1
|
Kamatham PT, Shukla R, Khatri DK, Vora LK. Pathogenesis, diagnostics, and therapeutics for Alzheimer's disease: Breaking the memory barrier. Ageing Res Rev 2024; 101:102481. [PMID: 39236855 DOI: 10.1016/j.arr.2024.102481] [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: 05/06/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for 60-70 % of all cases. It affects millions of people worldwide. AD poses a substantial economic burden on societies and healthcare systems. AD is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. As the prevalence of AD continues to increase, understanding its pathogenesis, improving diagnostic methods, and developing effective therapeutics have become paramount. This comprehensive review delves into the intricate mechanisms underlying AD, explores the current state of diagnostic techniques, and examines emerging therapeutic strategies. By revealing the complexities of AD, this review aims to contribute to the growing body of knowledge surrounding this devastating disease.
Collapse
Affiliation(s)
- Pushpa Tryphena Kamatham
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rashi Shukla
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland BT9 7BL, UK.
| |
Collapse
|
2
|
Abdi SMY, Al-Bakri SSM, Nordin N. Insights on the Characteristics and Therapeutic Potential of Mesenchymal Stem Cell-derived Exosomes for Mitigation of Alzheimer's Disease's Pathogenicity: A Systematic Review. Cell Biochem Biophys 2024:10.1007/s12013-024-01598-x. [PMID: 39436580 DOI: 10.1007/s12013-024-01598-x] [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] [Accepted: 04/22/2024] [Indexed: 10/23/2024]
Abstract
Alzheimer's disease (AD) remains a progressive neurodegenerative disease with no cure. Treatment of AD relies on administering drugs that only subside the symptoms. In recent studies, mesenchymal stem cell (MSC)-exosomes have been marked to possess therapeutic potential for treating AD. This study aims to systematically review and analyse findings that focus on the isolation, characterisation, and sources of MSC-derived exosomes used to unravel the therapeutic potential of these exosomes targeting AD using in vitro and in vivo models. It is hypothesised that MSC-exosomes exhibit high therapeutic potential for AD treatment by exerting various modes of action. PubMed, Scopus, and Medline were used to find relevant published works from January 2016 until December 2020, using assigned keywords including "Alzheimer's disease", "secretome", and "exosomes". Only research articles meeting the predefined inclusion/exclusion criteria were selected and analysed. The risk of bias was assessed using the Office of Health Assessment and Translation tool (OHAT). A total of 17 eligible in vivo and in vitro studies were included in this review. Bone marrow-derived stem cells (BMSCs) were the most used source for exosome isolation, even though studies on exosomes from adipose-derived stem cells (ADSCs) and human umbilical cord stem cells (HUCSCs) provide more information on the characteristics. When the risk of bias was assessed, the studies presented various levels of biases. Notably, the in vitro and in vivo studies revealed neuroprotective properties of MSC-exosomes through different modes of action to alleviate AD pathology. Our review discovered that most MSC exosomes could degrade Aβ plaques, enhance neurogenesis, extenuate neuroinflammatory response through microglial activation, regulate apoptosis and reduce oxidative stress. Delivery of exosomal micro-RNAs was also found to reduce neuroinflammation. Findings from this review provided convincing systematic evidence highlighting the therapeutic properties of MSC-derived exosomes as a prospective source for cell-free (acellular) therapy in treating AD.
Collapse
Affiliation(s)
- Sarah Mohammed Yousuf Abdi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Siti Sarah Mustaffa Al-Bakri
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Norshariza Nordin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- Malaysian Research Institute on Ageing (MyAgeing™), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Genetics & Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
| |
Collapse
|
3
|
Honda Pazili T. A Severe Alzheimer's Disease Patient Improved by Intravenous Mesenchymal Stem Cell Transplant. Case Rep Neurol Med 2024; 2024:8353492. [PMID: 39040486 PMCID: PMC11262880 DOI: 10.1155/2024/8353492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/13/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder and is the most common form of dementia. The terminal stage of AD is characterized by severe cognitive and substantial functional decline, requiring extensive assistance with daily activities. As effective therapies at this stage are not fully available, development of therapeutics that can recover any symptoms would be important to improve the quality of life. Recently, stem cell therapy has gathered a lot of attention in several neurological diseases, including AD. Here, we report an AD patient at the terminal stage whose symptoms were improved by the intravenous administration of ex vivo-expanded bone marrow-derived mesenchymal stem cells (MSC). The case is a 61-year-old woman with severe Alzheimer's disease who had been admitted to the special nursing home. She could neither walk nor sit up independently. She also did neither smile nor gaze properly when talked to. Rigidity including neck motion was observed. She was on dysphagia diets. We cultured her bone-marrow-derived MSCs and intravenously administered 1,5 × 108 cells. After the treatment, smile loss, eye movement dysfunction, and neck immobility were improved. This is the first case report that showed the therapeutic effects of MSCs on terminal symptoms of AD.
Collapse
Affiliation(s)
- Takahiro Honda Pazili
- Regenerative MedicineDepartment of Cell TherapyJapan Tokyo Stem Cell Transplant Research Institute Ginza Clinic, Ginza 4-3-9, Chuo-ku, Tokyo 104-0067, Japan
| |
Collapse
|
4
|
Ataei B, Hokmabadi M, Asadi S, Asadifard E, Aghaei Zarch SM, Najafi S, Bagheri-Mohammadi S. A review of the advances, insights, and prospects of gene therapy for Alzheimer's disease: A novel target for therapeutic medicine. Gene 2024; 912:148368. [PMID: 38485038 DOI: 10.1016/j.gene.2024.148368] [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: 08/01/2023] [Revised: 02/24/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD) are still an important issue for scientists because it is difficult to cure with the available molecular medications and conventional treatments. Due to the complex nature of the brain structures and heterogeneous morphological and physiological properties of neuronal cells, interventions for cerebral-related disorders using surgical approaches, and classical and ongoing treatments remain hard for physicians. Furthermore, the development of newly designed medications attempts to target AD are not successful in improving AD, because abnormalities of tau protein, aggregation of amyloid β (Aβ) peptide, inflammatory responses, etc lead to advanced neurodegeneration processes that conventional treatments cannot stop them. In recent years, novel diagnostic strategies and therapeutic approaches have been developed to identify and cure early pathological events of AD. Accordingly, many gene-based therapies have been developed and introduce the therapeutic potential to prevent and cure AD. On the other hand, genetic investigations and postmortem assessments have detected a large number of factors associated with AD pathology. Also, genetically diverse animal models of AD help us to detect and prioritize novel resilience mechanisms. Hence, gene therapy can be considered an effective and powerful tool to identify and treat human diseases. Ultimately, gene study and gene-based therapy with a critical role in the detection and cure of various human disorders will have a fundamental role in our lives forever. This scientific review paper discusses the present status of different therapeutic strategies, particularly gene-based therapy in treating AD, along with its challenges.
Collapse
Affiliation(s)
- Bahar Ataei
- Department of Genetics, Faculty of Basic Science, Shahrekord University, Shahrekord, Iran
| | - Mahsa Hokmabadi
- Department of Molecular Diagnosis, Armin Pathobiology and Medical Genetics Laboratory, Tehran, Iran; Medical Genomics Research Center, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | - Sahar Asadi
- Department of Community and Family Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elnaz Asadifard
- Medical Genomics Research Center, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | - Seyed Mohsen Aghaei Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Bagheri-Mohammadi
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
5
|
Kozisek T, Samuelson L, Hamann A, Pannier AK. Systematic comparison of nonviral gene delivery strategies for efficient co-expression of two transgenes in human mesenchymal stem cells. J Biol Eng 2023; 17:76. [PMID: 38062439 PMCID: PMC10704746 DOI: 10.1186/s13036-023-00394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/20/2023] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Human mesenchymal stem cells (hMSCs) are being researched for cell-based therapies due to a host of unique properties, however, genetic modification of hMSCs, accomplished through nonviral gene delivery, could greatly advance their therapeutic potential. Furthermore, expression of multiple transgenes in hMSCs could greatly advance their clinical significance for treatment of multifaceted diseases, as individual transgenes could be expressed that target separate pathogenic drivers of complex diseases. Expressing multiple transgenes can be accomplished by delivering multiple DNA vectors encoding for each transgene, or by delivering a single poly-cistronic vector that encodes for each transgene and accomplishes expression through either use of multiple promoters, an internal ribosome entry site (IRES), or a 2A peptide sequence. These different transgene expression strategies have been used to express multiple transgenes in various mammalian cells, however, they have not been fully evaluated in difficult-to-transfect primary cells, like hMSCs. This study systematically compared four transgene expression and delivery strategies for expression of two reporter transgenes in four donors of hMSCs from two tissue sources using lipid- and polymer-mediate transfection, as follows: (i) delivery of separate DNA vectors in separate nanoparticles; (ii) delivery of separate DNA vectors combined in the same nanoparticle; (iii) delivery of a bi-cistronic DNA vector with an IRES sequence via nanoparticles; and (iv) delivery of a bi-cistronic DNA vector with a dual 2A peptide sequence via nanoparticles. RESULTS Our results indicate that expression of two transgenes in hMSCs, independent of expression or delivery strategy, is inefficient compared to expressing a single transgene. However, delivery of separate DNA vectors complexed in the same nanoparticle, or delivery of a bi-cistronic DNA vector with a dual 2A peptide sequence, significantly increased the number of hMSCs expressing both transgenes compared to other conditions tested. CONCLUSION Separate DNA vectors delivered in the same nanoparticle and bi-cistronic DNA vectors with dual 2A peptide sequences are highly efficient at simultaneously expressing two transgenes in multiple donors of hMSCs from different tissue sources. The data presented in this work can guide the development of hMSC transfection systems for delivery of multiple transgenes, with the goal of producing clinically relevant, genetically modified hMSCs.
Collapse
Affiliation(s)
- Tyler Kozisek
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583-0900, USA
| | - Luke Samuelson
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583-0900, USA
| | - Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583-0900, USA
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68583-0900, USA.
| |
Collapse
|
6
|
Zhou AK, Jou E, Lu V, Zhang J, Chabra S, Abishek J, Wong E, Zeng X, Guo B. Using Pre-Clinical Studies to Explore the Potential Clinical Uses of Exosomes Secreted from Induced Pluripotent Stem Cell-Derived Mesenchymal Stem cells. Tissue Eng Regen Med 2023; 20:793-809. [PMID: 37651091 PMCID: PMC10519927 DOI: 10.1007/s13770-023-00557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 09/01/2023] Open
Abstract
Recent studies of exosomes derived from mesenchymal stem cells (MSCs) have indicated high potential clinical applications in many diseases. However, the limited source of MSCs impedes their clinical research and application. Most recently, induced pluripotent stem cells (iPSCs) have become a promising source of MSCs. Exosome therapy based on iPSC-derived MSCs (iMSCs) is a novel technique with much of its therapeutic potential untapped. Compared to MSCs, iMSCs have proved superior in cell proliferation, immunomodulation, generation of exosomes capable of controlling the microenvironment, and bioactive paracrine factor secretion, while also theoretically eliminating the dependence on immunosuppression drugs. The therapeutic effects of iMSC-derived exosomes are explored in many diseases and are best studied in wound healing, cardiovascular disease, and musculoskeletal pathology. It is pertinent clinicians have a strong understanding of stem cell therapy and the latest advances that will eventually translate into clinical practice. In this review, we discuss the various applications of exosomes derived from iMSCs in clinical medicine.
Collapse
Affiliation(s)
- Andrew Kailin Zhou
- Addenbrookes Major Trauma Unit, Department of Trauma And Orthopaedics, Cambridge University Hospitals, Cambridge, UK
- Watford General Hospital, London, UK
| | - Eric Jou
- Addenbrookes Major Trauma Unit, Department of Trauma And Orthopaedics, Cambridge University Hospitals, Cambridge, UK
- School Of Clinical Medicine, University Of Cambridge, Cambridge, UK
| | - Victor Lu
- Addenbrookes Major Trauma Unit, Department of Trauma And Orthopaedics, Cambridge University Hospitals, Cambridge, UK
- School Of Clinical Medicine, University Of Cambridge, Cambridge, UK
| | - James Zhang
- Addenbrookes Major Trauma Unit, Department of Trauma And Orthopaedics, Cambridge University Hospitals, Cambridge, UK
- School Of Clinical Medicine, University Of Cambridge, Cambridge, UK
| | - Shirom Chabra
- School Of Clinical Medicine, University Of Cambridge, Cambridge, UK
| | | | | | - Xianwei Zeng
- Beijing Rehabilitation Hospital Affiliated to National Research Centre for Rehabilitation Technical Aids, Ministry of Civil Affairs of China, Beijing, China.
- Weifang People's Hospital, Weifang City, Shandong Province, China.
| | - Baoqiang Guo
- Department of Life Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK.
| |
Collapse
|
7
|
Duan Y, Lyu L, Zhan S. Stem Cell Therapy for Alzheimer's Disease: A Scoping Review for 2017-2022. Biomedicines 2023; 11:biomedicines11010120. [PMID: 36672626 PMCID: PMC9855936 DOI: 10.3390/biomedicines11010120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) has been a major causal factor for mortality among elders around the world. The treatments for AD, however, are still in the stage of development. Stem cell therapy, compared to drug therapies and many other therapeutic options, has many advantages and is very promising in the future. There are four major types of stem cells used in AD therapy: neural stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells. All of them have applications in the treatments, either at the (1) cellular level, in an (2) animal model, or at the (3) clinical level. In general, many more types of stem cells were studied on the cellular level and animal model, than the clinical level. We suggest for future studies to increase research on various types of stem cells and include cross-disciplinary research with other diseases. In the future, there could also be improvements in the timeliness of research and individualization for stem cell therapies for AD.
Collapse
Affiliation(s)
- Yunxiao Duan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Linshuoshuo Lyu
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA
| | - Siyan Zhan
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
- Correspondence:
| |
Collapse
|
8
|
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
|
9
|
Abdi S, Javanmehr N, Ghasemi-Kasman M, Bali HY, Pirzadeh M. Stem Cell-based Therapeutic and Diagnostic Approaches in Alzheimer's Disease. Curr Neuropharmacol 2022; 20:1093-1115. [PMID: 34970956 PMCID: PMC9886816 DOI: 10.2174/1570159x20666211231090659] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/24/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative impairment mainly recognized by memory loss and cognitive deficits. However, the current therapies against AD are mostly limited to palliative medications, prompting researchers to investigate more efficient therapeutic approaches for AD, such as stem cell therapy. Recent evidence has proposed that extensive neuronal and synaptic loss and altered adult neurogenesis, which is perceived pivotal in terms of plasticity and network maintenance, occurs early in the course of AD, which exacerbates neuronal vulnerability to AD. Thus, regeneration and replenishing the depleted neuronal networks by strengthening the endogenous repair mechanisms or exogenous stem cells and their cargoes is a rational therapeutic approach. Currently, several stem cell-based therapies as well as stem cell products like exosomes, have shown promising results in the early diagnosis of AD. OBJECTIVE This review begins with a comparison between AD and normal aging pathophysiology and a discussion on open questions in the field. Next, summarizing the current stem cell-based therapeutic and diagnostic approaches, we declare the advantages and disadvantages of each method. Also, we comprehensively evaluate the human clinical trials of stem cell therapies for AD. METHODOLOGY Peer-reviewed reports were extracted through Embase, PubMed, and Google Scholar until 2021. RESULTS With several ongoing clinical trials, stem cells and their derivatives (e.g., exosomes) are an emerging and encouraging field in diagnosing and treating neurodegenerative diseases. Although stem cell therapies have been successful in animal models, numerous clinical trials in AD patients have yielded unpromising results, which we will further discuss.
Collapse
Affiliation(s)
- Sadaf Abdi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran;,Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran,Address correspondence to this author at the Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, P.O. Box 4136747176, Babol, Iran; Tel/Fax: +98-11-32190557; E-mail:
| | | | - Marzieh Pirzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
10
|
Zahedipour F, Hosseini SA, Henney NC, Barreto GE, Sahebkar A. Phytochemicals as inhibitors of tumor necrosis factor alpha and neuroinflammatory responses in neurodegenerative diseases. Neural Regen Res 2022; 17:1675-1684. [PMID: 35017414 PMCID: PMC8820712 DOI: 10.4103/1673-5374.332128] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
Inflammatory processes and proinflammatory cytokines have a key role in the cellular processes of neurodegenerative diseases and are linked to the pathogenesis of functional and mental health disorders. Tumor necrosis factor alpha has been reported to play a major role in the central nervous system in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis and many other neurodegenerative diseases. Therefore, a potent proinflammatory/proapoptotic tumor necrosis factor alpha could be a strong candidate for targeted therapy. Plant derivatives have now become promising candidates as therapeutic agents because of their antioxidant and chemical characteristics, and anti-inflammatory features. Recently, phytochemicals including flavonoids, terpenoids, alkaloids, and lignans have generated interest as tumor necrosis factor alpha inhibitor candidates for a number of diseases involving inflammation within the nervous system. In this review, we discuss how phytochemicals as tumor necrosis factor alpha inhibitors are a therapeutic strategy targeting neurodegeneration.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology, School of Medicine; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Atefe Hosseini
- Department of Medical Biotechnology, School of Medicine; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neil C Henney
- Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - George E Barreto
- Department of Biological Sciences; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Campos HC, Ribeiro DE, Hashiguchi D, Hukuda DY, Gimenes C, Romariz SAA, Ye Q, Tang Y, Ulrich H, Longo BM. Distinct Effects of the Hippocampal Transplantation of Neural and Mesenchymal Stem Cells in a Transgenic Model of Alzheimer's Disease. Stem Cell Rev Rep 2022; 18:781-791. [PMID: 34997526 DOI: 10.1007/s12015-021-10321-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a severe disabling condition with no cure currently available, which accounts for 60-70% of all dementia cases worldwide. Therefore, the investigation of possible therapeutic strategies for AD is necessary. To this end, animal models corresponding to the main aspects of AD in humans have been widely used. Similar to AD patients, the double transgenic APPswe/PS1dE9 (APP/PS1) mice show cognitive deficits, hyperlocomotion, amyloid-β (Αβ) plaques in the cortex and hippocampus, and exacerbated inflammatory responses. Recent studies have shown that these neuropathological features could be reversed by stem cell transplantation. However, the effects induced by neural (NSC) and mesenchymal (MSC) stem cells has never been compared in an AD animal model. Therefore, the present study aimed to investigate whether transplantation of NSC or MSC into the hippocampus of APP/PS1 mice reverses AD-induced pathological alterations, evaluated by the locomotor activity (open field test), short- and long-term memory (object recognition) tests, Αβ plaques (6-E10), microglia distribution (Iba-1), M1 (iNOS) and M2 (ARG-1) microglial phenotype frequencies. NSC and MSC engraftment reduced the number of Αβ plaques and produced an increase in M2 microglia polarization in the hippocampus of APP/PS1 mice, suggesting an anti-inflammatory effect of stem cell transplantation. NSC also reversed the hyperlocomotor activity and increased the number of microglia in the hippocampus of APP/PS1 mice. No impairment of short or long-term memory was observed in APP/PS1 mice. Overall, this study highlights the potential beneficial effects of transplanting NSC or MSC for AD treatment.
Collapse
Affiliation(s)
- Henrique C Campos
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Debora Hashiguchi
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.,Laboratório de Plasticidade Sináptica, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Caixa Postal: 1524, Brazil
| | - Deborah Y Hukuda
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Christiane Gimenes
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Simone A A Romariz
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Qing Ye
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.,International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Yong Tang
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.,International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Beatriz Monteiro Longo
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
12
|
Kozisek T, Hamann A, Samuelson L, Fudolig M, Pannier AK. Comparison of promoter, DNA vector, and cationic carrier for efficient transfection of hMSCs from multiple donors and tissue sources. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:81-93. [PMID: 34513295 PMCID: PMC8413668 DOI: 10.1016/j.omtn.2021.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/25/2021] [Indexed: 01/22/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are primary cells with high clinical relevance that could be enhanced through genetic modification. However, gene delivery, particularly through nonviral routes, is inefficient. To address the shortcomings of nonviral gene delivery to hMSCs, our lab has previously demonstrated that pharmacological "priming" of hMSCs with clinically approved drugs can increase transfection in hMSCs by modulating transfection-induced cytotoxicity. However, even with priming, hMSC transfection remains inefficient for clinical applications. This work takes a complementary approach to addressing the challenges of transfecting hMSCs by systematically investigating key transfection parameters for their effect on transgene expression. Specifically, we investigated two promoters (cytomegalovirus [CMV] and elongation factor 1 alpha), four DNA vectors (plasmid, plasmid with no F1 origin, minicircle, and mini-intronic plasmid), two cationic carriers (Lipofectamine 3000 and Turbofect), and four donors of hMSCs from two tissues (adipose and bone marrow) for efficient hMSC transfection. Following systematic comparison of each variable, we identified adipose-derived hMSCs transfected with mini-intronic plasmids containing the CMV promoter delivered using Lipofectamine 3000 as the parameters that produced the highest transfection levels. The data presented in this work can guide the development of other hMSC transfection systems with the goal of producing clinically relevant, genetically modified hMSCs.
Collapse
Affiliation(s)
- Tyler Kozisek
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrew Hamann
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Luke Samuelson
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Miguel Fudolig
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Angela K. Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
13
|
Alastra G, Aloe L, Baldassarro VA, Calzà L, Cescatti M, Duskey JT, Focarete ML, Giacomini D, Giardino L, Giraldi V, Lorenzini L, Moretti M, Parmeggiani I, Sannia M, Tosi G. Nerve Growth Factor Biodelivery: A Limiting Step in Moving Toward Extensive Clinical Application? Front Neurosci 2021; 15:695592. [PMID: 34335170 PMCID: PMC8319677 DOI: 10.3389/fnins.2021.695592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022] Open
Abstract
Nerve growth factor (NGF) was the first-discovered member of the neurotrophin family, a class of bioactive molecules which exerts powerful biological effects on the CNS and other peripheral tissues, not only during development, but also during adulthood. While these molecules have long been regarded as potential drugs to combat acute and chronic neurodegenerative processes, as evidenced by the extensive data on their neuroprotective properties, their clinical application has been hindered by their unexpected side effects, as well as by difficulties in defining appropriate dosing and administration strategies. This paper reviews aspects related to the endogenous production of NGF in healthy and pathological conditions, along with conventional and biomaterial-assisted delivery strategies, in an attempt to clarify the impediments to the clinical application of this powerful molecule.
Collapse
Affiliation(s)
- Giuseppe Alastra
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
| | | | - Vito Antonio Baldassarro
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- IRET Foundation, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Jason Thomas Duskey
- Nanotech Laboratory, TeFarTI Center, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Letizia Focarete
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Daria Giacomini
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Luciana Giardino
- IRET Foundation, Bologna, Italy
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Valentina Giraldi
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Bologna, Italy
| | - Luca Lorenzini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | | | - Irene Parmeggiani
- Nanotech Laboratory, TeFarTI Center, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Michele Sannia
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
| | - Giovanni Tosi
- Nanotech Laboratory, TeFarTI Center, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
14
|
Meldolesi J. Extracellular vesicles (exosomes and ectosomes) play key roles in the pathology of brain diseases. MOLECULAR BIOMEDICINE 2021; 2:18. [PMID: 35006460 PMCID: PMC8607397 DOI: 10.1186/s43556-021-00040-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Last century, neurons and glial cells were mostly believed to play distinct functions, relevant for the brain. Progressively, however, it became clear that neurons, astrocytes and microglia co-operate intensely with each other by release/binding of signaling factors, direct surface binding and generation/release of extracellular vesicles, the exosomes and ectosomes, called together vesicles in this abstract. The present review is focused on these vesicles, fundamental in various brain diseases. Their properties are extraordinary. The specificity of their membrane governs their fusion with distinct target cells, variable depending on the state and specificity of their cells of origin and target. Result of vesicle fusion is the discharge of their cargos into the cytoplasm of target cells. Cargos are composed of critical molecules, from proteins (various nature and function) to nucleotides (especially miRNAs), playing critical roles in immune and neurodegenerative diseases. Among immune diseases is multiple sclerosis, affected by extensive dysregulation of co-trafficking neural and glial vesicles, with distinct miRNAs inducing severe or reducing effects. The vesicle-dependent differences between progressive and relapsing-remitting forms of the disease are relevant for clinical developments. In Alzheimer’s disease the vesicles can affect the brain by changing their generation and inducing co-release of effective proteins, such Aβ and tau, from neurons and astrocytes. Specific miRNAs can delay the long-term development of the disease. Upon their traffic through the blood-brainbarrier, vesicles of various origin reach fluids where they are essential for the identification of biomarkers, important for diagnostic and therapeutic innovations, critical for the future of many brain patients.
Collapse
Affiliation(s)
- Jacopo Meldolesi
- Division of Neuroscience, San Raffaele Institute and Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
15
|
The Protective A673T Mutation of Amyloid Precursor Protein (APP) in Alzheimer's Disease. Mol Neurobiol 2021; 58:4038-4050. [PMID: 33914267 DOI: 10.1007/s12035-021-02385-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/05/2021] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder characterized by extracellular amyloid beta peptides and neurofibrillary tangles consisted of intracellular hyperphosphorylated Tau in the hippocampus and cerebral cortex. Most of the mutations in key genes that code for amyloid precursor protein can lead to significant accumulation of these peptides in the brain and cause Alzheimer's disease. Moreover, some point mutations in amyloid precursor protein can cause familial Alzheimer's disease, such as Swedish mutation (KM670/671NL) and A673V mutation. However, recent studies have found that the A673T mutation in amyloid precursor protein gene can protect against Alzheimer's disease, even if it is located next to the Swedish mutation (KM670/671NL) and at the same site as A673V mutation, which are pathogenic. It makes us curious about the protective A673T mutation. Here, we summarize the most recent insights of A673T mutation, focus on their roles in protective mechanisms against Alzheimer's disease, and discuss their involvement in future treatment.
Collapse
|
16
|
Loeffler DA. Modifiable, Non-Modifiable, and Clinical Factors Associated with Progression of Alzheimer's Disease. J Alzheimers Dis 2021; 80:1-27. [PMID: 33459643 DOI: 10.3233/jad-201182] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There is an extensive literature relating to factors associated with the development of Alzheimer's disease (AD), but less is known about factors which may contribute to its progression. This review examined the literature with regard to 15 factors which were suggested by PubMed search to be positively associated with the cognitive and/or neuropathological progression of AD. The factors were grouped as potentially modifiable (vascular risk factors, comorbidities, malnutrition, educational level, inflammation, and oxidative stress), non-modifiable (age at clinical onset, family history of dementia, gender, Apolipoprotein E ɛ4, genetic variants, and altered gene regulation), and clinical (baseline cognitive level, neuropsychiatric symptoms, and extrapyramidal signs). Although conflicting results were found for the majority of factors, a positive association was found in nearly all studies which investigated the relationship of six factors to AD progression: malnutrition, genetic variants, altered gene regulation, baseline cognitive level, neuropsychiatric symptoms, and extrapyramidal signs. Whether these or other factors which have been suggested to be associated with AD progression actually influence the rate of decline of AD patients is unclear. Therapeutic approaches which include addressing of modifiable factors associated with AD progression should be considered.
Collapse
Affiliation(s)
- David A Loeffler
- Beaumont Research Institute, Department of Neurology, Beaumont Health, Royal Oak, MI, USA
| |
Collapse
|
17
|
Kopach O, Pavlov AM, Sindeeva OA, Sukhorukov GB, Rusakov DA. Biodegradable Microcapsules Loaded with Nerve Growth Factor Enable Neurite Guidance and Synapse Formation. Pharmaceutics 2020; 13:E25. [PMID: 33375672 PMCID: PMC7823884 DOI: 10.3390/pharmaceutics13010025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders and traumas often involve loss of specific neuronal connections, which would require intervention with high spatial precision. We have previously demonstrated the biocompatibility and therapeutic potential of the layer-by-layer (LbL)-fabricated microcapsules aimed at the localized delivery of specific channel blockers to peripheral nerves. Here, we explore the potential of LbL-microcapsules to enable site-specific, directional action of neurotrophins to stimulate neuronal morphogenesis and synaptic circuit formation. We find that nanoengineered biodegradable microcapsules loaded with nerve growth factor (NGF) can guide the morphological development of hippocampal neurons in vitro. The presence of NGF-loaded microcapsules or their clusters increases the neurite outgrowth rate while boosting neurite branching. Microcapsule clusters appear to guide the trajectory of developing individual axons leading to the formation of functional synapses. Our observations highlight the potential of NGF-loaded, biodegradable LbL-microcapsules to help guide axonal development and possibly circuit regeneration in neuropathology.
Collapse
Affiliation(s)
- Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Anton M. Pavlov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; (A.M.P.); (O.A.S.)
- Remote Controlled Theranostic Systems Laboratory, Saratov State University, 83 Astrakhanskaya Street, 410012 Saratov, Russia
| | - Olga A. Sindeeva
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; (A.M.P.); (O.A.S.)
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 3 Nobel Street, 143005 Moscow, Russia
| | - Gleb B. Sukhorukov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; (A.M.P.); (O.A.S.)
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 3 Nobel Street, 143005 Moscow, Russia
| | - Dmitri A. Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
18
|
Chang CC, Li HH, Tsou SH, Hung HC, Liu GY, Korolenko TA, Lai TJ, Ho YJ, Lin CL. The Pluripotency Factor Nanog Protects against Neuronal Amyloid β-Induced Toxicity and Oxidative Stress through Insulin Sensitivity Restoration. Cells 2020; 9:cells9061339. [PMID: 32471175 PMCID: PMC7348813 DOI: 10.3390/cells9061339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 11/16/2022] Open
Abstract
Amyloid β (Aβ) is a peptide fragment of the amyloid precursor protein that triggers the progression of Alzheimer's Disease (AD). It is believed that Aβ contributes to neurodegeneration in several ways, including mitochondria dysfunction, oxidative stress and brain insulin resistance. Therefore, protecting neurons from Aβ-induced neurotoxicity is an effective strategy for attenuating AD pathogenesis. Recently, applications of stem cell-based therapies have demonstrated the ability to reduce the progression and outcome of neurodegenerative diseases. Particularly, Nanog is recognized as a stem cell-related pluripotency factor that enhances self-renewing capacities and helps reduce the senescent phenotypes of aged neuronal cells. However, whether the upregulation of Nanog can be an effective approach to alleviate Aβ-induced neurotoxicity and senescence is not yet understood. In the present study, we transiently overexpressed Nanog-both in vitro and in vivo-and investigated the protective effects and underlying mechanisms against Aβ. We found that overexpression of Nanog is responsible for attenuating Aβ-triggered neuronal insulin resistance, which restores cell survival through reducing intracellular mitochondrial superoxide accumulation and cellular senescence. In addition, upregulation of Nanog expression appears to increase secretion of neurotrophic factors through activation of the Nrf2 antioxidant defense pathway. Furthermore, improvement of memory and learning were also observed in rat model of Aβ neurotoxicity mediated by upregulation of Nanog in the brain. Taken together, our study suggests a potential role for Nanog in attenuating the neurotoxic effects of Aβ, which in turn, suggests that strategies to enhance Nanog expression may be used as a novel intervention for reducing Aβ neurotoxicity in the AD brain.
Collapse
Affiliation(s)
- Ching-Chi Chang
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
| | - Hsin-Hua Li
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
| | - Sing-Hua Tsou
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
| | - Hui-Chih Hung
- Department of Life Sciences and Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402204, Taiwan;
| | - Guang-Yaw Liu
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
| | - Tatiana A. Korolenko
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk 630117, Russia;
| | - Te-Jen Lai
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University, Taichung 402367, Taiwan
- Correspondence: (Y.-J.H.); (C.-L.L.); Tel.: +886-4-2473-0022-11673 (Y.-J.H.); +886-4-2473-0022-11690 (C.-L.L.)
| | - Chih-Li Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402367, Taiwan; (C.-C.C.); (H.-H.L.); (S.-H.T.); (G.-Y.L.); (T.-J.L.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402367, Taiwan
- Correspondence: (Y.-J.H.); (C.-L.L.); Tel.: +886-4-2473-0022-11673 (Y.-J.H.); +886-4-2473-0022-11690 (C.-L.L.)
| |
Collapse
|
19
|
Bahrami A, Fereidouni M, Pirro M, Bianconi V, Sahebkar A. Modulation of regulatory T cells by natural products in cancer. Cancer Lett 2019; 459:72-85. [DOI: 10.1016/j.canlet.2019.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
|
20
|
Mardani R, Jafari Najaf Abadi MH, Motieian M, Taghizadeh-Boroujeni S, Bayat A, Farsinezhad A, Gheibi Hayat SM, Motieian M, Pourghadamyari H. MicroRNA in leukemia: Tumor suppressors and oncogenes with prognostic potential. J Cell Physiol 2018; 234:8465-8486. [PMID: 30515779 DOI: 10.1002/jcp.27776] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
Leukemia is known as a progressive malignant disease, which destroys the blood-forming organs and results in adverse effects on the proliferation and development of leukocytes and their precursors in the blood and bone marrow. There are four main classes of leukemia including acute leukemia, chronic leukemia, myelogenous leukemia, and lymphocytic leukemia. Given that a variety of internal and external factors could be associated with the initiation and progression of different types of leukemia. One of the important factors is epigenetic regulators such as microRNAs (miRNAs) and long noncoding RNAs (ncRNA). MiRNAs are short ncRNAs which act as tumor suppressor (i.e., miR-15, miR-16, let-7, and miR-127) or oncogene (i.e., miR-155, miR-17-92, miR-21, miR-125b, miR-93, miR-143-p3, miR-196b, and miR-223) in leukemia. It has been shown that deregulation of these molecules are associated with the initiation and progression of leukemia. Hence, miRNAs could be used as potential therapeutic candidates in the treatment of patients with leukemia. Moreover, increasing evidence revealed that miRNAs could be used as diagnostic and prognostic biomarkers in monitoring patients in early stages of disease or after received chemotherapy regimen. It seems that identification and development of new miRNAs could pave to the way to the development new therapeutic platforms for patients with leukemia. Here, we summarized various miRNAs as tumor suppressor and oncogene which could be introduced as therapeutic targets in treatment of leukemia.
Collapse
Affiliation(s)
- Rajab Mardani
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | | | - Mahsa Motieian
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sima Taghizadeh-Boroujeni
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Borujen, Iran
| | - Amir Bayat
- Hematology, Oncology, and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Cell and Molecular Biology, College of Science, Kish International Campus, University of Tehran, Kish, Iran
| | - Alireza Farsinezhad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mahtab Motieian
- Department of Internal Medicine, Montefiore New Rochelle Hospital, Albert Einstein College of Medicine, New York, New York
| | - Hossein Pourghadamyari
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
21
|
Nejati M, Tameh AA, Atlasi MA. Role of toll‐like receptors 2 and 4 in the neuroprotective effects of bone marrow–derived mesenchymal stem cells in an experimental model of ischemic stroke. J Cell Biochem 2018. [DOI: 10.1002/jcb.28083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Majid Nejati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences Kashan Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences Kashan Iran
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences Kashan Iran
| |
Collapse
|