1
|
Hajivalili M, Nikkhoo N, Salahi S, Hosseini M. Traumatic brain Injury: Comprehensive overview from pathophysiology to Mesenchymal stem Cell-Based therapies. Int Immunopharmacol 2025; 146:113816. [PMID: 39708488 DOI: 10.1016/j.intimp.2024.113816] [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: 09/23/2024] [Revised: 11/16/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
Traumatic brain injury (TBI) is a disastrous phenomenon which is considered to cause high mortality and morbidity rate. Regarding the importance of TBI due to its prevalence and its effects on the brain and other organs, finding new therapeutic methods and improvement of conventional therapies seems to be vital. TBI involves a complex physiological mechanism, with inflammation being a key component among various contributing factors. After incidence of TBI, inflammation can act as a double-edged sword in the process. Inflammation actually plays its role both as initiator and progressive index during TBI which can accumulate myeloid and lymphoid immune cells and trigger cell death pathways. Through this study we made this concept bold that that besides conventional therapies that could be used for traumatic brain injury, treatments based on mesenchyme stem cells (MSCs) and their derivatives including secretomes and exosomes demonstrate more efficacies particularly in preventing secondary injuries caused by TBI. Of note, we highlighted the valuable features of MSC-based therapies such as self-direction toward inflamed tissues and amplifying neuro-regenerative aspects. We listed possible challenges in the way of reaching this therapy to clinic to provide a clear and updated of the field.
Collapse
Affiliation(s)
- Mahsa Hajivalili
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nima Nikkhoo
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sarvenaz Salahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Hosseini
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Chen Q, Wang Z, Zhang S. Mechanism, application and effect evaluation of nerve mobilization in the treatment of low back pain: A narrative review. Medicine (Baltimore) 2023; 102:e34961. [PMID: 37653794 PMCID: PMC10470699 DOI: 10.1097/md.0000000000034961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/14/2023] [Accepted: 08/04/2023] [Indexed: 09/02/2023] Open
Abstract
Lower back pain is a prevalent condition affecting people across all age groups and causing significant personal and societal burdens. While numerous treatments exist, nerve mobilization has emerged as a promising approach for managing lower back pain. Nerve mobilization involves applying gentle and rhythmic movements to the affected nerves, promoting normal nerve function and releasing tension. It has been well documented that nerve mobilization can be effective in reducing pain and improving function in patients with lower back pain, but the underlying mechanisms have not been clarified. This study aims to review the mechanisms of nerve mobilization in the management of lower back pain, its application, and effectiveness evaluation, and provide a potential solution for managing lower back pain.
Collapse
Affiliation(s)
- Quanzheng Chen
- Department of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Zhenshan Wang
- Department of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Shuna Zhang
- Department of Physical Education and Health, Guangxi Normal University, Guilin, China
| |
Collapse
|
3
|
Kim J, Kim J, Park HJ, Jeon EJ, Cho SW. A microfluidic platform for simulating stem cell migration using in vivo-like gradients of stem cell mobilizer. KOREAN J CHEM ENG 2023. [DOI: 10.1007/s11814-023-1390-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
4
|
Liu WS, Liu Y, Gao J, Zheng H, Lu ZM, Li M. Biomembrane-Based Nanostructure- and Microstructure-Loaded Hydrogels for Promoting Chronic Wound Healing. Int J Nanomedicine 2023; 18:385-411. [PMID: 36703725 PMCID: PMC9871051 DOI: 10.2147/ijn.s387382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Wound healing is a complex and dynamic process, and metabolic disturbances in the microenvironment of chronic wounds and the severe symptoms they cause remain major challenges to be addressed. The inherent properties of hydrogels make them promising wound dressings. In addition, biomembrane-based nanostructures and microstructures (such as liposomes, exosomes, membrane-coated nanostructures, bacteria and algae) have significant advantages in the promotion of wound healing, including special biological activities, flexible drug loading and targeting. Therefore, biomembrane-based nanostructure- and microstructure-loaded hydrogels can compensate for their respective disadvantages and combine the advantages of both to significantly promote chronic wound healing. In this review, we outline the loading strategies, mechanisms of action and applications of different types of biomembrane-based nanostructure- and microstructure-loaded hydrogels in chronic wound healing.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Yu Liu
- Department of Gastroenterology, Jinling Hospital, Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Hao Zheng
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Zheng-Mao Lu
- Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China,Zheng-Mao Lu, Department of General Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China, Tel +086-13651688596, Fax +086-021-31161589, Email
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China,Correspondence: Meng Li, Department of Dermatology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, People’s Republic of China, Tel +086-15000879978, Fax +086-021-23271699, Email
| |
Collapse
|
5
|
Ghozy S, El-Qushayri AE, Varney J, Kacimi SEO, Bahbah EI, Morra ME, Shah J, Kallmes KM, Abbas AS, Elfil M, Alghamdi BS, Ashraf G, Alhabbab R, Dmytriw AA. The prognostic value of neutrophil-to-lymphocyte ratio in patients with traumatic brain injury: A systematic review. Front Neurol 2022; 13:1021877. [PMID: 36353130 PMCID: PMC9638118 DOI: 10.3389/fneur.2022.1021877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) places a heavy load on healthcare systems worldwide. Despite significant advancements in care, the TBI-related mortality is 30–50% and in most cases involves adolescents or young adults. Previous literature has suggested that neutrophil-to-lymphocyte ratio (NLR) may serve as a sensitive biomarker in predicting clinical outcomes following TBI. With conclusive evidence in this regard lacking, this study aimed to systematically review all original studies reporting the effectiveness of NLR as a predictor of TBI outcomes. A systematic search of eight databases was conducted according to the Preferred Reporting Items for Systematic Review and Meta-Analyses statement (PRISMA) recommendations. The risk of bias was assessed using the Quality in Prognostic Studies (QUIPS) tool. Eight studies were ultimately included in the study. In most of the studies interrogated, severity outcomes were successfully predicted by NLR in both univariate and multivariate prediction models, in different follow-up durations up to 6 months. A high NLR at 24 and 48 h after TBI in pediatric patients was associated with worse clinical outcomes. On pooling the NLR values within studies assessing its association with the outcome severity (favorable or not), patients with favorable outcomes had 37% lower NLR values than those with unfavorable ones (RoM= 0.63; 95% CI = 0.44–0.88; p = 0.007). However, there were considerable heterogeneity in effect estimates (I2 = 99%; p < 0.001). Moreover, NLR was a useful indicator of mortality at both 6-month and 1-year intervals. In conjunction with clinical and radiographic parameters, NLR might be a useful, inexpensive marker in predicting clinical outcomes in patients with TBI. However, the considerable heterogeneity in current literature keeps it under investigation with further studies are warranted to confirm the reliability of NLR in predicting TBI outcomes.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States
- Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC Program), Oxford University, Oxford, United Kingdom
- *Correspondence: Sherief Ghozy
| | | | - Joseph Varney
- School of Medicine, American University of the Caribbean, Philipsburg, Sint Maarten
| | | | | | | | - Jaffer Shah
- Drexel University College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Kevin M. Kallmes
- Nested Knowledge, Saint Paul, MN, United States
- Superior Medical Experts, Saint Paul, MN, United States
| | | | - Mohamed Elfil
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Badrah S. Alghamdi
- Neuroscience Unit, Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Ghulam Ashraf
| | - Rowa Alhabbab
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adam A. Dmytriw
- Neurointerventional Program, Departments of Medical Imaging and Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON, Canada
- Neuroendovascular Program, Massachusetts General Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Pischiutta F, Caruso E, Cavaleiro H, Salgado AJ, Loane DJ, Zanier ER. Mesenchymal stromal cell secretome for traumatic brain injury: Focus on immunomodulatory action. Exp Neurol 2022; 357:114199. [PMID: 35952763 DOI: 10.1016/j.expneurol.2022.114199] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/14/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022]
Abstract
The severity and long-term consequences of brain damage in traumatic brain injured (TBI) patients urgently calls for better neuroprotective/neuroreparative strategies for this devastating disorder. Mesenchymal stromal cells (MSCs) hold great promise and have been shown to confer neuroprotection in experimental TBI, mainly through paracrine mechanisms via secreted bioactive factors (i.e. secretome), which indicates significant potential for a cell-free neuroprotective approach. The secretome is composed of cytokines, chemokines, growth factors, proteins, lipids, nucleic acids, metabolites, and extracellular vesicles; it may offer advantages over MSCs in terms of delivery, safety, and variability of therapeutic response for brain injury. Immunomodulation by molecular factors secreted by MSCs is considered to be a key mechanism involved in their multi-potential therapeutic effects. Regulated neuroinflammation is required for healthy remodeling of central nervous system during development and adulthood. Moreover, immune cells and their secreted factors can also contribute to tissue repair and neurological recovery following acute brain injury. However, a chronic and maladaptive neuroinflammatory response can exacerbate TBI and contribute to progressive neurodegeneration and long-term neurological impairments. Here, we review the evidence for MSC-derived secretome as a therapy for TBI. Our framework incorporates a detailed analysis of in vitro and in vivo studies investigating the effects of the secretome on clinically relevant neurological and histopathological outcomes. We also describe the activation of immune cells after TBI and the immunomodulatory properties exerted by mediators released in the secretome. We then describe how ageing modifies central and systemic immune responses to TBI and discuss challenges and opportunities of developing secretome based neuroprotective therapies for elderly TBI populations. Finally, strategies aimed at modulating the secretome in order to boost its efficacy for TBI will also be discussed.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| | - Enrico Caruso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy; Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Helena Cavaleiro
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; Stemmatters, Biotechnology and Regenerative Medicine, Guimarães, Portugal
| | - Antonio J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Elisa R Zanier
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy.
| |
Collapse
|
7
|
Sachdev KR, Lynch KJ, Barreto GE. Exploration of novel ligands to target C-C Motif Chemokine Receptor 2 (CCR2) as a promising pharmacological treatment against traumatic brain injury. Biomed Pharmacother 2022; 151:113155. [PMID: 35598371 DOI: 10.1016/j.biopha.2022.113155] [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: 04/04/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/02/2022] Open
Abstract
It is widely reported that the overexpression of the C-C Motif Chemokine Receptor 2 (CCR2) has negative implications in neuroinflammatory diseases such as traumatic brain injury (TBI), although promising drugs to tackle this have been less forthcoming. As of 2016, only 2 drugs specifically targeting this receptor have made their way to market, with unsuccessful outcome unfortunately, suggesting that the search for more specific and precise ligands is utterly necessary. In this paper we hypothesized that by targeting Glu291, Met295, Trp98, Leu45 and Val189 amino acids, essential in the binding of CCR2 with C-C Motif Chemokine Ligand 2 (CCL2), the endogenous substrate, mitigates its activity in TBI. We used a pharmacophore model to screen for suitable ligands that may bind to CCR2, which returned 871 ligands. Docking and molecular dynamics results uncovered that two ligands (A102) and (A435) contained several of those important residues and showed a stability and compactness when in complex with CCR2, with these results confirmed by MMGBSA calculations with A102 recording a better interaction compared to A435. Finally, a PPI network was built to explore downstream signaling being regulated by both ligands in TBI, showing amyloid precursor protein (APP) as a key target and neuroactive-ligand receptor interaction (1.80E-27) the top functional annotated category. In conclusion, for the first time we report novel ligands A102 and A435 targeting CCR2 as a potential new pharmacological approach to target inflammation post-TBI.
Collapse
Affiliation(s)
- Kilian R Sachdev
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Kevin J Lynch
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| |
Collapse
|
8
|
McGovern AJ, González J, Ramírez D, Barreto GE. Identification of HMGCR, PPGARG and prohibitin as potential druggable targets of dihydrotestosterone for treatment against traumatic brain injury using system pharmacology. Int Immunopharmacol 2022; 108:108721. [PMID: 35344815 DOI: 10.1016/j.intimp.2022.108721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Traumatic Brain Injury (TBI) has long-term devastating effects for which there is no accurate and effective treatment for inflammation and chronic oxidative stress. As a disease that affects multiple signalling pathways, the search for a drug with a broader spectrum of pharmacological action is of clinical interest. The fact that endocrine disruption (e.g hypogonadism) has been observed in TBI patients suggests that endogenous therapy with testosterone, or its more androgenic derivative, dihydrotestosterone (DHT), may attenuate, at least in part, the TBI-induced inflammation, but the underlying molecular mechanisms by which this occurs are still not completely clear. AIMS AND METHODS In this study, the main aim was to investigate proteins that may be related to the pathophysiological mechanism of TBI and also be pharmacological targets of DHT in order to explore a possible therapy with this androgen using network pharmacology. RESULTS AND CONCLUSIONS We identified 2.700 proteins related to TBI and 1.567 that are potentially molecular targets of DHT. Functional enrichment analysis showed that steroid (p-value: 2.1-22), lipid metabolism (p-value: 2.8-21) and apoptotic processes (p-value: 5.2-21) are mainly altered in TBI. Furthermore, being mitochondrion an organelle involved on these molecular processes we next identified that out of 32 mitochondrial-related proteins 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), peroxisome proliferator activated receptor gamma (PPGARG) and prohibitin are those found highly regulated in the network and potential targets of DHT in TBI. In conclusion, the identification of these cellular nodes may prove to be essential as targets of DHT for therapy against post-TBI inflammation.
Collapse
Affiliation(s)
- Andrew J McGovern
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| |
Collapse
|
9
|
Ge H, Zhang B, Li T, Yang Q, Tang Y, Liu J, Zhang T. In vivo and in silico studies on the mechanisms of egg white peptides in relieving acute colitis symptoms. Food Funct 2021; 12:12774-12787. [PMID: 34851341 DOI: 10.1039/d1fo03095g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Active peptides, as an alternative nutrition supplement, have been confirmed to have beneficial efficacy against acute colitis. Herein, egg white peptides (EWPs) were used as a nutritional supplement to relieve dextran sulfate sodium-induced acute colitis symptoms. The potential multi-component synergetic pharmacological intervention mechanism of EWPs was investigated on the basis of in silico pharmacology, bioinformatics analysis, and molecular docking. In vitro experiments demonstrated that the migration rate of HSF cells was enhanced 5.30-fold upon treatment with EWPs relative to the control group. After administration with EWPs, colitis symptoms were alleviated in a dose-dependent manner and the serum amino acid content was significantly enhanced, especially for Ala, Leu, Ser, Thr, and Met. Four peptides identified from EWPs showed a total of 52 acute colitis-related potential targets (Fit score >3.8) with network pharmacology analysis, and the targets participated in 31 signaling pathways (p < 0.001). Among these pathways, PI3K-Akt, VEGF, Ras, TNF, and MAPK signaling pathways may exert essential anti-inflammatory effects and accelerate repairing intestinal mucosa. Molecular docking showed that the majority binding energy of peptides-targets was between -10.35 kcal mol-1 and -18.72 kcal mol-1, and peptides mainly interacted with the core targets (Btk, Gstm1, and Rac1) by hydrogen-bonding interactions. The current study confirmed that EWPs as supplementary nutrition can alleviate acute colitis.
Collapse
Affiliation(s)
- Huifang Ge
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Biying Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Ting Li
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Qi Yang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Yuanhu Tang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Jingbo Liu
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| | - Ting Zhang
- Jilin Provincial Key Laboratory of Nutrition and Functional Food and College of Food Science and Engineering, Jilin University, Changchun, 130062, People's Republic of China.
| |
Collapse
|
10
|
Wu YH, Rosset S, Lee TR, Dragunow M, Park T, Shim V. In Vitro Models of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2021; 38:2336-2372. [PMID: 33563092 DOI: 10.1089/neu.2020.7402] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health challenge that is also the third leading cause of death worldwide. It is also the leading cause of long-term disability in children and young adults worldwide. Despite a large body of research using predominantly in vivo and in vitro rodent models of brain injury, there is no medication that can reduce brain damage or promote brain repair mainly due to our lack of understanding in the mechanisms and pathophysiology of the TBI. The aim of this review is to examine in vitro TBI studies conducted from 2008-2018 to better understand the TBI in vitro model available in the literature. Specifically, our focus was to perform a detailed analysis of the in vitro experimental protocols used and their subsequent biological findings. Our review showed that the uniaxial stretch is the most frequently used way of load application, accounting for more than two-thirds of the studies reviewed. The rate and magnitude of the loading were varied significantly from study to study but can generally be categorized into mild, moderate, and severe injuries. The in vitro studies reviewed here examined key processes in TBI pathophysiology such as membrane disruptions leading to ionic dysregulation, inflammation, and the subsequent damages to the microtubules and axons, as well as cell death. Overall, the studies examined in this review contributed to the betterment of our understanding of TBI as a disease process. Yet, our review also revealed the areas where more work needs to be done such as: 1) diversification of load application methods that will include complex loading that mimics in vivo head impacts; 2) more widespread use of human brain cells, especially patient-matched human cells in the experimental set-up; and 3) need for building a more high-throughput system to be able to discover effective therapeutic targets for TBI.
Collapse
Affiliation(s)
- Yi-Han Wu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Samuel Rosset
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Tae-Rin Lee
- Advanced Institute of Convergence Technology, Seoul National University, Seoul, Korea
| | - Mike Dragunow
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Thomas Park
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Vickie Shim
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Extracellular vesicles isolated from mesenchymal stromal cells primed with neurotrophic factors and signaling modifiers as potential therapeutics for neurodegenerative diseases. Curr Res Transl Med 2021; 69:103286. [DOI: 10.1016/j.retram.2021.103286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 12/17/2022]
|
12
|
Chubarev VN, Beeraka NM, Sinelnikov MY, Bulygin KV, Nikolenko VN, Mihaylenko E, Tarasov VV, Mikhaleva LM, Poltronieri P, Viswanadha VP, Somasundaram SG, Kirkland CE, Chen K, Liu J, Fan R, Kamal MA, Mironov AA, Madhunapantula SV, Pretorius E, Dindyaev SV, Muresanu C, Sukocheva OA. Health Science Community Will Miss This Bright and Uniting Star: In Memory of Professor Gjumrakch Aliev, M.D, Ph.D. Cancers (Basel) 2021; 13:1965. [PMID: 33921833 PMCID: PMC8072812 DOI: 10.3390/cancers13081965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/05/2023] Open
Abstract
It is with deep sadness that we offer our memorial on the unexpected demise of our dear colleague, Professor Gjumrakch Aliev [...].
Collapse
Affiliation(s)
- Vladimir N. Chubarev
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Narasimha M. Beeraka
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Bannimantapa, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka 570 015, India;
| | - Mikhail Y. Sinelnikov
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Kirill V. Bulygin
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 117192 Moscow, Russia
| | - Vladimir N. Nikolenko
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 117192 Moscow, Russia
| | - Elizaveta Mihaylenko
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Vadim V. Tarasov
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | | | - Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, via Monteroni km 7, 73100 Lecce, Italy;
| | | | - Siva G. Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Cecil E. Kirkland
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Kuo Chen
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Mohammad Amjad Kamal
- West China School of Nursing/Institutes for Systems Genetics, The Frontier Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China;
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Alexander A. Mironov
- Laboratory of Electron Microscopy, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy;
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Bannimantapa, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka 570 015, India;
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa;
| | - Sergey V. Dindyaev
- Department of Histology, Embryology & Cytology, Pediatric Faculty, Federal State Budgetary Educational Institution of Higher Education “Ivanovo State Medical Academy” of the Ministry of Healthcare of the Russian Federation (FSBEI HE IvSMA MOH Russia), 8 Sheremetyevsky Ave., 153012 Ivanovo, Russia;
| | - Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478 Cluj-Napoca, Romania;
| | - Olga A. Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University of South Australia, Adelaide 5001, Australia
| |
Collapse
|
13
|
Zhang GL, Qian C, Zhang SZ, Tuo YH, Zeng BY, Ji YX, Wang YZ. Effect of conditioned medium from neural stem cells on glioma progression and its protein expression profile analysis. World J Stem Cells 2020; 12:1396-1409. [PMID: 33312406 PMCID: PMC7705462 DOI: 10.4252/wjsc.v12.i11.1396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/04/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Emerging evidence suggests that the spread of glioma to the subventricular zone (SVZ) is closely related to glioma recurrence and patient survival. Neural stem cells (NSCs) are the main cell type in the SVZ region and exhibit tumor-homing ability.
AIM To evaluate the effects of conditioned medium (CM) derived from SVZ NSCs on the cancer-related behaviors of glioma cells.
METHODS The characteristics of SVZ hNSCs were identified by immunofluorescence. The normoxic-hNSC-CM and hypoxic-hNSC-CM (3% O2, oxygen-glucose deprived [OGD] culturing) were collected from 80%-90% confluent SVZ NSCs in sterile conditions. The CCK8 and Transwell assays were used to compare and evaluate the effects of normoxic-CM and hypoxic-CM on glioma proliferation and invasion. Then proteins secreted from SVZ NSCs into the CM were investigated by mass spectrometry, and the potential effects of candidate protein NCAN in the regulation of glioma progression were examined by CCK8 and Transwell assays.
RESULTS The CM from SVZ NSCs significantly increased the proliferation and invasion of glioma cells, particularly the CM from OGD NSCs induced under hypoxic conditions. Furthermore, the secreted protein neurocan (NCAN) in CM from OGD NSCs was identified by proteomic analysis. NCAN was expressed in glioma cells and played regulatory roles in mediating the progression of glioma cells mainly via the Rho/Rho-associated protein kinase pathway.
CONCLUSION Our study identified a potential interactive mechanism between SVZ NSCs and glioma cells, in which SVZ NSCs promote glioma progression via the secreted protein NCAN. These findings suggested that exploring the CM derived from cells could be a novel strategy for optimizing treatments and that NCAN derived from SVZ NSCs may be a potential new target in glioma progression.
Collapse
Affiliation(s)
- Gui-Long Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province China
| | - Cheng Qian
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Shi-Zhen Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Yong-Hua Tuo
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Bai-Yun Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Yun-Xiang Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| | - Ye-Zhong Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, Guangdong Province, China
| |
Collapse
|
14
|
Omelchenko A, Singh NK, Firestein BL. Current advances in in vitro models of central nervous system trauma. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 14:34-41. [PMID: 32671312 PMCID: PMC7363028 DOI: 10.1016/j.cobme.2020.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CNS trauma is a prominent cause of mortality and morbidity, and although much effort has focused on developing treatments for CNS trauma-related pathologies, little progress has been made. Pre-clinical models of TBI and SCI suffer from significant drawbacks, which result in substantial failures during clinical translation of promising pre-clinical therapies. Here, we review recent advances made in the development of in vitro models of CNS trauma, the promises and drawbacks of current in vitro CNS injury models, and the attributes necessary for future models to accurately mimic the trauma microenvironment and facilitate CNS trauma drug discovery. The goal is to provide insight for the development of future CNS injury models and to aid researchers in selecting effective models for pre-clinical research of trauma therapeutics.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience; Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
| | - Nisha K. Singh
- Department of Cell Biology and Neuroscience; Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
- Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience; Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
| |
Collapse
|
15
|
Nebie O, Barro L, Wu YW, Knutson F, Buée L, Devos D, Peng CW, Blum D, Burnouf T. Heat-treated human platelet pellet lysate modulates microglia activation, favors wound healing and promotes neuronal differentiation in vitro. Platelets 2020; 32:226-237. [PMID: 32106742 DOI: 10.1080/09537104.2020.1732324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The neurorestorative efficacy of human platelet lysates in neurodegenerative disorders is still under investigation. Platelets prepared from standard and pathogen reduced platelet concentrates were pelletized, washed, concentrated, and subjected to freeze-thawing. The lysate was heated to 56°C for 30 min and characterized. Toxicity was evaluated using SH-SY5Y neuroblastoma, BV-2 microglial, and EA-hy926 endothelial cells. Inflammatory activity was tested by examining tumor necrosis factor (TNF) and cyclooxygenase (COX)-2 expressions by BV-2 microglia with or without stimulation by lipopolysaccharides (LPS). The capacity to stimulate wound healing was evaluated by a scratch assay, and the capacity to differentiate SH-SY5Y into neurons was also examined. Platelet lysates contained a range of neurotrophins. They were not toxic to SH-SY5Y, EA-hy926, or BV-2 cells, did not induce the expression of TNF or COX-2 inflammatory markers by BV-2 microglia, and decreased inflammation after LPS stimulation. They stimulated the wound closure in the scratch assay and induced SH-SY5Y differentiation as revealed by the increased length of neurites as well as β3-tubulin and neurofilament staining. These data confirm the therapeutic potential of platelet lysates in the treatment of disorders of the central nervous system and support further evaluation as novel neurorestorative biotherapy in preclinical models.
Collapse
Affiliation(s)
- Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Folke Knutson
- Clinical Immunology and Transfusion Medicine IGP, Uppsala University, Uppsala, Sweden
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, U1172, Lille Neuroscience & Cognition, France
| | - David Devos
- Univ. Lille, Inserm, CHU-Lille, U1172, Lille Neuroscience & Cognition, France
| | - Chih-Wei Peng
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - David Blum
- Univ. Lille, Inserm, CHU-Lille, U1172, Lille Neuroscience & Cognition, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,PhD Program in Mind, Brain & Consciousness, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
16
|
Gorabi AM, Kiaie N, Barreto GE, Read MI, Tafti HA, Sahebkar A. The Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes in Treatment of Neurodegenerative Diseases. Mol Neurobiol 2019; 56:8157-8167. [PMID: 31197655 DOI: 10.1007/s12035-019-01663-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/27/2019] [Indexed: 12/13/2022]
Abstract
Neurologic complications are commonly regarded as irreversible impairments that stem from limited potential of regeneration of the central nervous system (CNS). On the other side, the regenerative potential of stem cells has been evaluated in basic research, as well as in preclinical studies. Mesenchymal stem cells (MSCs) have been regarded as candidate cell sources for therapeutic purposes of various neurological disorders, because of their self-renewal ability, plasticity in differentiation, neurotrophic characteristics, and immunomodulatory properties. Exosomes are extracellular vesicles which can deliver biological information over long distances and thereby influencing normal and abnormal processes in cells and tissues. The therapeutic capacity of exosomes relies on the type of cell, as well as on the physiological condition of a given cell. Therefore, based on tissue type and physiological condition of CNS, exosomes may function as contributors or suppressors of pathological conditions in this tissue. When it comes to the therapeutic viewpoint, the most promising cellular source of exosomes is considered to be MSCs. The aim of this review article is to discuss the current knowledge around the potential of stem cells and MSC-derived exosomes in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Morgayn I Read
- Department of Pharmacology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Tehran, Iran.
- School of Medicine, Mashhad University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
The glucagon-like peptide-1 receptor agonist reduces inflammation and blood-brain barrier breakdown in an astrocyte-dependent manner in experimental stroke. J Neuroinflammation 2019; 16:242. [PMID: 31779652 PMCID: PMC6883580 DOI: 10.1186/s12974-019-1638-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 11/11/2019] [Indexed: 11/30/2022] Open
Abstract
Background Preserving the integrity of the blood-brain barrier (BBB) is beneficial to avoid further brain damage after acute ischemic stroke (AIS). Astrocytes, an important component of the BBB, promote BBB breakdown in subjects with AIS by secreting inflammatory factors. The glucagon-like peptide-1 receptor (GLP-1R) agonist exendin-4 (Ex-4) protects the BBB and reduces brain inflammation from cerebral ischemia, and GLP-1R is expressed on astrocytes. However, the effect of Ex-4 on astrocytes in subjects with AIS remains unclear. Methods In the present study, we investigated the effect of Ex-4 on astrocytes cultured under oxygen-glucose deprivation (OGD) plus reoxygenation conditions and determined whether the effect influences bEnd.3 cells. We used various methods, including permeability assays, western blotting, immunofluorescence staining, and gelatin zymography, in vitro and in vivo. Results Ex-4 reduced OGD-induced astrocyte-derived vascular endothelial growth factor (VEGF-A), matrix metalloproteinase-9 (MMP-9), chemokine monocyte chemoattractant protein-1 (MCP-1), and chemokine C-X-C motif ligand 1 (CXCL-1). The reduction in astrocyte-derived VEGF-A and MMP-9 was related to the increased expression of tight junction proteins (TJPs) in bEnd.3 cells. Ex-4 improved neurologic deficit scores, reduced the infarct area, and ameliorated BBB breakdown as well as decreased astrocyte-derived VEGF-A, MMP-9, CXCL-1, and MCP-1 levels in ischemic brain tissues from rats subjected to middle cerebral artery occlusion. Ex-4 reduced the activation of the JAK2/STAT3 signaling pathway in astrocytes following OGD. Conclusion Based on these findings, ischemia-induced inflammation and BBB breakdown can be improved by Ex-4 through an astrocyte-dependent manner.
Collapse
|
18
|
Baez-Jurado E, Hidalgo-Lanussa O, Barrera-Bailón B, Sahebkar A, Ashraf GM, Echeverria V, Barreto GE. Secretome of Mesenchymal Stem Cells and Its Potential Protective Effects on Brain Pathologies. Mol Neurobiol 2019; 56:6902-6927. [PMID: 30941733 DOI: 10.1007/s12035-019-1570-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
Previous studies have indicated that mesenchymal stem cells (MSCs) have a fundamental role in the repair and regeneration of damaged tissues. There is strong evidence showing that much of the beneficial effects of these cells are due to the secretion of bioactive molecules-besides microRNAs, hormones, and neurotrophins-with anti-inflammatory, immunoregulatory, angiogenic, and trophic effects. These factors have been reported by many studies to possess protective effects on the nervous tissue. Although the beneficial effects of the secretory factors of MSCs have been suggested for various neurological diseases, their actions on astrocytic cells are not well understood. Hence, it is important to recognize the specific effects of MSCs derived from adipose tissue, in addition to the differences presented by the secretome, depending on the source and methods of analysis. In this paper, the different sources of MSCs and their main characteristics are described, as well as the most significant advances in regeneration and protection provided by the secretome of MSCs. Also, we discuss the possible neuroprotective mechanisms of action of the MSC-derived biomolecules, with special emphasis on the effect of MSCs derived from adipose tissue and their impact on glial cells and brain pathologies.
Collapse
Affiliation(s)
- Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Biviana Barrera-Bailón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastian, Lientur 1457, 4080871, Concepción, Chile.,Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
| |
Collapse
|
19
|
Zhang Y, Yu M, Zhao X, Dai M, Chen C, Tian W. Optimizing adipose tissue extract isolation with stirred suspension culture. Connect Tissue Res 2019; 60:178-188. [PMID: 29852798 DOI: 10.1080/03008207.2018.1483357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Adherent culture which is used to collect adipose tissue extract (ATE) previously brings the problem of inhomogeneity and non-repeatability. Here we aim to extract ATE with stirred suspension culture to speed up the extraction process, stabilize the yield, and improve consistent potency metrics of ATE. MATERIALS AND METHODS ATE was collected with adherent culture (ATE-A) and stirred suspension culture (ATE-S) separately. Protein yield and composition were detected by SDS-PAGE, while cytokines in ATE were determined with ELISA. The adipogenic and angiogenic potential of ATE were compared by western blot and qPCR. In addition, haematoxylin and eosin staining and lactate dehydrogenase (LDH) activity assays were used to analyze the cell viability of adipose tissue cultured with different methods. RESULTS The yield of ATE-S was consistent while ATE-A varied notably. Characterization of the protein composition and exosome-like vesicles (ELVs) indicated no significant difference between ATE-S and ATE-A. The concentrations of cytokines (VEGF, bFGF, and IL-6) showed no significant difference, while IGF in ATE-S was higher than that in ATE-A. ATE-S showed upregulated adipogenic and angiogenic potential compared to ATE-A. Morever, stirred suspension culture decreased the LDH activity of ATE while increased the number of viable adipocytes and reduced adipose tissue necrosis. CONCLUSION Compared with adherent culture, stirred suspension culture is a reliable, time- and labor-saving method to collect ATE, which might be used to improve the downstream applications of ATE.
Collapse
Affiliation(s)
- Yan Zhang
- a State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology , Sichuan University , Chengdu , China
| | - Mei Yu
- a State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology , Sichuan University , Chengdu , China
| | - Xueyong Zhao
- a State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology , Sichuan University , Chengdu , China
| | - Minjia Dai
- a State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology , Sichuan University , Chengdu , China
| | - Chang Chen
- a State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology , Sichuan University , Chengdu , China
| | - Weidong Tian
- a State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology , Sichuan University , Chengdu , China
| |
Collapse
|
20
|
Mitochondrial Neuroglobin Is Necessary for Protection Induced by Conditioned Medium from Human Adipose-Derived Mesenchymal Stem Cells in Astrocytic Cells Subjected to Scratch and Metabolic Injury. Mol Neurobiol 2018; 56:5167-5187. [PMID: 30536184 DOI: 10.1007/s12035-018-1442-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Astrocytes are specialized cells capable of regulating inflammatory responses in neurodegenerative diseases or traumatic brain injury. In addition to playing an important role in neuroinflammation, these cells regulate essential functions for the preservation of brain tissue. Therefore, the search for therapeutic alternatives to preserve these cells and maintain their functions contributes in some way to counteract the progress of the injury and maintain neuronal survival in various brain pathologies. Among these strategies, the conditioned medium from human adipose-derived mesenchymal stem cells (CM-hMSCA) has been reported with a potential beneficial effect against several neuropathologies. In this study, we evaluated the potential effect of CM-hMSCA in a model of human astrocytes (T98G cells) subjected to scratch injury. Our findings demonstrated that CM-hMSCA regulates the cytokines IL-2, IL-6, IL-8, IL-10, GM-CSF, and TNF-α, downregulates calcium at the cytoplasmic level, and regulates mitochondrial dynamics and the respiratory chain. These actions are accompanied by modulation of the expression of different proteins involved in signaling pathways such as AKT/pAKT and ERK1/2/pERK, and may mediate the localization of neuroglobin (Ngb) at the cellular level. We also confirmed that Ngb mediated the protective effects of CM-hMSCA through regulation of proteins involved in survival pathways and oxidative stress. In conclusion, regulation of brain inflammation combined with the recovery of fundamental cellular aspects in the face of injury makes CM-hMSCA a promising candidate for the protection of astrocytes in brain pathologies.
Collapse
|
21
|
Role of GTPases in the Regulation of Mitochondrial Dynamics in Alzheimer's Disease and CNS-Related Disorders. Mol Neurobiol 2018; 56:4530-4538. [PMID: 30338485 DOI: 10.1007/s12035-018-1397-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/14/2018] [Indexed: 12/22/2022]
Abstract
Data obtained from several studies have shown that mitochondria are involved and play a central role in the progression of several distinct pathological conditions. Morphological alterations and disruptions on the functionality of mitochondria may be related to metabolic and energy deficiency in neurons in a neurodegenerative disorder. Several recent studies demonstrate the linkage between neurodegeneration and mitochondrial dynamics in the spectrum of a promising era called precision mitochondrial medicine. In this review paper, an analysis of the correlation between mitochondria, Alzheimer's disease, and other central nervous system (CNS)-related disorders like the Parkinson's disease and the autism spectrum disorder is under discussion. The role of GTPases like the mfn1, mfn2, opa1, and dlp1 in mitochondrial fission and fusion is also under investigation, influencing mitochondrial population and leading to oxidative stress and neuronal damage.
Collapse
|
22
|
Rahman S, Archana A, Jan AT, Minakshi R. Dissecting Endoplasmic Reticulum Unfolded Protein Response (UPR ER) in Managing Clandestine Modus Operandi of Alzheimer's Disease. Front Aging Neurosci 2018; 10:30. [PMID: 29467648 PMCID: PMC5808164 DOI: 10.3389/fnagi.2018.00030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/24/2018] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, is most common cause of dementia witnessed among aged people. The pathophysiology of AD develops as a consequence of neurofibrillary tangle formation which consists of hyperphosphorylated microtubule associated tau protein and senile plaques of amyloid-β (Aβ) peptide in specific brain regions that result in synaptic loss and neuronal death. The feeble buffering capacity of endoplasmic reticulum (ER) proteostasis in AD is evident through alteration in unfolded protein response (UPR), where UPR markers express invariably in AD patient's brain samples. Aging weakens UPRER causing neuropathology and memory loss in AD. This review highlights molecular signatures of UPRER and its key molecular alliance that are affected in aging leading to the development of intriguing neuropathologies in AD. We present a summary of recent studies reporting usage of small molecules as inhibitors or activators of UPRER sensors/effectors in AD that showcase avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Safikur Rahman
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Ayyagari Archana
- Department of Microbiology, Swami Shraddhanand College, University of Delhi, New Delhi, India
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Rinki Minakshi
- Institute of Home Economics, University of Delhi, New Delhi, India
| |
Collapse
|
23
|
Kosenko EA, Tikhonova LA, Montoliu C, Barreto GE, Aliev G, Kaminsky YG. Metabolic Abnormalities of Erythrocytes as a Risk Factor for Alzheimer's Disease. Front Neurosci 2018; 11:728. [PMID: 29354027 PMCID: PMC5760569 DOI: 10.3389/fnins.2017.00728] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/13/2017] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's disease (AD) is a slowly progressive, neurodegenerative disorder of uncertain etiology. According to the amyloid cascade hypothesis, accumulation of non-soluble amyloid β peptides (Aβ) in the Central Nervous System (CNS) is the primary cause initiating a pathogenic cascade leading to the complex multilayered pathology and clinical manifestation of the disease. It is, therefore, not surprising that the search for mechanisms underlying cognitive changes observed in AD has focused exclusively on the brain and Aβ-inducing synaptic and dendritic loss, oxidative stress, and neuronal death. However, since Aβ depositions were found in normal non-demented elderly people and in many other pathological conditions, the amyloid cascade hypothesis was modified to claim that intraneuronal accumulation of soluble Aβ oligomers, rather than monomer or insoluble amyloid fibrils, is the first step of a fatal cascade in AD. Since a characteristic reduction of cerebral perfusion and energy metabolism occurs in patients with AD it is suggested that capillary distortions commonly found in AD brain elicit hemodynamic changes that alter the delivery and transport of essential nutrients, particularly glucose and oxygen to neuronal and glial cells. Another important factor in tissue oxygenation is the ability of erythrocytes (red blood cells, RBC) to transport and deliver oxygen to tissues, which are first of all dependent on the RBC antioxidant and energy metabolism, which finally regulates the oxygen affinity of hemoglobin. In the present review, we consider the possibility that metabolic and antioxidant defense alterations in the circulating erythrocyte population can influence oxygen delivery to the brain, and that these changes might be a primary mechanism triggering the glucose metabolism disturbance resulting in neurobiological changes observed in the AD brain, possibly related to impaired cognitive function. We also discuss the possibility of using erythrocyte biochemical aberrations as potential tools that will help identify a risk factor for AD.
Collapse
Affiliation(s)
- Elena A Kosenko
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Lyudmila A Tikhonova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Carmina Montoliu
- Fundación Investigación Hospital Clínico, INCLIVA Instituto Investigación Sanitaria, Valencia, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Gjumrakch Aliev
- GALLY International Biomedical Research Institute Inc., San Antonio, TX, United States
| | - Yury G Kaminsky
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|