anti-inflammatory and antioxidant properties, combined with its ability to interfere with α-synuclein aggregation, make it an attractive candidate for repositioning in Parkinson's disease. Nevertheless, the antibiotic activity of doxycycline restricts its potential use for long-term treatment of Parkinsonian patients. In the search for non-antibiotic tetracyclines that could operate against Parkinson's disease pathomechanisms, eighteen novel doxycycline derivatives were designed. Specifically, the dimethyl-amino group at C4 was reduced, resulting in limited antimicrobial activity, and several coupling reactions were performed at position C9 of the aromatic D ring, this position being one of the most reactive for introducing substituents. Using the Thioflavin-T assay, we found seven compounds were more effective than doxycycline in inhibiting α-synuclein aggregation. Furthermore, two of these derivatives exhibited better anti-inflammatory effects than doxycycline in a culture system of microglial cells used to model Parkinson's disease neuroinflammatory processes. Overall, through structure-activity relationship studies, we identified two newly designed tetracyclines as promising drug candidates for Parkinson's disease treatment.
Collapse
Affiliation(s)
- Clémence Rose
- BioCIS, CNRS, Université Paris-Saclay, 91400, Orsay, France
| | | | - Brenda Zabala
- Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, Hôpital Pitié Salpêtrière, 75013, Paris, France
| | - Patrick Pierre Michel
- Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, Hôpital Pitié Salpêtrière, 75013, Paris, France
| | - Jean-Michel Brunel
- UMR_MD1 Membranes et Cibles Thérapeutiques, U1261 INSERM, Aix-Marseille Université, 13385, Marseille, France
| | - Rosana Chehín
- IMMCA, CONICET-UNT-SIPROSA, Tucumán, 4000, Argentina
| | - Rita Raisman-Vozari
- Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, Hôpital Pitié Salpêtrière, 75013, Paris, France
| | - Laurent Ferrié
- BioCIS, CNRS, Université Paris-Saclay, 91400, Orsay, France
| | - Bruno Figadère
- BioCIS, CNRS, Université Paris-Saclay, 91400, Orsay, France
| |
Collapse
|
9
|
Suslov AV, Panas A, Sinelnikov MY, Maslennikov RV, Trishina AS, Zharikova TS, Zharova NV, Kalinin DV, Pontes-Silva A, Zharikov YO. Applied physiology: gut microbiota and antimicrobial therapy. Eur J Appl Physiol 2024; 124:1631-1643. [PMID: 38683402 DOI: 10.1007/s00421-024-05496-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
The gut microbiota plays an important role in maintaining human health and in the pathogenesis of several diseases. Antibiotics are among the most commonly prescribed drugs and have a significant impact on the structure and function of the gut microbiota. The understanding that a healthy gut microbiota prevents the development of many diseases has also led to its consideration as a potential therapeutic target. At the same time, any factor that alters the gut microbiota becomes important in this approach. Exercise and antibacterial therapy have a direct effect on the microbiota. The review reflects the current state of publications on the mechanisms of intestinal bacterial involvement in the pathogenesis of cardiovascular, metabolic, and neurodegenerative diseases. The physiological mechanisms of the influence of physical activity on the composition of the gut microbiota are considered. The mechanisms of the common interface between exercise and antibacterial therapy will be considered using the example of several socially important diseases. The aim of the study is to show the physiological relationship between the effects of exercise and antibiotics on the gut microbiota.
Collapse
Affiliation(s)
- Andrey V Suslov
- Russian National Centre of Surgery, Avtsyn Research Institute of Human Morphology, Moscow, 117418, Russia
- Pirogov Russian National Research Medical University (RNRMU), Moscow, 117997, Russia
| | - Alin Panas
- N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Oncology, Radiotherapy and Reconstructive Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119048, Russia
| | - Roman V Maslennikov
- Department of Internal Medicine, Gastroenterology and Hepatology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Aleksandra S Trishina
- N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, Bld. 2, Moscow, 119991, Russia
| | - Tatyana S Zharikova
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 125009, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nataliya V Zharova
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 125009, Russia
| | - Dmitry V Kalinin
- Pathology Department, A.V. Vishnevsky National Medical Research Center of Surgery, Moscow, 115093, Russia
| | - André Pontes-Silva
- Postgraduate Program in Physical Therapy (PPGFT), Department of Physical Therapy (DFisio), Universidade Federal de São Carlos (UFSCar), São Carlos (SP), Brazil.
| | - Yury O Zharikov
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 125009, Russia
| |
Collapse
|
10
|
Gandini A, Gonçalves AE, Strocchi S, Albertini C, Janočková J, Tramarin A, Grifoni D, Poeta E, Soukup O, Muñoz-Torrero D, Monti B, Sabaté R, Bartolini M, Legname G, Bolognesi ML. Discovery of Dual Aβ/Tau Inhibitors and Evaluation of Their Therapeutic Effect on a Drosophila Model of Alzheimer's Disease. ACS Chem Neurosci 2022; 13:3314-3329. [PMID: 36445009 PMCID: PMC9732823 DOI: 10.1021/acschemneuro.2c00357] [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] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD), the most common type of dementia, currently represents an extremely challenging and unmet medical need worldwide. Amyloid-β (Aβ) and Tau proteins are prototypical AD hallmarks, as well as validated drug targets. Accumulating evidence now suggests that they synergistically contribute to disease pathogenesis. This could not only help explain negative results from anti-Aβ clinical trials but also indicate that therapies solely directed at one of them may have to be reconsidered. Based on this, herein, we describe the development of a focused library of 2,4-thiazolidinedione (TZD)-based bivalent derivatives as dual Aβ and Tau aggregation inhibitors. The aggregating activity of the 24 synthesized derivatives was tested in intact Escherichia coli cells overexpressing Aβ42 and Tau proteins. We then evaluated their neuronal toxicity and ability to cross the blood-brain barrier (BBB), together with the in vitro interaction with the two isolated proteins. Finally, the most promising (most active, nontoxic, and BBB-permeable) compounds 22 and 23 were tested in vivo, in a Drosophila melanogaster model of AD. The carbazole derivative 22 (20 μM) showed extremely encouraging results, being able to improve both the lifespan and the climbing abilities of Aβ42 expressing flies and generating a better outcome than doxycycline (50 μM). Moreover, 22 proved to be able to decrease Aβ42 aggregates in the brains of the flies. We conclude that bivalent small molecules based on 22 deserve further attention as hits for dual Aβ/Tau aggregation inhibition in AD.
Collapse
Affiliation(s)
- Annachiara Gandini
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy,Department
of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136Trieste, Italy
| | - Ana Elisa Gonçalves
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy,Pharmaceutical
Sciences Postgraduate Program, Center of Health Sciences, Universidade do Vale do Itajaí, Rua Uruguai 458, 88302-202Itajaí, Santa Catarina, Brazil
| | - Silvia Strocchi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy
| | - Claudia Albertini
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy
| | - Jana Janočková
- Biomedical
Research Center, University Hospital Hradec
Kralove, 500 00Hradec Kralove, Czech Republic
| | - Anna Tramarin
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy
| | - Daniela Grifoni
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy,Department
of Life, Health and Environmental Sciences, University of L’Aquila, Via Vetoio, Coppito II, 67100L’Aquila, Italy
| | - Eleonora Poeta
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy
| | - Ondrej Soukup
- Biomedical
Research Center, University Hospital Hradec
Kralove, 500 00Hradec Kralove, Czech Republic
| | - Diego Muñoz-Torrero
- Laboratory
of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy
and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona (UB), Av. Joan XXIII 27-31, E-08028Barcelona, Spain
| | - Barbara Monti
- Pharmaceutical
Sciences Postgraduate Program, Center of Health Sciences, Universidade do Vale do Itajaí, Rua Uruguai 458, 88302-202Itajaí, Santa Catarina, Brazil
| | - Raimon Sabaté
- Department
of Pharmacy and Pharmaceutical Technology and Physical Chemistry,
Faculty of Pharmacy and Food Science, University
of Barcelona, Av Joan
XXIII 27-31, E-08028Barcelona, Spain
| | - Manuela Bartolini
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy
| | - Giuseppe Legname
- Department
of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136Trieste, Italy
| | - Maria Laura Bolognesi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, I-40126Bologna, Italy,. Tel: +39 0512099718
| |
Collapse
|
11
|
Natale C, Barzago MM, Colnaghi L, De Luigi A, Orsini F, Fioriti L, Diomede L. A Combined Cell-Worm Approach to Search for Compounds Counteracting the Toxicity of Tau Oligomers In Vivo. Int J Mol Sci 2022; 23:11277. [PMID: 36232578 PMCID: PMC9569484 DOI: 10.3390/ijms231911277] [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: 07/26/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
A clear relationship between the tau assemblies and toxicity has still to be established. To correlate the tau conformation with its proteotoxic effect in vivo, we developed an innovative cell-worm-based approach. HEK293 cells expressing tau P301L under a tetracycline-inducible system (HEK T-Rex) were employed to produce different tau assemblies whose proteotoxic potential was evaluated using C. elegans. Lysates from cells induced for five days significantly reduced the worm's locomotor activity. This toxic effect was not related to the total amount of tau produced by cells or to its phosphorylation state but was related to the formation of multimeric tau assemblies, particularly tetrameric ones. We investigated the applicability of this approach for testing compounds acting against oligomeric tau toxicity, using doxycycline (Doxy) as a prototype drug. Doxy affected tau solubility and promoted the disassembly of already formed toxic aggregates in lysates of cells induced for five days. These effects translated into a dose-dependent protective action in C. elegans. These findings confirm the validity of the combined HEK T-Rex cells and the C. elegans-based approach as a platform for pharmacological screening.
Collapse
Affiliation(s)
- Carmina Natale
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Maria Monica Barzago
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luca Colnaghi
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Franca Orsini
- Dulbecco Telethon Institute and Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luana Fioriti
- Dulbecco Telethon Institute and Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| |
Collapse
|
12
|
Tomas-Grau R, González-Lizárraga F, Ploper D, Avila CL, Socías SB, Besnault P, Tourville A, Mella RM, Villacé P, Salado C, Rose C, Seon-Méniel B, Brunel JM, Ferrié L, Raisman-Vozari R, Michel PP, Figadère B, Chehín R. Neuroprotective Effects of a Novel Demeclocycline Derivative Lacking Antibiotic Activity: From a Hit to a Promising Lead Compound. Cells 2022; 11:cells11172759. [PMID: 36078167 PMCID: PMC9454755 DOI: 10.3390/cells11172759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/09/2022] Open
Abstract
The antibiotic tetracycline demeclocycline (DMC) was recently reported to rescue α-synuclein (α-Syn) fibril-induced pathology. However, the antimicrobial activity of DMC precludes its potential use in long-term neuroprotective treatments. Here, we synthesized a doubly reduced DMC (DDMC) derivative with residual antibiotic activity and improved neuroprotective effects. The molecule was obtained by removal the dimethylamino substituent at position 4 and the reduction of the hydroxyl group at position 12a on ring A of DMC. The modifications strongly diminished its antibiotic activity against Gram-positive and Gram-negative bacteria. Moreover, this compound preserved the low toxicity of DMC in dopaminergic cell lines while improving its ability to interfere with α-Syn amyloid-like aggregation, showing the highest effectiveness of all tetracyclines tested. Likewise, DDMC demonstrated the ability to reduce seeding induced by the exogenous addition of α-Syn preformed fibrils (α-SynPFF) in biophysical assays and in a SH-SY5Y-α-Syn-tRFP cell model. In addition, DDMC rendered α-SynPFF less inflammogenic. Our results suggest that DDMC may be a promising drug candidate for hit-to-lead development and preclinical studies in Parkinson's disease and other synucleinopathies.
Collapse
Affiliation(s)
- Rodrigo Tomas-Grau
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - César L. Avila
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Sergio B. Socías
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
| | - Pierre Besnault
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Aurore Tourville
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Rosa M. Mella
- Innoprot SL, Parque Tecnológico de Bizkaia, Edificio 502, 48160 Derio, Spain
| | - Patricia Villacé
- Innoprot SL, Parque Tecnológico de Bizkaia, Edificio 502, 48160 Derio, Spain
| | - Clarisa Salado
- Innoprot SL, Parque Tecnológico de Bizkaia, Edificio 502, 48160 Derio, Spain
| | - Clémence Rose
- BioCIS, Université Paris-Saclay, CNRS, 92290 Châtenay-Malabry, France
| | | | - Jean-Michel Brunel
- UMR_MD1 “Membranes et Cibles Thérapeutiques”, U1261 INSERM, Aix-Marseille Université, 13385 Marseille, France
| | - Laurent Ferrié
- BioCIS, Université Paris-Saclay, CNRS, 92290 Châtenay-Malabry, France
| | - Rita Raisman-Vozari
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Patrick P. Michel
- Paris Brain Institute-ICM, Inserm, CNRS, Sorbonne Université APHP, Hôpital de la Pitié la Pitié-Salpêtrière, 75013 Paris, France
| | - Bruno Figadère
- BioCIS, Université Paris-Saclay, CNRS, 92290 Châtenay-Malabry, France
- Correspondence: (B.F.); (R.C.)
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), Pasaje Dorrego 1080, San Miguel de Tucumán 4000, Argentina
- Correspondence: (B.F.); (R.C.)
| |
Collapse
|
13
|
Forloni G, La Vitola P, Balducci C. Oligomeropathies, inflammation and prion protein binding. Front Neurosci 2022; 16:822420. [PMID: 36081661 PMCID: PMC9445368 DOI: 10.3389/fnins.2022.822420] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The central role of oligomers, small soluble aggregates of misfolded proteins, in the pathogenesis of neurodegenerative disorders is recognized in numerous experimental conditions and is compatible with clinical evidence. To underline this concept, some years ago we coined the term oligomeropathies to define the common mechanism of action of protein misfolding diseases like Alzheimer, Parkinson or prion diseases. Using simple experimental conditions, with direct application of synthetic β amyloid or α-synuclein oligomers intraventricularly at micromolar concentrations, we could detect differences and similarities in the biological consequences. The two oligomer species affected cognitive behavior, neuronal dysfunction and cerebral inflammatory reactions with distinct mechanisms. In these experimental conditions the proposed mediatory role of cellular prion protein in oligomer activities was not confirmed. Together with oligomers, inflammation at different levels can be important early in neurodegenerative disorders; both β amyloid and α-synuclein oligomers induce inflammation and its control strongly affects neuronal dysfunction. This review summarizes our studies with β-amyloid or α-synuclein oligomers, also considering the potential curative role of doxycycline, a well-known antibiotic with anti-amyloidogenic and anti-inflammatory activities. These actions are analyzed in terms of the therapeutic prospects.
Collapse
|
14
|
The potential use of tetracyclines in neurodegenerative diseases and the role of nano-based drug delivery systems. Eur J Pharm Sci 2022; 175:106237. [PMID: 35710076 DOI: 10.1016/j.ejps.2022.106237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/07/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022]
Abstract
Neurodegenerative diseases are still a challenge for effective treatments. The high cost of approved drugs, severity of side effects, injection site pain, and restrictions on drug delivery to the Central Nervous System (CNS) can overshadow the management of these diseases. Due to the chronic and progressive evolution of neurodegenerative disorders and since there is still no cure for them, new therapeutic strategies such as the combination of several drugs or the use of existing drugs with new therapeutic applications are valuable strategies. Tetracyclines are traditionally classified as antibiotics. However, in this class of drugs, doxycycline and minocycline exhibit also anti-inflammatory effects by inhibiting microglia/macrophages. Hence, they have been studied as potential agents for the treatment of neurodegenerative diseases. The results of in vitro and in vivo studies confirm the effective role of these two drugs as anti-inflammatory agents in experimentally induced models of neurodegenerative diseases. In clinical studies, satisfactory results have been obtained in Multiple sclerosis (MS) but not yet in other disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), or Amyotrophic lateral sclerosis (ALS). In recent years, researchers have developed and evaluated nanoparticulate drug delivery systems to improve the clinical efficacy of these two tetracyclines for their potential application in neurodegenerative diseases. This study reviews the neuroprotective roles of minocycline and doxycycline in four of the main neurodegenerative disorders: AD, PD, ALS and MS. Moreover, the potential applications of nanoparticulate delivery systems developed for both tetracyclines are also reviewed.
Collapse
|
15
|
Treatment of Transthyretin Amyloid Cardiomyopathy: The Current Options, the Future, and the Challenges. J Clin Med 2022; 11:jcm11082148. [PMID: 35456241 PMCID: PMC9031576 DOI: 10.3390/jcm11082148] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 12/13/2022] Open
Abstract
Transthyretin amyloid cardiomyopathy (ATTR-CM) is a progressively debilitating, rare disease associated with high mortality. ATTR-CM occurs when TTR amyloid protein builds up in the myocardium along with different organs, most commonly the peripheral and the autonomic nervous systems. Managing the cardiac complications with standard heart failure medications is difficult due to the challenge to maintain a balance between the high filling pressure associated with restricted ventricular volume and the low cardiac output. To date, tafamidis is the only agent approved for ATTR-CM treatment. Besides, several agents, including green tea, tolcapone, and diflunisal, are used off-label in ATTR-CM patients. Novel therapies using RNA interference also offer clinical promise. Patisiran and inotersen are currently approved for ATTR-polyneuropathy of hereditary origin and are under investigation for ATTR-CM. Monoclonal antibodies in the early development phases carry hope for amyloid deposit clearance. Despite several drug candidates in the clinical development pipeline, the small ATTR-CM patient population raises several challenges. This review describes current and future therapies for ATTR-CM and sheds light on the clinical development hurdles facing them.
Collapse
|
16
|
Liu W, Wang G, Wang Z, Wang G, Huang J, Liu B. Repurposing small-molecule drugs for modulating toxic protein aggregates in neurodegenerative diseases. Drug Discov Today 2022; 27:1994-2007. [PMID: 35395400 DOI: 10.1016/j.drudis.2022.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/18/2022] [Accepted: 04/01/2022] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases (NDs) are often age-related disorders that can cause dementia in people, usually over 65 years old, are still lacking effective therapies. Some NDs have recently been linked to toxic protein aggregates, for example Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and Huntington disease; therefore, mulating toxic protein aggregates would be a promising therapeutic strategy. Moreover, drug repurposing, in other words exploiting drugs that are already in use for another indication, has been attracting mounting attention for potential therapeutic purposes in NDs. Thus, in this review, we focus on summarizing a series of repurposed small-molecule drugs for eliminating or inhibiting toxic protein aggregates and further discuss their intricate molecular mechanisms to improve the current ND treatment. Taken together, these findings will shed new light on exploiting more repurposed small-molecule drugs targeting different types of toxic proteins to fight NDs in the future. Teaser: Drug repurposing has been gaining attention to yield therapeutic potential in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and Huntington disease.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Biotherapy and Cancer Center, and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Wang
- State Key Laboratory of Biotherapy and Cancer Center, and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiwen Wang
- State Key Laboratory of Biotherapy and Cancer Center, and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | | | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, and Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
17
|
Markulin I, Matasin M, Turk VE, Salković-Petrisic M. Challenges of repurposing tetracyclines for the treatment of Alzheimer's and Parkinson's disease. J Neural Transm (Vienna) 2022; 129:773-804. [PMID: 34982206 DOI: 10.1007/s00702-021-02457-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
The novel antibiotic-exploiting strategy in the treatment of Alzheimer's (AD) and Parkinson's (PD) disease has emerged as a potential breakthrough in the field. The research in animal AD/PD models provided evidence on the antiamyloidogenic, anti-inflammatory, antioxidant and antiapoptotic activity of tetracyclines, associated with cognitive improvement. The neuroprotective effects of minocycline and doxycycline in animals initiated investigation of their clinical efficacy in AD and PD patients which led to inconclusive results and additionally to insufficient safety data on a long-standing doxycycline and minocycline therapy in these patient populations. The safety issues should be considered in two levels; in AD/PD patients (particularly antibiotic-induced alteration of gut microbiota and its consequences), and as a world-wide threat of development of bacterial resistance to these antibiotics posed by a fact that AD and PD are widespread incurable diseases which require daily administered long-lasting antibiotic therapy. Recently proposed subantimicrobial doxycycline doses should be thoroughly explored for their effectiveness and long-term safety especially in AD/PD populations. Keeping in mind the antibacterial activity-related far-reaching undesirable effects both for the patients and globally, further work on repurposing these drugs for a long-standing therapy of AD/PD should consider the chemically modified tetracycline compounds tailored to lack antimicrobial but retain (or introduce) other activities effective against the AD/PD pathology. This strategy might reduce the risk of long-term therapy-related adverse effects (particularly gut-related ones) and development of bacterial resistance toward the tetracycline antibiotic agents but the therapeutic potential and desirable safety profile of such compounds in AD/PD patients need to be confirmed.
Collapse
Affiliation(s)
- Iva Markulin
- Community Health Centre Zagreb-Centre, Zagreb, Croatia
| | | | - Viktorija Erdeljic Turk
- Division of Clinical Pharmacology, Department of Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Melita Salković-Petrisic
- Department of Pharmacology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia.
| |
Collapse
|
18
|
Ferreira Junior NC, dos Santos Pereira M, Francis N, Ramirez P, Martorell P, González-Lizarraga F, Figadère B, Chehin R, Del Bel E, Raisman-Vozari R, Michel PP. The Chemically-Modified Tetracycline COL-3 and Its Parent Compound Doxycycline Prevent Microglial Inflammatory Responses by Reducing Glucose-Mediated Oxidative Stress. Cells 2021; 10:cells10082163. [PMID: 34440932 PMCID: PMC8392055 DOI: 10.3390/cells10082163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
We used mouse microglial cells in culture activated by lipopolysaccharide (LPS) or α-synuclein amyloid aggregates (αSa) to study the anti-inflammatory effects of COL-3, a tetracycline derivative without antimicrobial activity. Under LPS or αSa stimulation, COL-3 (10, 20 µM) efficiently repressed the induction of the microglial activation marker protein Iba-1 and the stimulated-release of the pro-inflammatory cytokine TNF-α. COL-3′s inhibitory effects on TNF-α were reproduced by the tetracycline antibiotic doxycycline (DOX; 50 µM), the glucocorticoid dexamethasone, and apocynin (APO), an inhibitor of the superoxide-producing enzyme NADPH oxidase. This last observation suggested that COL-3 and DOX might also operate themselves by restraining oxidative stress-mediated signaling events. Quantitative measurement of intracellular reactive oxygen species (ROS) levels revealed that COL-3 and DOX were indeed as effective as APO in reducing oxidative stress and TNF-α release in activated microglia. ROS inhibition with COL-3 or DOX occurred together with a reduction of microglial glucose accumulation and NADPH synthesis. This suggested that COL-3 and DOX might reduce microglial oxidative burst activity by limiting the glucose-dependent synthesis of NADPH, the requisite substrate for NADPH oxidase. Coherent with this possibility, the glycolysis inhibitor 2-deoxy-D-glucose reproduced the immunosuppressive action of COL-3 and DOX in activated microglia. Overall, we propose that COL-3 and its parent compound DOX exert anti-inflammatory effects in microglial cells by inhibiting glucose-dependent ROS production. These effects might be strengthened by the intrinsic antioxidant properties of DOX and COL-3 in a self-reinforcing manner.
Collapse
Affiliation(s)
- Nilson Carlos Ferreira Junior
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Maurício dos Santos Pereira
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Nour Francis
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paola Ramirez
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Paula Martorell
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
| | - Florencia González-Lizarraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Bruno Figadère
- BioCIS, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France;
| | - Rosana Chehin
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (CONICET-UNT-SIPROSA), CP 4000 Tucumán, Argentina; (F.G.-L.); (R.C.)
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Av. Café, s/no, Ribeirão Preto 14040-904, Brazil;
- USP, Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), São Paulo 05508-220, Brazil
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| | - Patrick Pierre Michel
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; (N.C.F.J.); (M.d.S.P.); (N.F.); (P.R.); (P.M.)
- Correspondence: (R.R.-V.); (P.P.M.); Tel.: +33-(0)157274550 (R.R.-V.); +33-(0)157274534 (P.P.M.)
| |
Collapse
|
19
|
González C, Cartagena C, Caballero L, Melo F, Areche C, Cornejo A. The Fumarprotocetraric Acid Inhibits Tau Covalently, Avoiding Cytotoxicity of Aggregates in Cells. Molecules 2021; 26:molecules26123760. [PMID: 34205516 PMCID: PMC8234475 DOI: 10.3390/molecules26123760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative disorders, including Tauopathies that involve tau protein, base their pathological mechanism on forming proteinaceous aggregates, which has a deleterious effect on cells triggering an inflammatory response. Moreover, tau inhibitors can exert their mechanism of action through noncovalent and covalent interactions. Thus, Michael's addition appears as a feasible type of interaction involving an α, β unsaturated carbonyl moiety to avoid pathological confirmation and further cytotoxicity. Moreover, we isolated three compounds from Antarctic lichens Cladonia cariosa and Himantormia lugubris: protolichesterinic acid (1), fumarprotocetraric acid (2), and lichesterinic acid (3). The maleimide cysteine labeling assay showed that compounds 1, 2, and 3 inhibit at 50 µM, but compounds 2 and 3 are statistically significant. Based on its inhibition capacity, we decided to test compound 2 further. Thus, our results suggest that compound 2 remodel soluble oligomers and diminish β sheet content, as demonstrated through ThT experiments. Hence, we added externally treated oligomers with compound 2 to demonstrate that they are harmless in cell culture. First, the morphology of cells in the presence of aggregates does not suffer evident changes compared to the control. Additionally, the externally added aggregates do not provoke a substantial LDH release compared to the control, indicating that treated oligomers do not provoke membrane damage in cell culture compared with aggregates alone. Thus, in the present work, we demonstrated that Michael's acceptors found in lichens could serve as a scaffold to explore different mechanisms of action to turn tau aggregates into harmless species.
Collapse
Affiliation(s)
- Camila González
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Andres Bello, Santiago 8370071, Chile; (C.G.); (C.C.)
| | - Constanza Cartagena
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Andres Bello, Santiago 8370071, Chile; (C.G.); (C.C.)
| | - Leonardo Caballero
- Departamento de Física, Center for Soft Matter Research, SMAT-C, Usach, Avenida Ecuador, Estación Central, Santiago 9170124, Chile; (L.C.); (F.M.)
| | - Francisco Melo
- Departamento de Física, Center for Soft Matter Research, SMAT-C, Usach, Avenida Ecuador, Estación Central, Santiago 9170124, Chile; (L.C.); (F.M.)
| | - Carlos Areche
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Ñuñoa, Santiago 7800003, Chile;
| | - Alberto Cornejo
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Andres Bello, Santiago 8370071, Chile; (C.G.); (C.C.)
- Correspondence:
| |
Collapse
|