1
|
Peyvand P, Allami P, Rezaei N. From genetic roots to recent advancements in gene therapy targeting amyloid beta in Alzheimer's disease. Rev Neurosci 2025:revneuro-2025-0025. [PMID: 40448320 DOI: 10.1515/revneuro-2025-0025] [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: 02/15/2025] [Accepted: 05/10/2025] [Indexed: 06/02/2025]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders. The pathological hallmarks of AD are amyloid-beta (Aβ) plaques and tau protein tangles, which cause neurodegeneration and lead to cognitive decline. The distinguished role of Aβ plaques in the onset of the disease, especially in familial AD, alongside the genetic complexity of AD, underscores the need for precise and targeted genetic interventions targeting Aβ. This review first highlights the amyloidogenic and non-amyloidogenic pathways and inflammatory mechanisms contributing to Aβ accumulation. It also introduces the role of genetic variants such as amyloid precursor protein (APP), presenilin (PSEN1), PSEN2, and Apolipoprotein E (APOE) alongside the molecular and cellular mechanisms involved in Aβ pathology. Then, gene therapy techniques are discussed for their potential to target Aβ either directly by inhibiting its production or enhancing its degradation or indirectly by targeting APOE, inflammatory pathways, and neurotrophic factors. While these approaches show significant preclinical promise, challenges such as timing, safety, and delivery across the blood-brain barrier persist and need further investigation.
Collapse
Affiliation(s)
- Pinar Peyvand
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran, 1416634793, Iran
| | - Pantea Allami
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- School of Medicine, 48439 Tehran University of Medical Sciences , Pour Sina St, Tehran, 1416634793, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Italy St Cross, St, Vesal Shirazi, Tehran, 1417755331, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
| |
Collapse
|
2
|
Mitrovic M, Selakovic D, Jovicic N, Ljujic B, Rosic G. BDNF/proBDNF Interplay in the Mediation of Neuronal Apoptotic Mechanisms in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:4926. [PMID: 40430064 PMCID: PMC12112594 DOI: 10.3390/ijms26104926] [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: 04/19/2025] [Revised: 05/12/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
The neurotrophic system includes neurotrophins, such as brain-derived neurotrophic factor (BDNF) and its precursor proBDNF, which play conflicting roles in neuronal survival and apoptosis, with their balance having a significant impact on neurodegenerative outcomes. While BDNF is widely acknowledged as a potent neurotrophin that promotes neuronal survival and differentiation, its precursor, proBDNF, has the opposite effect, promoting apoptosis and neuronal death. This review highlights the new and unique aspects of BDNF/proBDNF interaction in the modulation of neuronal apoptotic pathways in neurodegenerative disorders. It systematically discusses the cross-talk in apoptotic signaling at the molecular level, whereby BDNF activates survival pathways such as PI3K/Akt and MAPK/ERK, whereas proBDNF activates p75NTR and sortilin to induce neuronal apoptosis via JNK, RhoA, NFkB, and Rac-GTPase pathways such as caspase activation and mitochondrial injury. Moreover, this review emphasizes the factors that affect the balance between proBDNF and BDNF levels within the context of neurodegeneration, including proteolytic processing, the expression of TrkB and p75NTR receptors, and extrinsic gene transcription regulators. Cellular injury, stress, or signaling pathway alterations can disrupt the balance of BDNF/proBDNF, which may be involved in apoptotic-related neurodegenerative diseases like Alzheimer's, Parkinson's, and Huntington's diseases. This review provides a comprehensive framework for targeting neurotrophin signaling in the development of innovative therapies for neuronal survival and managing apoptotic-related neurodegenerative disorders, addressing the mechanistic complexity and clinical feasibility of BDNF/proBDNF interaction.
Collapse
Affiliation(s)
- Marina Mitrovic
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| |
Collapse
|
3
|
Borkowska P, Kowalczyk M, Zielińska A, Poskrobko K, Rother MB, Paul-Samojedny M, Kowalski J. NGF regulates survival and differentiation of umbilical mesenchymal stem/stromal cells into GABAergic, dopaminergic and cholinergic lineages. Eur J Pharm Sci 2025; 208:107053. [PMID: 40010415 DOI: 10.1016/j.ejps.2025.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
Mesenchymal stem cells advantageous properties have led scientists to conduct trials on a range of medical conditions, including incurable neurodegenerative diseases. Wharton-Jelly derived mesenchymal stem cells, given their ease of collection, are frequently selected for these studies. This research aimed to investigate the effects of nerve growth factor (NGF) gene overexpression on the neural differentiation, survivability, and gene and protein expression of these cells. The level of gene expression was tested using the ddPCR method. Six umbilical cords from donors were collected, and three randomly chosen primary cultures of Wharton-Jelly derived mesenchymal stem cells were used in experiment. Cells were transduced with lentiviral vectors and underwent a 12-day differentiation process. The results revealed neuron-like cells with significantly high expression of CHAT, GAD2 and TH genes. A corresponding increase in protein expression was also observed. Immunostaining demonstrated notable differences in neuron-like phenotypes, contingent on the environmental conditions of the research groups. Throughout the experiment, samples with transduced mesenchymal stem cells overexpressing the NGF gene showed the highest expression levels from almost all of studied genes and proteins, and were also the most phenotypically similar to neuron-like cells. The study concluded that sustained overexpression of NGF: guides mesenchymal stem cells towards the neural pathway, facilitates the differentiation of modified mesenchymal stem cells into GABAergic, dopaminergic, and cholinergic neuron-like cells, suggests that GABAergic neurons' marker predominantly co-expresses with other neurons' markers, such as cholinergic or dopaminergic ones, increases survivability of modified mesenchymal stem cells in toxic conditions; The limitations of the study is that we merely know that cells have begun to express neurogenic markers, but in the absence of standards for mature neuronal markers, we do not yet know how far they have progressed as differentiating cells.
Collapse
Affiliation(s)
- Paulina Borkowska
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Małgorzata Kowalczyk
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Aleksandra Zielińska
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Karol Poskrobko
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| | - Magdalena B Rother
- Department of Human Genetics, Leiden University Medical Center (LUMC), 2300 RC, The Netherlands
| | | | - Jan Kowalski
- Department of Medical Genetics, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
4
|
Gu CL, Zhang L, Zhu Y, Bao TY, Zhu YT, Chen YT, Pang HQ. Exploring the cellular and molecular basis of nerve growth factor in cerebral ischemia recovery. Neuroscience 2025; 566:190-197. [PMID: 39742942 DOI: 10.1016/j.neuroscience.2024.12.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Vascular obstruction often causes inadequate oxygen and nutrient supply to the brain. This deficiency results in cerebral ischemic injury, which significantly impairs neurological function. This review aimed to explore the neuroprotective and regenerative effects of nerve growth factor (NGF) in cerebral ischemic injury. NGF, a crucial neurotrophic factor, could inhibit neuronal apoptosis, reduce inflammatory responses, and promote axon regeneration and angiogenesis through its interaction with TrkA, a high-affinity receptor. These functions were closely related to the activation of Phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) and Mitogen-Activated Protein Kinase (MAPK) pathways. Moreover, the mechanisms of NGF in the acute and recovery phases, along with the strategies to enhance its therapeutic effects using delivery systems (such as intranasal administration, nanovesicles, and gene therapy) were also summarized. Although NGF shows great potential for clinical application, its delivery efficiency and long-term safety still need more research and improvements. Future research should focus on exploring the specific action mechanism of NGF, optimizing the delivery strategy, and evaluating its long-term efficacy and safety to facilitate its clinical transformation in cerebral ischemic stroke.
Collapse
Affiliation(s)
- Chen-Lin Gu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Lu Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yan Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Ting-Yu Bao
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Ting Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Tong Chen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Han-Qing Pang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
5
|
Inamdar A, Gurupadayya B, Halagali P, Nandakumar S, Pathak R, Singh H, Sharma H. Cutting-edge Strategies for Overcoming Therapeutic Barriers in Alzheimer's Disease. Curr Pharm Des 2025; 31:598-618. [PMID: 39492772 DOI: 10.2174/0113816128344571241018154506] [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: 08/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) remains one of the hardest neurodegenerative diseases to treat due to its enduring cognitive deterioration and memory loss. Despite extensive research, few viable treatment approaches have been found; these are mostly due to several barriers, such as the disease's complex biology, limited pharmaceutical efficacy, and the BBB. This presentation discusses current strategies for addressing these therapeutic barriers to enhance AD treatment. Innovative drug delivery methods including liposomes, exosomes, and nanoparticles may be able to pass the blood-brain barrier and allow medicine to enter specific brain regions. These innovative strategies of medicine distribution reduce systemic side effects by improving absorption. Moreover, the development of disease-modifying treatments that target tau protein tangles, amyloid-beta plaques, and neuroinflammation offers the chance to influence the course of the illness rather than only treat its symptoms. Furthermore, gene therapy and CRISPR-Cas9 technologies have surfaced as potentially groundbreaking methods for addressing the underlying genetic defects associated with AD. Furthermore, novel approaches to patient care may involve the utilization of existing medications having neuroprotective properties, such as those for diabetes and cardiovascular conditions. Furthermore, biomarker research and personalized medicine have made individualized therapy approaches possible, ensuring that patients receive the best care possible based on their unique genetic and molecular profiles.
Collapse
Affiliation(s)
- Aparna Inamdar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Bannimath Gurupadayya
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Prashant Halagali
- Department of Pharmaceutical Quality Assurance, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi 590010, Karnataka, India
| | - S Nandakumar
- Associate Scientist, Corteva Agriscience, Hyderabad 500081, Telangana, India
| | - Rashmi Pathak
- Department of Pharmacy, Invertis University, Bareilly (UP) 243123, India
| | - Himalaya Singh
- Department of Medicine, Government Institute of Medical Sciences, Greater Noida (UP) 201312, India
| | - Himanshu Sharma
- Department of Pharmacy, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad (UP) 244001, India
| |
Collapse
|
6
|
Șovrea AS, Boșca AB, Dronca E, Constantin AM, Crintea A, Suflețel R, Ștefan RA, Ștefan PA, Onofrei MM, Tschall C, Crivii CB. Non-Drug and Non-Invasive Therapeutic Options in Alzheimer's Disease. Biomedicines 2025; 13:84. [PMID: 39857667 PMCID: PMC11760896 DOI: 10.3390/biomedicines13010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Despite the massive efforts of modern medicine to stop the evolution of Alzheimer's disease (AD), it affects an increasing number of people, changing individual lives and imposing itself as a burden on families and the health systems. Considering that the vast majority of conventional drug therapies did not lead to the expected results, this review will discuss the newly developing therapies as an alternative in the effort to stop or slow AD. Focused Ultrasound (FUS) and its derived Transcranial Pulse Stimulation (TPS) are non-invasive therapeutic approaches. Singly or as an applied technique to change the permeability of the blood-brain-barrier (BBB), FUS and TPS have demonstrated the benefits of use in treating AD in animal and human studies. Adipose-derived stem Cells (ADSCs), gene therapy, and many other alternative methods (diet, sleep pattern, physical exercise, nanoparticle delivery) are also new potential treatments since multimodal approaches represent the modern trend in this disorder research therapies.
Collapse
Affiliation(s)
- Alina Simona Șovrea
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Adina Bianca Boșca
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Eleonora Dronca
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Anne-Marie Constantin
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Andreea Crintea
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Rada Suflețel
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Roxana Adelina Ștefan
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Paul Andrei Ștefan
- Radiology and Imaging Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania;
| | - Mădălin Mihai Onofrei
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Christoph Tschall
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Carmen-Bianca Crivii
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| |
Collapse
|
7
|
Ortega A, Chernicki B, Ou G, Parmar MS. From Lab Bench to Hope: Emerging Gene Therapies in Clinical Trials for Alzheimer's Disease. Mol Neurobiol 2025; 62:1112-1135. [PMID: 38958888 DOI: 10.1007/s12035-024-04285-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder that affects memory and cognitive abilities, affecting millions of people around the world. Current treatments focus on the management of symptoms, as no effective therapy has been approved to modify the underlying disease process. Gene therapy is a promising approach that can offer disease-modifying treatment for AD, targeting various aspects of the pathophysiology of the disease. This review presents a comprehensive overview of the current state of gene therapy research for AD, with a specific focus on clinical trials and preclinical studies that have used nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), apolipoprotein E2 (APOE2), and human telomerase reverse transcriptase (hTERT) as therapeutic gene therapy approaches. These gene targets have shown potential to alleviate the neuropathology of AD in animal studies and have demonstrated feasibility and safety in non-human primates. Despite the failure of the NGF gene therapy approach in clinical trials, we have reviewed and highlighted the reported findings and evaluations from the trials. Furthermore, the review included the conclusions of postmortem brain tissue analysis of AD patients who received NGF gene therapy. The goal is to learn from the failed trials and improve the approach in the future. Although gene therapy shows promise, it faces several challenges and limitations, including optimizing gene delivery methods, enhancing safety and efficacy profiles, and determining long-term results. This review contributes to the growing body of literature on innovative treatments for AD and highlights the need for more research and development to advance gene therapy as a viable treatment option for AD.
Collapse
Affiliation(s)
- Angelica Ortega
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Brendan Chernicki
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Grace Ou
- College of Arts and Sciences, Cornell University, Ithaca, NY, USA
| | - Mayur S Parmar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
- Department of Foundational Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
| |
Collapse
|
8
|
Mufson EJ, Perez SE. The cholinotrophic system in Down syndrome. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:185-213. [PMID: 40340061 DOI: 10.1016/b978-0-443-19088-9.00017-2] [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: 05/10/2025]
Abstract
Cholinergic basal forebrain (CBF) projection neurons within the nucleus basalis and striatal cholinergic interneurons degenerate in individuals with Down syndrome (DS). However, the neuropathobiology of these diverse cholinergic phenotypes remains underinvestigated. This review summarizes the alterations of cholinergic, neurotrophic survival and cell death factors as well as tau pathology and amyloidopathy, and their effects upon these cell types in DS. In trisomy, the developing cholinergic system remains stable, whereas the neurotrophic receptors are compromised between control and DS cases. Both cholinergic neuronal phenotypes display severe cellular degeneration in both adult and the aged people with DS. Although developing cholinergic striatal neurons display a similar morphology between phenotypes, cholinergic striatal neurons appear dystrophic in adults with DS. Both cholinergic cell types display tau tangle pathology in elders with DS. Novel findings suggest that alterations in plasma and cerebral spinal fluid levels of proNGF, NGF metabolites, and select classes of neuronal genes are potential biomarkers to distinguish nondemented from demented people with DS. Compounds that target cholinergic pathways, TrkA agonists, p75NTR/proNGF small molecular antagonists, NGF metabolites, and select gene ontology classes are potential targets to slow degeneration of the CBF memory connectome in DS with translation to AD.
Collapse
Affiliation(s)
- Elliott J Mufson
- Department of Translational Neuroscience, St. Joseph's Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, United States; Department of Neurology, St. Joseph's Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, United States.
| | - Sylvia E Perez
- Department of Translational Neuroscience, St. Joseph's Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
9
|
Mitra S, Gera R, Eriksdotter M. NGF-based cholinergic therapies in Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:123-135. [PMID: 40340057 DOI: 10.1016/b978-0-443-19088-9.00007-x] [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: 05/10/2025]
Abstract
The cholinergic system is part of the parasympathetic nervous system, which works in tandem with the sympathetic and enteric nervous systems to maintain the physiologic functioning of our body. The neurotransmitter acetylcholine (ACh) facilitates cholinergic signaling pathways by activating specific cell surface receptors (nicotinic and muscarinic receptors). Altered cholinergic signaling has been implicated in several pathologic conditions. In this chapter, conditions where cholinergic modulation in the central nervous system occurs through the neurotrophin nerve growth factor (NGF) are addressed. NGF is the master regulator of several pathways, ultimately leading to cell survival, ACh production, regenerative signaling, and anti-inflammatory tone. NGF and cholinergic-related pathways have been reported to be severely affected in the case of Alzheimer disease (AD), the most common dementia disorder. In AD, the cholinergic nuclei of the basal forebrain are affected early during the AD continuum, resulting in cholinergic cell loss and hampered ACh production, which overall affects the propagation of cholinergic signals in other brain regions. Since the 1990s clinically relevant strategies to treat AD patients have been the drugs that enhance cholinergic signaling-termed cholinesterase inhibitors (ChEIs), however, other strategies in AD have been and are presently being assessed for clinical efficacy. Delivery of NGF to the basal forebrain is considered crucial to revive the cholinergic cell bodies, restore ACh production, and sustain cognitive function. This chapter provides a description of the relevance of NGF-based therapies targeted for AD treatment, technical approaches for NGF delivery to the brain, and the status of ongoing clinical studies are provided.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ruchi Gera
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
10
|
Juengling F, Wuest F, Schirrmacher R, Abele J, Thiel A, Soucy JP, Camicioli R, Garibotto V. PET Imaging in Dementia: Mini-Review and Canadian Perspective for Clinical Use. Can J Neurol Sci 2025; 52:26-38. [PMID: 38433571 DOI: 10.1017/cjn.2024.31] [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: 03/05/2024]
Abstract
PET imaging is increasingly recognized as an important diagnostic tool to investigate patients with cognitive disturbances of possible neurodegenerative origin. PET with 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), assessing glucose metabolism, provides a measure of neurodegeneration and allows a precise differential diagnosis among the most common neurodegenerative diseases, such as Alzheimer's disease, frontotemporal dementia or dementia with Lewy bodies. PET tracers specific for the pathological deposits characteristic of different neurodegenerative processes, namely amyloid and tau deposits typical of Alzheimer's Disease, allow the visualization of these aggregates in vivo. [18F]FDG and amyloid PET imaging have reached a high level of clinical validity and are since 2022 investigations that can be offered to patients in standard clinical care in most of Canada.This article will briefly review and summarize the current knowledge on these diagnostic tools, their integration into diagnostic algorithms as well as perspectives for future developments.
Collapse
Affiliation(s)
- Freimut Juengling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Faculty, University of Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ralf Schirrmacher
- Division of Oncologic Imaging and Radionuclide Therapy, Cross Cancer Institute, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Jonathan Abele
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Department of Neurology and Neurosurgery, Lady Davis Institute for Medical Research, McGill University, Montréal, QC, Canada
| | - Jean-Paul Soucy
- Montréal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Richard Camicioli
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valentina Garibotto
- Diagnostic Department, Nuclear Medicine and Molecular Imaging Division, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
11
|
Day IL, Tamboline M, Lipshutz GS, Xu S. Recent developments in translational imaging of in vivo gene therapy outcomes. Mol Ther 2024:S1525-0016(24)00849-9. [PMID: 39741403 DOI: 10.1016/j.ymthe.2024.12.049] [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: 07/31/2024] [Revised: 11/18/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Gene therapy achieves therapeutic benefits by delivering genetic materials, packaged within a delivery vehicle, to target cells with defective genes. This approach has shown promise in treating various conditions, including cancer, metabolic disorders, and tissue-degenerative diseases. Over the past 5 years, molecular imaging has increasingly supported gene therapy development in both preclinical and clinical studies. High-quality images from positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and computed tomography (CT) enable quantitative and reliable monitoring of gene therapy. Most reported studies have applied imaging biomarkers to non-invasively evaluate the outcomes of gene therapy. This review aims to inform researchers in molecular imaging and gene therapy about the integration of these two disciplines. We highlight recent developments in using imaging biomarkers to monitor the outcome of in vivo gene therapy, where the therapeutic delivery vehicle is administered systemically. In addition, we discuss prospects for further incorporating imaging biomarkers to support the development and application of gene therapy.
Collapse
Affiliation(s)
- Isabel L Day
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mikayla Tamboline
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shili Xu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
12
|
Zhao H, Li H, Meng L, Du P, Mo X, Gong M, Chen J, Liao Y. Disrupting heroin-associated memory reconsolidation through actin polymerization inhibition in the nucleus accumbens core. Int J Neuropsychopharmacol 2024; 28:pyae065. [PMID: 39716383 DOI: 10.1093/ijnp/pyae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Understanding drug addiction as a disorder of maladaptive learning, where drug-associated or environmental cues trigger drug cravings and seeking, is crucial for developing effective treatments. Actin polymerization, a biochemical process, plays a crucial role in drug-related memory formation, particularly evident in conditioned place preference paradigms involving drugs like morphine and methamphetamine. However, the role of actin polymerization in the reconsolidation of heroin-associated memories remains understudied. METHODS This study employed a rodent model of self-administered heroin to investigate the involvement of actin polymerization in the reconsolidation of heroin-associated memories. Rats underwent ten days of intravenous heroin self-administration paired with conditioned cues. Subsequently, a 10-day extinction phase aimed to reduce heroin-seeking behaviors. Following this, rats participated in a 15-minute retrieval trial with or without cues. Immediately post-retrieval, rats received bilateral injections of the actin polymerization inhibitor Latrunculin A (Lat A) into the nucleus accumbens core (NACc), a critical brain region for memory reconsolidation. RESULTS Immediate administration of Lat A into the NACc post-retrieval significantly reduced cue-induced and heroin-primed reinstatement of heroin-seeking behavior for at least 28 days. However, administering Lat A 6-hour post-retrieval or without a retrieval trial, as well as administering Jasplakionlide prior to memory reactivation did not affect heroin-seeking behaviors. CONCLUSIONS Inhibiting actin polymerization during the reconsolidation window disrupts heroin-associated memory reconsolidation, leading to decreased heroin-seeking behavior and prevention of relapse. These effects are contingent upon the presence of a retrieval trial and exhibit temporal specificity, shedding light on addiction mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Haiting Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyu Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Meng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Du
- Department of Neurosurgery, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Xin Mo
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengqi Gong
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaxin Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiwei Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Ozgür-Gunes Y, Le Stunff C, Bougnères P. Oligodendrocytes, the Forgotten Target of Gene Therapy. Cells 2024; 13:1973. [PMID: 39682723 PMCID: PMC11640421 DOI: 10.3390/cells13231973] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
If the billions of oligodendrocytes (OLs) populating the central nervous system (CNS) of patients could express their feelings, they would undoubtedly tell gene therapists about their frustration with the other neural cell populations, neurons, microglia, or astrocytes, which have been the favorite targets of gene transfer experiments. This review questions why OLs have been left out of most gene therapy attempts. The first explanation is that the pathogenic role of OLs is still discussed in most CNS diseases. Another reason is that the so-called ubiquitous CAG, CBA, CBh, or CMV promoters-widely used in gene therapy studies-are unable or poorly able to activate the transcription of episomal transgene copies brought by adeno-associated virus (AAV) vectors in OLs. Accordingly, transgene expression in OLs has either not been found or not been evaluated in most gene therapy studies in rodents or non-human primates. The aims of the current review are to give OLs their rightful place among the neural cells that future gene therapy could target and to encourage researchers to test the effect of OL transduction in various CNS diseases.
Collapse
Affiliation(s)
- Yasemin Ozgür-Gunes
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA;
| | - Catherine Le Stunff
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l’Energie Atomique, 92260 Fontenay-aux-Roses, France;
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm and University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Pierre Bougnères
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l’Energie Atomique, 92260 Fontenay-aux-Roses, France;
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| |
Collapse
|
14
|
Mansoor S, Jindal A, Badu NYA, Katiki C, Ponnapalli VJS, Desai KJ, Nassar ST. Role of Neurotrophins in the Development and Treatment of Neurodegenerative Diseases: A Systematic Review. Cureus 2024; 16:e74048. [PMID: 39712854 PMCID: PMC11660190 DOI: 10.7759/cureus.74048] [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] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
A considerable amount of morbidity and disability are caused by a wide variety of neurological illnesses together referred to as neurodegenerative diseases. Among them, Alzheimer's and Parkinson's diseases are the most prevalent and have been thoroughly studied. The development of intervention techniques that focus on the unfavorable elements of these diseases, particularly those that could help halt their course, has become increasingly important. This study aims to explain the most current findings about the function of neurotrophins, the signaling pathways they follow in neurodegenerative illnesses, and their possible therapeutic applications. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 criteria served as the foundation for this systematic review. In April 2024, a thorough search was conducted through the Cochrane Library, Google Scholar, PubMed, PubMed Central, and ScienceDirect databases. The predetermined criteria used to choose the research were the English language, narrative and systematic reviews, observational studies, and randomized and non-randomized clinical trials published within the last ten years. Subsequently, each study type-specific quality assessment was conducted utilizing the available assessment method. Of the 3,322 studies found during the first search, 15 were ultimately chosen for inclusion in the final selection. One cohort, one non-randomized clinical trial, one randomized clinical trial, three meta-analyses and systematic reviews, and nine narrative reviews were included. This review has explained in detail the current understanding of how neurotrophins play an essential role in neuroplasticity and neurogenesis, as well as their complex downstream signaling that leads to the process of neurodegeneration. Our study has also highlighted previous studies showing the efficacy of neurotrophins in clinical trials, but the data is limited; more preclinical and clinical studies are needed in this regard. These have also drawn our attention to future clinical trials that will address the challenges faced in their delivery and associated complications. Altogether, neurotrophins could serve as promising targets for therapeutic intervention that could stop or even reverse the development of neuropathology associated with neurodegenerative illnesses.
Collapse
Affiliation(s)
- Sadia Mansoor
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | | | | | - Chiko Katiki
- Emergency Medicine, American International Medical School, Santa Rosa, USA
| | | | | | - Sondos T Nassar
- Medicine and Surgery, Jordan University of Science and Technology, Amman, JOR
| |
Collapse
|
15
|
Svendsen SP, Svendsen CN. Cell therapy for neurological disorders. Nat Med 2024; 30:2756-2770. [PMID: 39407034 DOI: 10.1038/s41591-024-03281-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/30/2024] [Indexed: 10/18/2024]
Abstract
Cell therapies for neurological disorders are entering the clinic and present unique challenges and opportunities compared with conventional medicines. They have the potential to replace damaged nervous tissue and integrate into the brain or spinal cord to produce functional effects for the lifetime of the patient, which could revolutionize the way clinicians treat debilitating neurological disorders. The major challenge has been cell sourcing, which historically relied mainly on fetal brain tissue. This has largely been overcome with the advent of pluripotent stem cell technology and the ability to make almost any cell of the nervous system at scale. Furthermore, advances in gene editing now allow the generation of genetically modified cells that could perform better and evade the immune system. With all the remarkable new approaches to treat neurological disorders, we take a critical look at the state of current clinical trials and how challenges may be overcome with the evolving technology and innovation occurring in the stem cell field.
Collapse
Affiliation(s)
- Soshana P Svendsen
- Cedars-Sinai Board of Governors Regenerative Medicine Institute, Los Angeles, CA, USA
| | - Clive N Svendsen
- Cedars-Sinai Board of Governors Regenerative Medicine Institute, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Ebrahimi P, Davoudi E, Sadeghian R, Zadeh AZ, Razmi E, Heidari R, Morowvat MH, Sadeghian I. In vivo and ex vivo gene therapy for neurodegenerative diseases: a promise for disease modification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7501-7530. [PMID: 38775852 DOI: 10.1007/s00210-024-03141-4] [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] [Received: 01/17/2024] [Accepted: 05/01/2024] [Indexed: 10/04/2024]
Abstract
Neurodegenerative diseases (NDDs), including AD, PD, HD, and ALS, represent a growing public health concern linked to aging and lifestyle factors, characterized by progressive nervous system damage leading to motor and cognitive deficits. Current therapeutics offer only symptomatic management, highlighting the urgent need for disease-modifying treatments. Gene therapy has emerged as a promising approach, targeting the underlying pathology of diseases with diverse strategies including gene replacement, gene silencing, and gene editing. This innovative therapeutic approach involves introducing functional genetic material to combat disease mechanisms, potentially offering long-term efficacy and disease modification. With advancements in genomics, structural biology, and gene editing tools such as CRISPR/Cas9, gene therapy holds significant promise for addressing the root causes of NDDs. Significant progress in preclinical and clinical studies has demonstrated the potential of in vivo and ex vivo gene therapy to treat various NDDs, offering a versatile and precise approach in comparison to conventional treatments. The current review describes various gene therapy approaches employed in preclinical and clinical studies for the treatment of NDDs, including AD, PD, HD, and ALS, and addresses some of the key translational challenges in this therapeutic approach.
Collapse
Affiliation(s)
- Pouya Ebrahimi
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Davoudi
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | | | - Amin Zaki Zadeh
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Emran Razmi
- Arak University of Medical Sciences, Arak, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Morowvat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
17
|
Magrì A, Tomasello B, Naletova I, Tabbì G, Cairns WRL, Greco V, Sciuto S, La Mendola D, Rizzarelli E. New BDNF and NT-3 Cyclic Mimetics Concur with Copper to Activate Trophic Signaling Pathways as Potential Molecular Entities to Protect Old Brains from Neurodegeneration. Biomolecules 2024; 14:1104. [PMID: 39334869 PMCID: PMC11430436 DOI: 10.3390/biom14091104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
A low level of Neurotrophins (NTs), their Tyrosine Kinase Receptors (Trks), Vascular Endothelial Growth Factors (VEGFs) and their receptors, mainly VEGFR1 and VEGFR2, characterizes AD brains. The use of NTs and VEGFs as drugs presents different issues due to their low permeability of the blood-brain barrier, the poor pharmacokinetic profile, and the relevant side effects. To overcome these issues, different functional and structural NT mimics have been employed. Being aware that the N-terminus domain as the key domain of NTs for the binding selectivity and activation of Trks and the need to avoid or delay proteolysis, we herein report on the mimicking ability of two cyclic peptide encompassing the N-terminus of Brain Derived Growth Factor (BDNF), (c-[HSDPARRGELSV-]), cBDNF(1-12) and of Neurotrophin3 (NT3), (c-[YAEHKSHRGEYSV-]), cNT3(1-13). The two cyclic peptide features were characterized by a combined thermodynamic and spectroscopic approach (potentiometry, NMR, UV-vis and CD) that was extended to their copper(II) ion complexes. SH-SY5Y cell assays show that the Cu2+ present at the sub-micromolar level in the complete culture media affects the treatments with the two peptides. cBDNF(1-12) and cNT3(1-13) act as ionophores, induce neuronal differentiation and promote Trks and CREB phosphorylation in a copper dependent manner. Consistently, both peptide and Cu2+ stimulate BDNF and VEGF expression as well as VEGF release; cBDNF(1-12) and cNT3(1-13) induce the expression of Trks and VEGFRs.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Irina Naletova
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Giovanni Tabbì
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
| | - Warren R. L. Cairns
- CNR-Institute of Polar Sciences (CNR-ISP), 155 Via Torino, 30172 Venice, Italy;
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy; (A.M.); (I.N.); (G.T.)
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (V.G.); (S.S.)
| |
Collapse
|
18
|
Issa S, Fayoud H, Shaimardanova A, Sufianov A, Sufianova G, Solovyeva V, Rizvanov A. Growth Factors and Their Application in the Therapy of Hereditary Neurodegenerative Diseases. Biomedicines 2024; 12:1906. [PMID: 39200370 PMCID: PMC11351319 DOI: 10.3390/biomedicines12081906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Hereditary neurodegenerative diseases (hNDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and others are primarily characterized by their progressive nature, severely compromising both the cognitive and motor abilities of patients. The underlying genetic component in hNDDs contributes to disease risk, creating a complex genetic landscape. Considering the fact that growth factors play crucial roles in regulating cellular processes, such as proliferation, differentiation, and survival, they could have therapeutic potential for hNDDs, provided appropriate dosing and safe delivery approaches are ensured. This article presents a detailed overview of growth factors, and explores their therapeutic potential in treating hNDDs, emphasizing their roles in neuronal survival, growth, and synaptic plasticity. However, challenges such as proper dosing, delivery methods, and patient variability can hinder their clinical application.
Collapse
Affiliation(s)
- Shaza Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (S.I.); (H.F.)
| | - Haidar Fayoud
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (S.I.); (H.F.)
| | - Alisa Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
| | - Albert Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia;
- The Research and Educational Institute of Neurosurgery, Peoples’ Friendship University of Russia (RUDN), 117198 Moscow, Russia
| | - Galina Sufianova
- Department of Pharmacology, Tyumen State Medical University, 625023 Tyumen, Russia;
| | - Valeriya Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| |
Collapse
|
19
|
Roberts WS, Price S, Wu M, Parmar MS. Emerging Gene Therapies for Alzheimer's and Parkinson's Diseases: An Overview of Clinical Trials and Promising Candidates. Cureus 2024; 16:e67037. [PMID: 39286667 PMCID: PMC11405083 DOI: 10.7759/cureus.67037] [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: 06/24/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Gene therapy as a disease-modifying therapeutic approach for neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), is a promising avenue. Promising results in the preclinical studies involving rodents and nonhuman primates utilizing gene therapy have led to multiple clinical trials evaluating various genes of interest for AD and PD. In AD, clinical trials are assessing gene therapy involving brain-derived neurotrophic factor (BDNF) and other targets such as apolipoprotein E2 (APOE2) and human telomerase reverse transcriptase (hTERT). In PD, clinical trials are evaluating gene therapy delivering neurotrophic factors, such as glial cell line-derived neurotrophic factor (GDNF). Additionally, gene therapy delivering enzymes aromatic L-amino acid decarboxylase (AADC) and glutamic acid decarboxylase (GAD) are also being evaluated for PD. All these trials primarily utilized adeno-associated virus (AAV) to deliver the above transgene of interest. This review summarizes the current clinical trials involving gene therapy for AD and PD. It also discusses the challenges and opportunities associated with the gene therapy approach in AD and PD and ongoing developments related to increasing the safety and efficacy of the gene therapy for long-term outcomes, which include evaluation of various serotypes and administration routes. This comprehensive review emphasizes translating preclinical findings into clinical trials, further directions, and the potential for this promising therapeutic approach to alleviate neurodegenerative disease.
Collapse
Affiliation(s)
- Will S Roberts
- Osteopathic Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine Nova Southeastern University, Clearwater, USA
| | - Shawn Price
- Osteopathic Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine Nova Southeastern University, Clearwater, USA
| | - Michael Wu
- Osteopathic Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine Nova Southeastern University, Clearwater, USA
| | - Mayur S Parmar
- Foundational Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine Nova Southeastern University, Clearwater, USA
| |
Collapse
|
20
|
Doshi V, Joshi G, Sharma S, Choudhary D. Gene therapy: an alternative to treat Alzheimer's disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3675-3693. [PMID: 38078920 DOI: 10.1007/s00210-023-02873-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/22/2023] [Indexed: 05/23/2024]
Abstract
Alzheimer's disease (AD), a neuro-degenerative disease that primarily affects the elderly, is a worldwide phenomenon. Loss of memory, cognitive decline, behavioural changes, and many other signs are used to classify it. Various hypotheses that may contribute to Alzheimer's disease have been found during decades of survey, including tau theory, the amyloid theory, the cholinergic hypothesis, and the oxidative stress hypothesis. According to some theories, the two leading causes of AD are the accumulation of amyloid beta plaque and development of NFTs in the brain. The hippocampus and cerebral cortex are the primary sites where amyloid beta plaques gather in the body. NFT formation in the brain impairs the brain's neurons' potential of signalling. According to the age at which it manifests in a person, there are two subtypes of AD: 'LOAD (Late Onset Alzheimer's Disease)' and 'EOAD (Early Onset Alzheimer's Disease)'. Long-term research into AD treatment has resulted in the introduction of some medications that provided symptomatic relief to patients but did not alter the disease's pathophysiology, like cholinesterase inhibitors, inhibitors of tau aggregation, and monoclonal antibodies to Aβ aggregation. Even though the medications did not halt the progression of AD, researchers did not discontinue their work, which lead to the introduction of gene therapy - a recently created cutting-edge method of delivering genes to target sites where they can express the intended functionalities. Viral or non-viral vectors could be used to deliver the gene, each with advantages and limitations of their own. Gene therapy is proven to be a potential disease-modifying treatment for AD. This article discusses about gene therapy, its merits and demerits and the various ways of gene delivery. Additionally, it focuses on AD as the target for treatment through gene therapy, the pathophysiology of AD, and the multiple targets for gene therapy in the treatment of AD.
Collapse
Affiliation(s)
- Vanshika Doshi
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, Maharashtra, 400056, India
| | - Garima Joshi
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan, 313001, India
| | - Sanjay Sharma
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, Maharashtra, 400056, India.
| | - Deepak Choudhary
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan, 313001, India.
| |
Collapse
|
21
|
Song M, Kang K, Wang S, Zhang C, Zhao X, Song F. Elevated intracellular Ca 2+ functions downstream of mitodysfunction to induce Wallerian-like degeneration and necroptosis in organophosphorus-induced delayed neuropathy. Toxicology 2024; 504:153812. [PMID: 38653376 DOI: 10.1016/j.tox.2024.153812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/06/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Neurotoxic organophosphorus compounds can induce a type of delayed neuropathy in humans and sensitive animals, known as organophosphorus-induced delayed neuropathy (OPIDN). OPIDN is characterized by axonal degeneration akin to Wallerian-like degeneration, which is thought to be caused by increased intra-axonal Ca2+ concentrations. This study was designed to investigate that deregulated cytosolic Ca2+ may function downstream of mitodysfunction in activating Wallerian-like degeneration and necroptosis in OPIDN. Adult hens were administrated a single dosage of 750 mg/kg tri-ortho-cresyl phosphate (TOCP), and then sacrificed at 1 day, 5 day, 10 day and 21 day post-exposure, respectively. Sciatic nerves and spinal cords were examined for pathological changes and proteins expression related to Wallerian-like degeneration and necroptosis. In vitro experiments using differentiated neuro-2a (N2a) cells were conducted to investigate the relationship among mitochondrial dysfunction, Ca2+ influx, axonal degeneration, and necroptosis. The cells were co-administered with the Ca2+-chelator BAPTA-AM, the TRPA1 channel inhibitor HC030031, the RIPK1 inhibitor Necrostatin-1, and the mitochondrial-targeted antioxidant MitoQ along with TOCP. Results demonstrated an increase in cytosolic calcium concentration and key proteins associated with Wallerian degeneration and necroptosis in both in vivo and in vitro models after TOCP exposure. Moreover, co-administration with BATPA-AM or HC030031 significantly attenuated the loss of NMNAT2 and STMN2 in N2a cells, as well as the upregulation of SARM1, RIPK1 and p-MLKL. In contrast, Necrostatin-1 treatment only inhibited the TOCP-induced elevation of p-MLKL. Notably, pharmacological protection of mitochondrial function with MitoQ effectively alleviated the increase in intracellular Ca2+ following TOCP and mitigated axonal degeneration and necroptosis in N2a cells, supporting mitochondrial dysfunction as an upstream event of the intracellular Ca2+ imbalance and neuronal damage in OPIDN. These findings suggest that mitochondrial dysfunction post-TOCP intoxication leads to an elevated intracellular Ca2+ concentration, which plays a pivotal role in the initiation and development of OPIDN through inducing SARM1-mediated axonal degeneration and activating the necroptotic signaling pathway.
Collapse
Affiliation(s)
- Mingxue Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Kang Kang
- Qingdao Municipal Center for Disease Control & Prevention, Qingdao, Shandong 266033, PR China
| | - Shuai Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Cuiqin Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Xiulan Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
22
|
Suresh S, Vellapandian C. Cyanidin Ameliorates Bisphenol A-Induced Alzheimer's Disease Pathology by Restoring Wnt/β-Catenin Signaling Cascade: an In Vitro Study. Mol Neurobiol 2024; 61:2064-2080. [PMID: 37843801 DOI: 10.1007/s12035-023-03672-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/22/2023] [Indexed: 10/17/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder causing memory loss and cognitive decline, linked to amyloid-beta (Aβ) plaques and hyperphosphorylated tau protein accumulation in the brain. Environmental pollutant bisphenol A (BPA) has been implicated in AD pathology due to its neurotoxic effects. This study aims to evaluate cyanidin from flower bracts of Musa acuminata Colla (red variety; AAA group) for its neuroprotective properties against BPA-induced AD pathology. The extraction of cyanidin was optimized using 70% ethanol in acidified water, showing promising anti-acetylcholinesterase activity. Cyanidin was effectively purified from the resultant extract and characterized using spectroscopic techniques. Two gradient doses of cyanidin (90 and 10 µg/ml) were determined based on cell viability assay. The role of cyanidin in promoting nerve growth and differentiation was assessed in PC12 cells for up to 72 h. A discernible and statistically significant difference was assessed in neurite extension at both doses at 72 h, followed by pre-treatment with cyanidin. BPA stimulation significantly increased the p-tau expression compared to the control (p < 0.0001). Pre-treatment with cyanidin reduced the tau expression; however, a significant difference was observed compared to control cells (p = 0.0003). Cyanidin significantly enhanced the mRNA expression of Wnt3a (p < 0.0001), β-catenin (p = 0.0004), and NeuroD1 (p = 0.0289), and decreased the expression of WIF1(p = 0.0040) and DKK1 (p < 0.0001), which are Wnt antagonist when compared to cells stimulated with BPA. Conclusively, our finding suggests that cyanidin could agonize nerve growth factor and promote neuronal differentiation, reduce tau-hyperphosphorylation by restoring the Wnt/β-catenin signaling cascade, and thereby render its neuroprotective potential against BPA-induced AD pathology.
Collapse
Affiliation(s)
- Swathi Suresh
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamil Nadu, India
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamil Nadu, India.
| |
Collapse
|
23
|
Firdaus Z, Li X. Unraveling the Genetic Landscape of Neurological Disorders: Insights into Pathogenesis, Techniques for Variant Identification, and Therapeutic Approaches. Int J Mol Sci 2024; 25:2320. [PMID: 38396996 PMCID: PMC10889342 DOI: 10.3390/ijms25042320] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Genetic abnormalities play a crucial role in the development of neurodegenerative disorders (NDDs). Genetic exploration has indeed contributed to unraveling the molecular complexities responsible for the etiology and progression of various NDDs. The intricate nature of rare and common variants in NDDs contributes to a limited understanding of the genetic risk factors associated with them. Advancements in next-generation sequencing have made whole-genome sequencing and whole-exome sequencing possible, allowing the identification of rare variants with substantial effects, and improving the understanding of both Mendelian and complex neurological conditions. The resurgence of gene therapy holds the promise of targeting the etiology of diseases and ensuring a sustained correction. This approach is particularly enticing for neurodegenerative diseases, where traditional pharmacological methods have fallen short. In the context of our exploration of the genetic epidemiology of the three most prevalent NDDs-amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease, our primary goal is to underscore the progress made in the development of next-generation sequencing. This progress aims to enhance our understanding of the disease mechanisms and explore gene-based therapies for NDDs. Throughout this review, we focus on genetic variations, methodologies for their identification, the associated pathophysiology, and the promising potential of gene therapy. Ultimately, our objective is to provide a comprehensive and forward-looking perspective on the emerging research arena of NDDs.
Collapse
Affiliation(s)
- Zeba Firdaus
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
24
|
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and behavioral impairments. Despite extensive research efforts, effective treatment options for AD remain limited. Recently, gene therapy has emerged as a promising avenue for targeted intervention in the pathogenesis of AD. This review will provide an overview of clinical and preclinical studies where gene therapy techniques have been utilized in the context of AD, highlighting their potential as novel therapeutic strategies. While challenges remain, ongoing research and technological advancement continue to enhance the potential of gene therapy as a targeted and personalized therapeutic approach for AD.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
25
|
Schindowski K. Differential Regulation of Neurotrophic Factors During Pathogenic Tau-Aggregation in a Tau Transgenic Mouse Model for Alzheimer's Disease: A Protocol for Double-Labeling mRNA by In Situ Hybridization and Protein Epitopes by Immunohistochemistry. Methods Mol Biol 2024; 2754:361-385. [PMID: 38512677 DOI: 10.1007/978-1-0716-3629-9_20] [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: 03/23/2024]
Abstract
Alzheimer's disease (AD), most tauopathies, and other neurodegenerative diseases are highly associated to impaired neurotrophin regulation and imbalanced neurotrophin transport and distribution. Neurotrophins are crucial for the survival and maintenance of distinct neuronal population therefore their supply is essential for a healthy brain. Tau phosphorylation occurs at different sites of the tau protein and some phospho-epitopes are highly associated to AD (e.g., abnormally phosphorylated tau at Thr212/Ser214). Though the importance of neurotrophins is well known, their analysis in tissue is not trivial and needs careful consideration. Here a detailed protocol is presented, which combines in situ hybridization (ISH) with immunohistochemistry (IHC) to analyze neurotrophin mRNA expression during tau neuropathology and the results were confirmed by immunological methods.With this protocol, it was demonstrated that Brain-Derived Neurotrophic Factor (BDNF) and its receptor Tropomyosin receptor kinase B (TrkB) were significantly decreased in tau-transgenic mice compared to their age-matched littermates. Neurotrophin-3 (NT-3) and its receptor TrkC were not altered with statistical significance, but a tendency for decreased NT-3 and slightly increased TrkC expression was observed in tau transgenic mice. The loss of BDNF-ISH signal was predominantly observed in hippocampus (CA1 and CA3) and cortex (layer II-VI) and verified by BDNF-immunoreactivity. Decreased BDNF and TrkB mRNA was negatively correlated with abnormal tau phosphorylation at Thr212/Ser214 in cortical neurons in transgenic mice. Strikingly, no correlation was observed with age-related phospho-epitopes such as Ser202/Thr205. Interestingly, both, the mRNA and protein levels of Nerve Growth Factor (NGF) were significantly increased in hippocampal neurons in the tau models as demonstrated by ISH, immunofluorescence, and Western Blotting. Here, some co-localization of NGF mRNA and phospho-tau (Thr212/Ser214) was observed but was a rare event. Since there is growing evidence for the relevance of neurotrophic factor distribution in the pathogenesis of neurodegeneration, this technique is a useful tool to investigate the underlying mechanisms and potential therapeutic intervention.
Collapse
Affiliation(s)
- Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany.
| |
Collapse
|
26
|
Phan P, Deshwal A, McMahon TA, Slikas M, Andrews E, Becker B, Kumar TKS. A Review of Rattlesnake Venoms. Toxins (Basel) 2023; 16:2. [PMID: 38276526 PMCID: PMC10818703 DOI: 10.3390/toxins16010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
Venom components are invaluable in biomedical research owing to their specificity and potency. Many of these components exist in two genera of rattlesnakes, Crotalus and Sistrurus, with high toxicity and proteolytic activity variation. This review focuses on venom components within rattlesnakes, and offers a comparison and itemized list of factors dictating venom composition, as well as presenting their known characteristics, activities, and significant applications in biosciences. There are 64 families and subfamilies of proteins present in Crotalus and Sistrurus venom. Snake venom serine proteases (SVSP), snake venom metalloproteases (SVMP), and phospholipases A2 (PLA2) are the standard components in Crotalus and Sistrurus venom. Through this review, we highlight gaps in the knowledge of rattlesnake venom; there needs to be more information on the venom composition of three Crotalus species and one Sistrurus subspecies. We discuss the activity and importance of both major and minor components in biomedical research and drug development.
Collapse
Affiliation(s)
- Phuc Phan
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Anant Deshwal
- Department of Biology, Bradley University, Peoria, IL 61625, USA; (T.A.M.); (M.S.); (E.A.)
| | - Tyler Anthony McMahon
- Department of Biology, Bradley University, Peoria, IL 61625, USA; (T.A.M.); (M.S.); (E.A.)
| | - Matthew Slikas
- Department of Biology, Bradley University, Peoria, IL 61625, USA; (T.A.M.); (M.S.); (E.A.)
| | - Elodie Andrews
- Department of Biology, Bradley University, Peoria, IL 61625, USA; (T.A.M.); (M.S.); (E.A.)
| | - Brian Becker
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA;
| | | |
Collapse
|
27
|
Soltani Dehnavi S, Cembran A, Mahmoudi N, Caballero Aguilar LM, Wang Y, Cheeseman S, Malagutti N, Franks S, Long B, Lisowski L, Harvey AR, Parish CL, Williams RJ, Nisbet DR. Molecular camouflage by a context-specific hydrogel as the key to unlock the potential of viral vector gene therapy. CHEMICAL ENGINEERING JOURNAL 2023; 477:146857. [DOI: 10.1016/j.cej.2023.146857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
28
|
Liu J, van Beusekom H, Bu X, Chen G, Henrique Rosado de Castro P, Chen X, Chen X, Clarkson AN, Farr TD, Fu Y, Jia J, Jolkkonen J, Kim WS, Korhonen P, Li S, Liang Y, Liu G, Liu G, Liu Y, Malm T, Mao X, Oliveira JM, Modo MM, Ramos‐Cabrer P, Ruscher K, Song W, Wang J, Wang X, Wang Y, Wu H, Xiong L, Yang Y, Ye K, Yu J, Zhou X, Zille M, Masters CL, Walczak P, Boltze J, Ji X, Wang Y. Preserving cognitive function in patients with Alzheimer's disease: The Alzheimer's disease neuroprotection research initiative (ADNRI). NEUROPROTECTION 2023; 1:84-98. [PMID: 38223913 PMCID: PMC10783281 DOI: 10.1002/nep3.23] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 01/16/2024]
Abstract
The global trend toward aging populations has resulted in an increase in the occurrence of Alzheimer's disease (AD) and associated socioeconomic burdens. Abnormal metabolism of amyloid-β (Aβ) has been proposed as a significant pathomechanism in AD, supported by results of recent clinical trials using anti-Aβ antibodies. Nonetheless, the cognitive benefits of the current treatments are limited. The etiology of AD is multifactorial, encompassing Aβ and tau accumulation, neuroinflammation, demyelination, vascular dysfunction, and comorbidities, which collectively lead to widespread neurodegeneration in the brain and cognitive impairment. Hence, solely removing Aβ from the brain may be insufficient to combat neurodegeneration and preserve cognition. To attain effective treatment for AD, it is necessary to (1) conduct extensive research on various mechanisms that cause neurodegeneration, including advances in neuroimaging techniques for earlier detection and a more precise characterization of molecular events at scales ranging from cellular to the full system level; (2) identify neuroprotective intervention targets against different neurodegeneration mechanisms; and (3) discover novel and optimal combinations of neuroprotective intervention strategies to maintain cognitive function in AD patients. The Alzheimer's Disease Neuroprotection Research Initiative's objective is to facilitate coordinated, multidisciplinary efforts to develop systemic neuroprotective strategies to combat AD. The aim is to achieve mitigation of the full spectrum of pathological processes underlying AD, with the goal of halting or even reversing cognitive decline.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Heleen van Beusekom
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Xian‐Le Bu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Gong Chen
- Guangdong‐HongKong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouGuangdongChina
| | | | - Xiaochun Chen
- Fujian Key Laboratory of Molecular Neurology, Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Institute of NeuroscienceFujian Medical UniversityFuzhouFujianChina
| | - Xiaowei Chen
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
- Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New ZealandUniversity of OtagoDunedinNew Zealand
| | - Tracy D. Farr
- School of Life SciencesUniversity of NottinghamNottinghamUK
| | - Yuhong Fu
- Brain and Mind Centre & School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Jukka Jolkkonen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Woojin Scott Kim
- Brain and Mind Centre & School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Guang‐Hui Liu
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Membrane Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Yu‐Hui Liu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Xiaobo Mao
- Institute for Cell Engineering, Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of MinhoGuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Mike M. Modo
- Department of Bioengineering, McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Radiology, McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Pedro Ramos‐Cabrer
- Magnetic Resonance Imaging LaboratoryCIC BiomaGUNE Research Center, Basque Research and Technology Alliance (BRTA)Donostia‐San SebastianSpain
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical SciencesLund UniversityLundSweden
| | - Weihong Song
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province. Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Medical UniversityZhejiangChina
| | - Jun Wang
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Xuanyue Wang
- School of Optometry and Vision ScienceUniversity of New South WalesSydneyNew South WalesAustralia
| | - Yun Wang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic, Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National, Health Commission and State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
- PKU‐IDG/McGovern Institute for Brain ResearchPeking UniversityBeijingChina
| | - Haitao Wu
- Department of NeurobiologyBeijing Institute of Basic Medical SciencesBeijingChina
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like Intelligence, Shanghai Fourth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi Yang
- Department of NeurologyThe First Hospital of Jilin University, Chang ChunJilinChina
| | - Keqiang Ye
- Faculty of Life and Health SciencesBrain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced TechnologyShenzhenChina
| | - Jin‐Tai Yu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin‐Fu Zhou
- Division of Health Sciences, School of Pharmacy and Medical Sciences and Sansom InstituteUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Suzhou Auzone BiotechSuzhouJiangsuChina
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and ToxicologyUniversity of ViennaViennaAustria
| | - Colin L. Masters
- The Florey InstituteThe University of Melbourne, ParkvilleVictoriaAustralia
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | | | - Xunming Ji
- Department of NeurosurgeryXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yan‐Jiang Wang
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
- Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| |
Collapse
|
29
|
Gross J, Knipper M, Mazurek B. Candidate Key Proteins in Tinnitus: A Bioinformatic Study of Synaptic Transmission in Spiral Ganglion Neurons. Cell Mol Neurobiol 2023; 43:4189-4207. [PMID: 37736859 PMCID: PMC10661836 DOI: 10.1007/s10571-023-01405-w] [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: 07/29/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023]
Abstract
To study key proteins associated with changes in synaptic transmission in the spiral ganglion in tinnitus, we build three gene lists from the GeneCard database: 1. Perception of sound (PoS), 2. Acoustic stimulation (AcouStim), and 3. Tinnitus (Tin). Enrichment analysis by the DAVID database resulted in similar Gene Ontology (GO) terms for cellular components in all gene lists, reflecting synaptic structures known to be involved in auditory processing. The STRING protein-protein interaction (PPI) network and the Cytoscape data analyzer were used to identify the top two high-degree proteins (HDPs) and their high-score interaction proteins (HSIPs) identified by the combined score (CS) of the corresponding edges. The top two protein pairs (key proteins) for the PoS are BDNF-GDNF and OTOF-CACNA1D and for the AcouStim process BDNF-NTRK2 and TH-CALB1. The Tin process showed BDNF and NGF as HDPs, with high-score interactions with NTRK1 and NGFR at a comparable level. Compared to the PoS and AcouStim process, the number of HSIPs of key proteins (CS > 90. percentile) increases strongly in Tin. In the PoS and AcouStim networks, BDNF receptor signaling is the dominant pathway, and in the Tin network, the NGF-signaling pathway is of similar importance. Key proteins and their HSIPs are good indicators of biological processes and of signaling pathways characteristic for the normal hearing on the one hand and tinnitus on the other.
Collapse
Affiliation(s)
- Johann Gross
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Leibniz Society of Science Berlin, Berlin, Germany.
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- Leibniz Society of Science Berlin, Berlin, Germany
| | - Birgit Mazurek
- Tinnitus Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
30
|
Gupta R, Advani D, Yadav D, Ambasta RK, Kumar P. Dissecting the Relationship Between Neuropsychiatric and Neurodegenerative Disorders. Mol Neurobiol 2023; 60:6476-6529. [PMID: 37458987 DOI: 10.1007/s12035-023-03502-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/11/2023] [Indexed: 09/28/2023]
Abstract
Neurodegenerative diseases (NDDs) and neuropsychiatric disorders (NPDs) are two common causes of death in elderly people, which includes progressive neuronal cell death and behavioral changes. NDDs include Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and motor neuron disease, characterized by cognitive defects and memory impairment, whereas NPDs include depression, seizures, migraine headaches, eating disorders, addictions, palsies, major depressive disorders, anxiety, and schizophrenia, characterized by behavioral changes. Mounting evidence demonstrated that NDDs and NPDs share an overlapping mechanism, which includes post-translational modifications, the microbiota-gut-brain axis, and signaling events. Mounting evidence demonstrated that various drug molecules, namely, natural compounds, repurposed drugs, multitarget directed ligands, and RNAs, have been potentially implemented as therapeutic agents against NDDs and NPDs. Herein, we highlighted the overlapping mechanism, the role of anxiety/stress-releasing factors, cytosol-to-nucleus signaling, and the microbiota-gut-brain axis in the pathophysiology of NDDs and NPDs. We summarize the therapeutic application of natural compounds, repurposed drugs, and multitarget-directed ligands as therapeutic agents. Lastly, we briefly described the application of RNA interferences as therapeutic agents in the pathogenesis of NDDs and NPDs. Neurodegenerative diseases and neuropsychiatric diseases both share a common signaling molecule and molecular phenomenon, namely, pro-inflammatory cytokines, γCaMKII and MAPK/ERK, chemokine receptors, BBB permeability, and the gut-microbiota-brain axis. Studies have demonstrated that any alterations in the signaling mentioned above molecules and molecular phenomena lead to the pathophysiology of neurodegenerative diseases, namely, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, and neuropsychiatric disorders, such as bipolar disorder, schizophrenia, depression, anxiety, autism spectrum disorder, and post-traumatic stress disorder.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Divya Yadav
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India.
| |
Collapse
|
31
|
Chen W, Zhang Y, Liang J, Zhang Z, Zhang L, Huang E, Zhang G, Lu L, Han Y, Shi J. Disrupting astrocyte-neuron lactate transport prevents cocaine seeking after prolonged withdrawal. SCIENCE ADVANCES 2023; 9:eadi4462. [PMID: 37878699 PMCID: PMC10599624 DOI: 10.1126/sciadv.adi4462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/21/2023] [Indexed: 10/27/2023]
Abstract
Energy supply, especially the transfer of lactate from astrocytes to neurons, is critical for neuronal plasticity. However, its role in the incubation of cocaine craving remains largely unknown. Using an extended-access self-administration model and in vivo 1H-magnetic resonance spectroscopy, we found that lactate synthesis in the central amygdala (CeA) is required for the intensified cocaine craving after prolonged withdrawal. Furthermore, incubated cocaine seeking was associated with a selective increase in monocarboxylate transporter 2 (MCT2) and MCT4 expression levels. Down-regulation of astrocytic MCT4 or neuronal MCT2 using targeted antisense oligonucleotides or cell type-specific shRNA attenuated cocaine craving and reduced the expression of plasticity-related proteins and excitatory synaptic transmission. Meanwhile, lactate administration rescued MCT4 but not MCT2 disruption-induced behavioral changes due to the inability of lactate to be transported into neurons. Together, our study highlights the critical role of astrocyte-neuron lactate transport in the CeA in the incubation of cocaine craving and suggests a potential therapeutic target for drug addiction.
Collapse
Affiliation(s)
- Wenjun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yan Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jie Liang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Zhongyu Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Libo Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Enze Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Guipeng Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lin Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
- The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing, 100191, China
- The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| |
Collapse
|
32
|
Tonev D, Momchilova A. Therapeutic Plasma Exchange and Multiple Sclerosis Dysregulations: Focus on the Removal of Pathogenic Circulatory Factors and Altering Nerve Growth Factor and Sphingosine-1-Phosphate Plasma Levels. Curr Issues Mol Biol 2023; 45:7749-7774. [PMID: 37886933 PMCID: PMC10605592 DOI: 10.3390/cimb45100489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/28/2023] Open
Abstract
Multiple sclerosis (MS) is predominantly an immune-mediated disease of the central nervous system (CNS) of unknown etiology with a possible genetic predisposition and effect of certain environmental factors. It is generally accepted that the disease begins with an autoimmune inflammatory reaction targeting oligodendrocytes followed by a rapid depletion of their regenerative capacity with subsequent permanent neurodegenerative changes and disability. Recent research highlights the central role of B lymphocytes and the corresponding IgG and IgM autoantibodies in newly forming MS lesions. Thus, their removal along with the modulation of certain bioactive molecules to improve neuroprotection using therapeutic plasma exchange (TPE) becomes of utmost importance. Recently, it has been proposed to determine the levels and precise effects of both beneficial and harmful components in the serum of MS patients undergoing TPE to serve as markers for appropriate TPE protocols. In this review we discuss some relevant examples, focusing on the removal of pathogenic circulating factors and altering the plasma levels of nerve growth factor and sphingosine-1-phosphate by TPE. Altered plasma levels of the reviewed molecular compounds in response to TPE reflect a successful reduction of the pro-inflammatory burden at the expense of an increase in anti-inflammatory potential in the circulatory and CNS compartments.
Collapse
Affiliation(s)
- Dimitar Tonev
- Department of Anesthesiology and Intensive Care, University Hospital “Tzaritza Yoanna—ISUL”, Medical University of Sofia, 1527 Sofia, Bulgaria
| | - Albena Momchilova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Science, 1113 Sofia, Bulgaria;
| |
Collapse
|
33
|
Carrera I, Corzo L, Martínez-Iglesias O, Naidoo V, Cacabelos R. Neuroprotective Effect of Nosustrophine in a 3xTg Mouse Model of Alzheimer's Disease. Pharmaceuticals (Basel) 2023; 16:1306. [PMID: 37765114 PMCID: PMC10535028 DOI: 10.3390/ph16091306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodegeneration, characterized by the progressive deterioration of neurons and glial cells, is a feature of Alzheimer's disease (AD). The present study aims to demonstrate that the onset and early progression of neurodegenerative processes in transgenic mice models of AD can be delayed by a cocktail of neurotrophic factors and derived peptides named Nosustrophine, a nootropic supplement made by a peptide complex extracted from the young porcine brain, ensuring neuroprotection and improving neuro-functional recovery. Experimental 3xTg-APP/Bin1/COPS5 transgenic mice models of AD were treated with Nosustrophine at two different early ages, and their neuropathological hallmark and behavior response were analyzed. Results showed that Nosustrophine increased the activity of the immune system and reduced pathological changes in the hippocampus and cortex by halting the development of amyloid plaques, mainly seen in mice of 3-4 months of age, indicating that its effect is more preventive than therapeutic. Taken together, the results indicate the potent neuroprotective activity of Nosustrophine and its stimulating effects on neuronal plasticity. This study shows for the first time an effective therapy using nootropic supplements against degenerative diseases, although further investigation is needed to understand their molecular pathways.
Collapse
Affiliation(s)
- Iván Carrera
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165 Bergondo, Corunna, Spain; (L.C.); (O.M.-I.); (V.N.); (R.C.)
| | | | | | | | | |
Collapse
|
34
|
Dafre R, Wasnik P. Current Diagnostic and Treatment Methods of Alzheimer's Disease: A Narrative Review. Cureus 2023; 15:e45649. [PMID: 37868425 PMCID: PMC10589453 DOI: 10.7759/cureus.45649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
To diagnose and predict the possibility of Alzheimer's or a different kind of dementia, medical professionals employ tests that look at a patient's mental competence; however, such methods are impracticable. A reliable diagnosis at the start of treatment is essential for therapy. Except in situations with apparent genetic variations, most Alzheimer's patients lack a known etiology. Therefore, every Alzheimer's patient receives the same treatment plan, regardless of the etiology, which may or may not be successful in slowing or preventing the disease's progression. Tau pathology is further complicated by the amyloid buildup that arises from the cellular phase of Alzheimer's disease (AD). Alzheimer's is a degenerative, diverse, complicated, and incurable neurological disorder primarily affecting elderly individuals. The currently accepted drugs available for treating AD, which involve cholinesterase inhibitors and N-methyl-D-aspartate (NMDA)-receptor antagonists only provide temporary relief from symptoms. The neurological disorder primarily affecting elderly individuals is degenerative, diverse, complicated, and incurable. Accurate diagnosis is the most essential prerequisite before beginning therapy. Most Alzheimer's patients' causes are still unclear, except for instances where hereditary variations have been noted. The gut microbiota composition significantly influences AD and any age-associated neurological illness. Therapies are very useful in improving the cognitive functions of AD. New microbiota-based therapy alternatives may now be available due to the more recent connection between the altered gut microbiome and neurodegeneration through the gut microbiota-brain axis.
Collapse
Affiliation(s)
- Rajshri Dafre
- Health Science, Jawaharlal Nehru Medical College, Wardha, IND
| | - Praful Wasnik
- Medicine, Jawaharlal Nehru Medical College, Wardha, IND
| |
Collapse
|
35
|
Gao J, Li L. Enhancement of neural regeneration as a therapeutic strategy for Alzheimer's disease (Review). Exp Ther Med 2023; 26:444. [PMID: 37614437 PMCID: PMC10443056 DOI: 10.3892/etm.2023.12143] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/18/2023] [Indexed: 08/25/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia worldwide, has gradually become a global health concern for society and individuals with the process of global ageing. Although extensive research has been carried out on AD, the etiology and pathological mechanism of the disease are still unclear, and there is no specific drug to cure or delay AD progression. The exploration of enhancing nerve regeneration in AD has gradually attracted increasing attention. In the current review, the existing therapeutic strategies were summarized to induce nerve regeneration which can increase the number of neurons, and improve the survival of neurons, the plasticity of synapses and synaptic activity. The strategies include increasing neurotrophic expression (such as brain-derived neurotrophic factor and nerve growth factor), inhibiting acetylcholinesterase (such as donepezil, tacrine, rivastigmine and galanthamine), elevating histone deacetylase levels (such as RGFP-966, Tasquinimod, CM-414 and 44B), stimulating the brain by physiotherapy (such as near-infrared light, repetitive transcranial magnetic stimulation, and transcranial direct current stimulation) and transplanting exogenous neural stem cells. However, further evaluations need to be performed to determine the optimal treatment. The present study reviews recent interventions for enhancing adult neurogenesis and attempts to elucidate their mechanisms of action, which may provide a theoretical basis for inducing nerve regeneration to fight against AD.
Collapse
Affiliation(s)
- Junyan Gao
- Department of Physiology and Pharmacology, Health Science Centre, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Liping Li
- Department of Physiology and Pharmacology, Health Science Centre, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
36
|
Quan M, Cao S, Wang Q, Wang S, Jia J. Genetic Phenotypes of Alzheimer's Disease: Mechanisms and Potential Therapy. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:333-349. [PMID: 37589021 PMCID: PMC10425323 DOI: 10.1007/s43657-023-00098-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 08/18/2023]
Abstract
Years of intensive research has brought us extensive knowledge on the genetic and molecular factors involved in Alzheimer's disease (AD). In addition to the mutations in the three main causative genes of familial AD (FAD) including presenilins and amyloid precursor protein genes, studies have identified several genes as the most plausible genes for the onset and progression of FAD, such as triggering receptor expressed on myeloid cells 2, sortilin-related receptor 1, and adenosine triphosphate-binding cassette transporter subfamily A member 7. The apolipoprotein E ε4 allele is reported to be the strongest genetic risk factor for sporadic AD (SAD), and it also plays an important role in FAD. Here, we reviewed recent developments in genetic and molecular studies that contributed to the understanding of the genetic phenotypes of FAD and compared them with SAD. We further reviewed the advancements in AD gene therapy and discussed the future perspectives based on the genetic phenotypes.
Collapse
Affiliation(s)
- Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
- National Medical Center for Neurological Disorders and National Clinical Research Center for Geriatric Diseases, Beijing, 100053 China
| | - Shuman Cao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
- National Medical Center for Neurological Disorders and National Clinical Research Center for Geriatric Diseases, Beijing, 100053 China
| | - Shiyuan Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China
- National Medical Center for Neurological Disorders and National Clinical Research Center for Geriatric Diseases, Beijing, 100053 China
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053 China
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053 China
- Center of Alzheimer’s Disease, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, 100053 China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053 China
| |
Collapse
|
37
|
Jeyaraman M, Rajendran RL, Muthu S, Jeyaraman N, Sharma S, Jha SK, Muthukanagaraj P, Hong CM, Furtado da Fonseca L, Santos Duarte Lana JF, Ahn BC, Gangadaran P. An update on stem cell and stem cell-derived extracellular vesicle-based therapy in the management of Alzheimer's disease. Heliyon 2023; 9:e17808. [PMID: 37449130 PMCID: PMC10336689 DOI: 10.1016/j.heliyon.2023.e17808] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 05/10/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Globally, neurological diseases pose a major burden to healthcare professionals in terms of the management and prevention of the disorder. Among neurological diseases, Alzheimer's disease (AD) accounts for 50%-70% of dementia and is the fifth leading cause of mortality worldwide. AD is a progressive, degenerative neurological disease, with the loss of neurons and synapses in the cerebral cortex and subcortical regions. The management of AD remains a debate among physicians as no standard and specific "disease-modifying" modality is available. The concept of 'Regenerative Medicine' is aimed at regenerating the degenerated neural tissues to reverse the pathology in AD. Genetically modified engineered stem cells modify the course of AD after transplantation into the brain. Extracellular vesicles (EVs) are an emerging new approach in cell communication that involves the transfer of cellular materials from parental cells to recipient cells, resulting in changes at the molecular and signaling levels in the recipient cells. EVs are a type of vesicle that can be transported between cells. Many have proposed that EVs produced from mesenchymal stem cells (MSCs) may have therapeutic promise in the treatment of AD. The biology of AD, as well as the potential applications of stem cells and their derived EVs-based therapy, were explored in this paper.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu, 600056, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
- Department of Orthopedics, Government Dindigul Medical College and Hospital, Dindigul, Tamil Nadu, 624001, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
- Department of Orthopedics, Shri Sathya Sai Medical College and Research Institute, Sri Balaji Vidyapeeth, Chengalpet, Tamil Nadu, 603108, India
| | - Shilpa Sharma
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, 226010, India
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Purushothaman Muthukanagaraj
- Department of Internal Medicine & Psychiatry, SUNY-Upstate Binghamton Clinical Campus, Binghamton, NY, 13904, USA
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Lucas Furtado da Fonseca
- Department of Orthopedics, The Federal University of São Paulo, São Paulo, 04023-062, SP, Brazil
| | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| |
Collapse
|
38
|
Madokoro Y, Kato D, Tsuda Y, Arakawa I, Suzuki K, Sato T, Mizuno M, Uchida Y, Ojika K, Matsukawa N. Direct Enhancement Effect of Hippocampal Cholinergic Neurostimulating Peptide on Cholinergic Activity in the Hippocampus. Int J Mol Sci 2023; 24:ijms24108916. [PMID: 37240261 DOI: 10.3390/ijms24108916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The cholinergic efferent network from the medial septal nucleus to the hippocampus is crucial for learning and memory. This study aimed to clarify whether hippocampal cholinergic neurostimulating peptide (HCNP) has a rescue function in the cholinergic dysfunction of HCNP precursor protein (HCNP-pp) conditional knockout (cKO). Chemically synthesized HCNP or a vehicle were continuously administered into the cerebral ventricle of HCNP-pp cKO mice and littermate floxed (control) mice for two weeks via osmotic pumps. We immunohistochemically measured the cholinergic axon volume in the stratum oriens and functionally evaluated the local field potential in the CA1. Furthermore, choline acetyltransferase (ChAT) and nerve growth factor (NGF) receptor (TrkA and p75NTR) abundances were quantified in wild-type (WT) mice administered HCNP or the vehicle. As a result, HCNP administration morphologically increased the cholinergic axonal volume and electrophysiological theta power in HCNP-pp cKO and control mice. Following the administration of HCNP to WT mice, TrkA and p75NTR levels also decreased significantly. These data suggest that extrinsic HCNP may compensate for the reduced cholinergic axonal volume and theta power in HCNP-pp cKO mice. HCNP may function complementarily to NGF in the cholinergic network in vivo. HCNP may represent a therapeutic candidate for neurological diseases with cholinergic dysfunction, e.g., Alzheimer's disease and Lewy body dementia.
Collapse
Affiliation(s)
- Yuta Madokoro
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Daisuke Kato
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yo Tsuda
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Itsumi Arakawa
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Kengo Suzuki
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Toyohiro Sato
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Masayuki Mizuno
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Yuto Uchida
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Kosei Ojika
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|
39
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
40
|
Palasz E, Wilkaniec A, Stanaszek L, Andrzejewska A, Adamczyk A. Glia-Neurotrophic Factor Relationships: Possible Role in Pathobiology of Neuroinflammation-Related Brain Disorders. Int J Mol Sci 2023; 24:ijms24076321. [PMID: 37047292 PMCID: PMC10094105 DOI: 10.3390/ijms24076321] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Neurotrophic factors (NTFs) play an important role in maintaining homeostasis of the central nervous system (CNS) by regulating the survival, differentiation, maturation, and development of neurons and by participating in the regeneration of damaged tissues. Disturbances in the level and functioning of NTFs can lead to many diseases of the nervous system, including degenerative diseases, mental diseases, and neurodevelopmental disorders. Each CNS disease is characterized by a unique pathomechanism, however, the involvement of certain processes in its etiology is common, such as neuroinflammation, dysregulation of NTFs levels, or mitochondrial dysfunction. It has been shown that NTFs can control the activation of glial cells by directing them toward a neuroprotective and anti-inflammatory phenotype and activating signaling pathways responsible for neuronal survival. In this review, our goal is to outline the current state of knowledge about the processes affected by NTFs, the crosstalk between NTFs, mitochondria, and the nervous and immune systems, leading to the inhibition of neuroinflammation and oxidative stress, and thus the inhibition of the development and progression of CNS disorders.
Collapse
Affiliation(s)
- Ewelina Palasz
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: (E.P.); (A.A.)
| | - Anna Wilkaniec
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Luiza Stanaszek
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Anna Andrzejewska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Agata Adamczyk
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: (E.P.); (A.A.)
| |
Collapse
|
41
|
Fitzgerald GS, Chuchta TG, McNay EC. Insulin‐like growth factor‐2 is a promising candidate for the treatment and prevention of Alzheimer's disease. CNS Neurosci Ther 2023; 29:1449-1469. [PMID: 36971212 PMCID: PMC10173726 DOI: 10.1111/cns.14160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 02/06/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Current AD treatments slow the rate of cognitive decline, but do not restore lost function. One reason for the low efficacy of current treatments is that they fail to target neurotrophic processes, which are thought to be essential for functional recovery. Bolstering neurotrophic processes may also be a viable strategy for preventative treatment, since structural losses are thought to underlie cognitive decline in AD. The challenge of identifying presymptomatic patients who might benefit from preventative treatment means that any such treatment must meet a high standard of safety and tolerability. The neurotrophic peptide insulin-like growth factor-2 (IGF2) is a promising candidate for both treating and preventing AD-induced cognitive decline. Brain IGF2 expression declines in AD patients. In rodent models of AD, exogenous IGF2 modulates multiple aspects of AD pathology, resulting in (1) improved cognitive function; (2) stimulation of neurogenesis and synaptogenesis; and, (3) neuroprotection against cholinergic dysfunction and beta amyloid-induced neurotoxicity. Preclinical evidence suggests that IGF2 is likely to be safe and tolerable at therapeutic doses. In the preventative treatment context, the intranasal route of administration is likely to be the preferred method for achieving the therapeutic effect without risking adverse side effects. For patients already experiencing AD dementia, routes of administration that deliver IGF2 directly access the CNS may be necessary. Finally, we discuss several strategies for improving the translational validity of animal models used to study the therapeutic potential of IGF2.
Collapse
Affiliation(s)
| | | | - E C McNay
- University at Albany, Albany, New York, USA
| |
Collapse
|
42
|
Lin L, Li C, Li T, Zheng J, Shu Y, Zhang J, Shen Y, Ren D. Plant‐derived peptides for the improvement of Alzheimer's disease: Production, functions, and mechanisms. FOOD FRONTIERS 2023. [DOI: 10.1002/fft2.210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Affiliation(s)
- Like Lin
- Key Laboratory of Synthetic and Natural Functional Molecule of Ministry of Education College of Chemistry and Materials Science National Demonstration Center for Experimental Chemistry Education Northwest University Xi'an Shaanxi China
| | - Cong Li
- Key Laboratory of Synthetic and Natural Functional Molecule of Ministry of Education College of Chemistry and Materials Science National Demonstration Center for Experimental Chemistry Education Northwest University Xi'an Shaanxi China
| | - Tingting Li
- Key Laboratory of Synthetic and Natural Functional Molecule of Ministry of Education College of Chemistry and Materials Science National Demonstration Center for Experimental Chemistry Education Northwest University Xi'an Shaanxi China
| | - Jingyi Zheng
- Key Laboratory of Synthetic and Natural Functional Molecule of Ministry of Education College of Chemistry and Materials Science National Demonstration Center for Experimental Chemistry Education Northwest University Xi'an Shaanxi China
| | - Yu Shu
- College of Food Science and Technology Northwest University Xi'an Shaanxi China
| | - Jingjing Zhang
- College of Chemical Engineering Northwest University Xi'an Shaanxi China
| | - Yehua Shen
- Key Laboratory of Synthetic and Natural Functional Molecule of Ministry of Education College of Chemistry and Materials Science National Demonstration Center for Experimental Chemistry Education Northwest University Xi'an Shaanxi China
| | - Difeng Ren
- Beijing Key Laboratory of Food Processing and Safety in Forestry Department of Food Science and Engineering, College of Biological Sciences and Biotechnology Beijing Forestry University Beijing China
| |
Collapse
|
43
|
Neurotrophic Factors as Regenerative Therapy for Neurodegenerative Diseases: Current Status, Challenges and Future Perspectives. Int J Mol Sci 2023; 24:ijms24043866. [PMID: 36835277 PMCID: PMC9968045 DOI: 10.3390/ijms24043866] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), spinal cord injury (SCI), and amyotrophic lateral sclerosis (ALS), are characterized by acute or chronic progressive loss of one or several neuronal subtypes. However, despite their increasing prevalence, little progress has been made in successfully treating these diseases. Research has recently focused on neurotrophic factors (NTFs) as potential regenerative therapy for neurodegenerative diseases. Here, we discuss the current state of knowledge, challenges, and future perspectives of NTFs with a direct regenerative effect in chronic inflammatory and degenerative disorders. Various systems for delivery of NTFs, such as stem and immune cells, viral vectors, and biomaterials, have been applied to deliver exogenous NTFs to the central nervous system, with promising results. The challenges that currently need to be overcome include the amount of NTFs delivered, the invasiveness of the delivery route, the blood-brain barrier permeability, and the occurrence of side effects. Nevertheless, it is important to continue research and develop standards for clinical applications. In addition to the use of single NTFs, the complexity of chronic inflammatory and degenerative diseases may require combination therapies targeting multiple pathways or other possibilities using smaller molecules, such as NTF mimetics, for effective treatment.
Collapse
|
44
|
Muresanu DF, Sharma A, Tian ZR, Lafuente JV, Nozari A, Feng L, Buzoianu AD, Wiklund L, Sharma HS. Nanowired Delivery of Cerebrolysin with Mesenchymal Stem Cells Attenuates Heat Stress-Induced Exacerbation of Neuropathology Following Brain Blast Injury. ADVANCES IN NEUROBIOLOGY 2023; 32:231-270. [PMID: 37480463 DOI: 10.1007/978-3-031-32997-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Blast brain injury (bBI) following explosive detonations in warfare is one of the prominent causes of multidimensional insults to the central nervous and other vital organs injury. Several military personnel suffered from bBI during the Middle East conflict at hot environment. The bBI largely occurs due to pressure waves, generation of heat together with release of shrapnel and gun powders explosion with penetrating and/or impact head trauma causing multiple brain damage. As a result, bBI-induced secondary injury causes breakdown of the blood-brain barrier (BBB) and edema formation that further results in neuronal, glial and axonal injuries. Previously, we reported endocrine imbalance and influence of diabetes on bBI-induced brain pathology that was significantly attenuated by nanowired delivery of cerebrolysin in model experiments. Cerebrolysin is a balanced composition of several neurotrophic factors, and active peptide fragment is capable of neuroprotection in several neurological insults. Exposure to heat stress alone causes BBB damage, edema formation and brain pathology. Thus, it is quite likely that hot environment further exacerbates the consequences of bBI. Thus, novel therapeutic strategies using nanodelivery of stem cell and cerebrolysin may further enhance superior neuroprotection in bBI at hot environment. Our observations are the first to show that combined nanowired delivery of mesenchymal stem cells (MSCs) and cerebrolysin significantly attenuated exacerbation of bBI in hot environment and induced superior neuroprotection, not reported earlier. The possible mechanisms of neuroprotection with MSCs and cerebrolysin in bBI are discussed in the light of current literature.
Collapse
Affiliation(s)
- Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan, Hebei Province, China
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
45
|
Wiart M, Tavakoli C, Hubert V, Hristovska I, Dumot C, Parola S, Lerouge F, Chauveau F, Canet-Soulas E, Pascual O, Cormode DP, Brun E, Elleaume H. Use of metal-based contrast agents for in vivo MR and CT imaging of phagocytic cells in neurological pathologies. J Neurosci Methods 2023; 383:109729. [PMID: 36272462 DOI: 10.1016/j.jneumeth.2022.109729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
The activation of phagocytic cells is a hallmark of many neurological diseases. Imaging them in their 3-dimensional cerebral environment over time is crucial to better understand their role in disease pathogenesis and to monitor their potential therapeutic effects. Phagocytic cells have the ability to internalize metal-based contrast agents both in vitro and in vivo and can thus be tracked by magnetic resonance imaging (MRI) or computed tomography (CT). In this review article, we summarize the different labelling strategies, contrast agents, and in vivo imaging modalities that can be used to monitor cells with phagocytic activity in the central nervous system using MRI and CT, with a focus on clinical applications. Metal-based nanoparticle contrast agents such as gadolinium, gold and iron are ideal candidates for these applications as they have favourable magnetic and/or radiopaque properties and can be fine-tuned for optimal uptake by phagocytic cells. However, they also come with downsides due to their potential toxicity, especially in the brain where they might accumulate. We therefore conclude our review by discussing the pitfalls, safety and potential for clinical translation of these metal-based neuroimaging techniques. Early results in patients with neuropathologies such as multiple sclerosis, stroke, trauma, cerebral aneurysm and glioblastoma are promising. If the challenges represented by safety issues are overcome, phagocytic cells imaging will be a very valuable tool for studying and understanding the inflammatory response and evaluating treatments that aim at mitigating this response in patients with neurological diseases.
Collapse
Affiliation(s)
- Marlène Wiart
- Univ. Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69003 Lyon, France; CNRS, Lyon, France.
| | - Clément Tavakoli
- Univ. Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69003 Lyon, France; Univ. Grenoble Alpes, INSERM UA7 STROBE, 38000 Grenoble, France
| | - Violaine Hubert
- Univ. Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69003 Lyon, France
| | | | - Chloé Dumot
- Univ. Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69003 Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Stéphane Parola
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, 69364 Lyon, France
| | - Frédéric Lerouge
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, 69364 Lyon, France
| | - Fabien Chauveau
- CNRS, Lyon, France; Univ. Lyon, Lyon Neurosciences Research Center, CNRS UMR5292, INSERM U1028, Université Claude Bernard Lyon 1, 69003 Lyon, France
| | - Emmanuelle Canet-Soulas
- Univ. Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69003 Lyon, France
| | | | - David P Cormode
- Department of Radiology, University of Pennsylvania, Pennsylvania, United States
| | - Emmanuel Brun
- Univ. Grenoble Alpes, INSERM UA7 STROBE, 38000 Grenoble, France
| | - Hélène Elleaume
- Univ. Grenoble Alpes, INSERM UA7 STROBE, 38000 Grenoble, France
| |
Collapse
|
46
|
Jiao L, Yu Z, Zhong X, Yao W, Xing L, Ma G, Shen J, Wu Y, Du K, Liu J, Tong J, Fu J, Wei M, Liu M. Cordycepin improved neuronal synaptic plasticity through CREB-induced NGF upregulation driven by MG-M2 polarization: a microglia-neuron symphony in AD. Biomed Pharmacother 2023; 157:114054. [PMID: 36462314 DOI: 10.1016/j.biopha.2022.114054] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/14/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
PURPOSE Microglia-neuron crosstalk is critically involved in synaptic plasticity and degeneration by releasing diverse mediators in Alzheimer's disease (AD). Therefore, determining contributors that modulate the systemic microenvironment is essential. Cordycepin (CCS) is a novel neuroprotective compound obtained from Cordyceps militaris. However, the anti-AD efficacy and potential mechanism of CCS treatment remain unclear. This study aimed to elucidate the microglia-neuron symphony in AD after CCS treatment and to explore the possible mechanisms of its neuroprotective efficacy. METHODS AND RESULTS CCS treatment improved learning and memory impairment in 9-month-old APP/PS1 mice by behavioral tests. CCS polarized the microglia from M1 to M2, inhibited neuronal apoptosis and promoted synaptic remodeling accompanied by in vivo and in vitro upregulation of NGF. The cAMP-response element-binding protein (CREB) was also activated after MG-M2 polarization. Further, we verified that the sg3 promoter region of NGF (-1018 to -1011) is the key binding site for CREB-induced NGF transcription, which increased NGF expression and secretion. Finally, microglia-derived NGF was confirmed as an important mediator in microglia-neuron symphony to improve the neuronal microenvironment after CCS treatment. CONCLUSIONS CCS improved the neuronal synaptic plasticity and senescence by promoting MG-M2 activation driven by CREB-induced NGF upregulation and facilitated symphony communication between the microglia and neuron in AD. This study provides a new perspective on the development of a novel strategy for anti-AD therapy and offers new targets for anti-AD drug development.
Collapse
Affiliation(s)
- Linchi Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Zhihua Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Xin Zhong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Lijuan Xing
- Precision Laboratory of Panjin Central Hospital, Panjin, 124000, China.
| | - Guowei Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Jiajia Shen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Yuqiang Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Junxiu Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Junhui Tong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Jia Fu
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110179, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110179, China.
| | - Mingyan Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
47
|
Toledano-Díaz A, Álvarez MI, Toledano A. The relationships between neuroglial and neuronal changes in Alzheimer's disease, and the related controversies II: gliotherapies and multimodal therapy. J Cent Nerv Syst Dis 2022; 14:11795735221123896. [PMID: 36407561 PMCID: PMC9666878 DOI: 10.1177/11795735221123896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/05/2022] [Indexed: 08/30/2023] Open
Abstract
Since the original description of Alzheimer´s disease (AD), research into this condition has mainly focused on assessing the alterations to neurons associated with dementia, and those to the circuits in which they are involved. In most of the studies on human brains and in many models of AD, the glial cells accompanying these neurons undergo concomitant alterations that aggravate the course of neurodegeneration. As a result, these changes to neuroglial cells are now included in all the "pathogenic cascades" described in AD. Accordingly, astrogliosis and microgliosis, the main components of neuroinflammation, have been integrated into all the pathogenic theories of this disease, as discussed in this part of the two-part monograph that follows an accompanying article on gliopathogenesis and glioprotection. This initial reflection verified the implication of alterations to the neuroglia in AD, suggesting that these cells may also represent therapeutic targets to prevent neurodegeneration. In this second part of the monograph, we will analyze the possibilities of acting on glial cells to prevent or treat the neurodegeneration that is the hallmark of AD and other pathologies. Evidence of the potential of different pharmacological, non-pharmacological, cell and gene therapies (widely treated) to prevent or treat this disease is now forthcoming, in most cases as adjuncts to other therapies. A comprehensive AD multimodal therapy is proposed in which neuronal and neuroglial pharmacological treatments are jointly considered, as well as the use of new cell and gene therapies and non-pharmacological therapies that tend to slow down the progress of dementia.
Collapse
|
48
|
Hayes G, Pinto J, Sparks SN, Wang C, Suri S, Bulte DP. Vascular smooth muscle cell dysfunction in neurodegeneration. Front Neurosci 2022; 16:1010164. [PMID: 36440263 PMCID: PMC9684644 DOI: 10.3389/fnins.2022.1010164] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 09/01/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the key moderators of cerebrovascular dynamics in response to the brain's oxygen and nutrient demands. Crucially, VSMCs may provide a sensitive biomarker for neurodegenerative pathologies where vasculature is compromised. An increasing body of research suggests that VSMCs have remarkable plasticity and their pathophysiology may play a key role in the complex process of neurodegeneration. Furthermore, extrinsic risk factors, including environmental conditions and traumatic events can impact vascular function through changes in VSMC morphology. VSMC dysfunction can be characterised at the molecular level both preclinically, and clinically ex vivo. However the identification of VSMC dysfunction in living individuals is important to understand changes in vascular function at the onset and progression of neurological disorders such as dementia, Alzheimer's disease, and Parkinson's disease. A promising technique to identify changes in the state of cerebral smooth muscle is cerebrovascular reactivity (CVR) which reflects the intrinsic dynamic response of blood vessels in the brain to vasoactive stimuli in order to modulate regional cerebral blood flow (CBF). In this work, we review the role of VSMCs in the most common neurodegenerative disorders and identify physiological systems that may contribute to VSMC dysfunction. The evidence collected here identifies VSMC dysfunction as a strong candidate for novel therapeutics to combat the development and progression of neurodegeneration, and highlights the need for more research on the role of VSMCs and cerebrovascular dynamics in healthy and diseased states.
Collapse
Affiliation(s)
- Genevieve Hayes
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Joana Pinto
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Sierra N. Sparks
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Congxiyu Wang
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Sana Suri
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Daniel P. Bulte
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Wang J, Li Z, Pan M, Fiaz M, Hao Y, Yan Y, Sun L, Yan F. Ultrasound-mediated blood-brain barrier opening: An effective drug delivery system for theranostics of brain diseases. Adv Drug Deliv Rev 2022; 190:114539. [PMID: 36116720 DOI: 10.1016/j.addr.2022.114539] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Blood-brain barrier (BBB) remains a significant obstacle to drug therapy for brain diseases. Focused ultrasound (FUS) combined with microbubbles (MBs) can locally and transiently open the BBB, providing a potential strategy for drug delivery across the BBB into the brain. Nowadays, taking advantage of this technology, many therapeutic agents, such as antibodies, growth factors, and nanomedicine formulations, are intensively investigated across the BBB into specific brain regions for the treatment of various brain diseases. Several preliminary clinical trials also have demonstrated its safety and good tolerance in patients. This review gives an overview of the basic mechanisms, ultrasound contrast agents, evaluation or monitoring methods, and medical applications of FUS-mediated BBB opening in glioblastoma, Alzheimer's disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Jieqiong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 201206, China
| | - Zhenzhou Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, China
| | - Min Pan
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Muhammad Fiaz
- Department of Radiology, Azra Naheed Medical College, Lahore, Pakistan
| | - Yongsheng Hao
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yiran Yan
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
50
|
Alfonsetti M, d’Angelo M, Castelli V. Neurotrophic factor-based pharmacological approaches in neurological disorders. Neural Regen Res 2022; 18:1220-1228. [PMID: 36453397 PMCID: PMC9838155 DOI: 10.4103/1673-5374.358619] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aging is a physiological event dependent on multiple pathways that are linked to lifespan and processes leading to cognitive decline. This process represents the major risk factor for aging-related diseases such as Alzheimer's disease, Parkinson's disease, and ischemic stroke. The incidence of all these pathologies increases exponentially with age. Research on aging biology has currently focused on elucidating molecular mechanisms leading to the development of those pathologies. Cognitive deficit and neurodegeneration, common features of aging-related pathologies, are related to the alteration of the activity and levels of neurotrophic factors, such as brain-derived neurotrophic factor, nerve growth factor, and glial cell-derived neurotrophic factor. For this reason, treatments that modulate neurotrophin levels have acquired a great deal of interest in preventing neurodegeneration and promoting neural regeneration in several neurological diseases. Those treatments include both the direct administration of neurotrophic factors and the induced expression with viral vectors, neurotrophins' binding with biomaterials or other molecules to increase their bioavailability but also cell-based therapies. Considering neurotrophins' crucial role in aging pathologies, here we discuss the involvement of several neurotrophic factors in the most common brain aging-related diseases and the most recent therapeutic approaches that provide direct and sustained neurotrophic support.
Collapse
Affiliation(s)
- Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy,Correspondence to: Vanessa Castelli, .
| |
Collapse
|