1
|
Basheer HA, Salman NM, Abdullah RM, Elsalem L, Afarinkia K. Metformin and glioma: Targeting metabolic dysregulation for enhanced therapeutic outcomes. Transl Oncol 2025; 53:102323. [PMID: 39970627 DOI: 10.1016/j.tranon.2025.102323] [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: 08/22/2024] [Revised: 12/09/2024] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
Glioma, a highly aggressive form of brain cancer, continues to pose significant therapeutic challenges in the field of medicine. Its invasive nature and resistance to traditional treatments make it particularly difficult to combat. This review examines the potential of metformin, a commonly prescribed antidiabetic medication, as a promising new treatment option for glioma. The potential of metformin to target crucial metabolic pathways in cancer cells presents an encouraging approach to improve therapeutic outcomes. The review explores the complexities of metabolic reprogramming in glioma and metformin's role in inhibiting these metabolic pathways. Preclinical studies demonstrate metformin's efficacy in reducing tumor growth and enhancing the sensitivity of glioma cells to chemotherapy and radiotherapy. Furthermore, clinical studies highlight metformin's potential in improving progression-free survival and overall survival rates in glioma patients. The review also addresses the synergistic effects of combining metformin with other therapeutic agents, such as temozolomide and radiotherapy, to overcome drug resistance and improve treatment efficacy. Despite the promising findings, the review acknowledges the need for further clinical trials to establish optimal dosing regimens, understand the molecular mechanisms underlying metformin's antitumor effects, and identify patient populations that would benefit the most from metformin-based therapies. Additionally, the potential side effects and the long-term impact of metformin on Glioma patients require careful evaluation. In conclusion, this review underscores the potential of metformin as a repurposed drug in glioma treatment, emphasizing its multifaceted role in targeting metabolic dysregulation. Metformin holds promise as part of a combination therapy approach to improve the therapeutic landscape of glioma and offers hope for better patient outcomes.
Collapse
Affiliation(s)
- Haneen A Basheer
- Department of Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan.
| | - Nadeem M Salman
- Department of Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan
| | - Rami M Abdullah
- Department of Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan
| | - Lina Elsalem
- Jordan University of Science and Technology, Faculty of Medicine, Department of Pharmacology, Irbid, Jordan
| | - Kamyar Afarinkia
- School of Medicine and Biosciences, University of West London, London W5 5RF, UK
| |
Collapse
|
2
|
Rawani NS, Chan AW, Todd KG, Baker GB, Dursun SM. The Role of Neuroglia in the Development and Progression of Schizophrenia. Biomolecules 2024; 15:10. [PMID: 39858403 PMCID: PMC11761573 DOI: 10.3390/biom15010010] [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: 09/25/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Schizophrenia is a complex heterogenous disorder thought to be caused by interactions between genetic and environmental factors. The theories developed to explain the etiology of schizophrenia have focused largely on the dysfunction of neurotransmitters such as dopamine, serotonin and glutamate with their receptors, although research in the past several decades has indicated strongly that other factors are also involved and that the role of neuroglial cells in psychotic disorders including schizophrenia should be given more attention. Although glia were originally thought to be present in the brain only to support neurons in a physical, metabolic and nutritional capacity, it has become apparent that these cells have a variety of important physiological roles and that abnormalities in their function may make significant contributions to the symptoms of schizophrenia. In the present paper, we review the interactions of brain microglia, astrocytes and oligodendroglia with aspects such as transmitter dysregulation, neuro-inflammation, oxidative stress, synaptic function, the gut microbiome, myelination and the blood-brain barrier that appear to affect the cause, development and treatment of schizophrenia. We also review crosstalk between microglia, astrocytes and oligodendrocytes and the effects of antipsychotics on neuroglia. Problems associated with studies on specific biomarkers for glia in schizophrenia are discussed.
Collapse
Affiliation(s)
| | | | | | - Glen B. Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2G3, Canada; (N.S.R.); (A.W.C.); (K.G.T.); (S.M.D.)
| | | |
Collapse
|
3
|
Vazquez C, Negatu SG, Bannerman CD, Sriram S, Ming GL, Jurado KA. Antiviral immunity within neural stem cells distinguishes Enterovirus-D68 strain differences in forebrain organoids. J Neuroinflammation 2024; 21:288. [PMID: 39501367 PMCID: PMC11539839 DOI: 10.1186/s12974-024-03275-5] [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: 07/23/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
Neural stem cells have intact innate immune responses that protect them from virus infection and cell death. Yet, viruses can antagonize such responses to establish neuropathogenesis. Using a forebrain organoid model system at two developmental time points, we identified that neural stem cells, in particular radial glia, are basally primed to respond to virus infection by upregulating several antiviral interferon-stimulated genes. Infection of these organoids with a neuropathogenic Enterovirus-D68 strain, demonstrated the ability of this virus to impede immune activation by blocking interferon responses. Together, our data highlight immune gene signatures present in different types of neural stem cells and differential viral capacity to block neural-specific immune induction.
Collapse
Affiliation(s)
- Christine Vazquez
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Seble G Negatu
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Carl D Bannerman
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sowmya Sriram
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kellie A Jurado
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Noor L, Upadhyay A, Joshi V. Role of T Lymphocytes in Glioma Immune Microenvironment: Two Sides of a Coin. BIOLOGY 2024; 13:846. [PMID: 39452154 PMCID: PMC11505600 DOI: 10.3390/biology13100846] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Glioma is known for its immunosuppressive microenvironment, which makes it challenging to target through immunotherapies. Immune cells like macrophages, microglia, myeloid-derived suppressor cells, and T lymphocytes are known to infiltrate the glioma tumor microenvironment and regulate immune response distinctively. Among the variety of immune cells, T lymphocytes have highly complex and multifaceted roles in the glioma immune landscape. T lymphocytes, which include CD4+ helper and CD8+ cytotoxic T cells, are known for their pivotal roles in anti-tumor responses. However, these cells may behave differently in the highly dynamic glioma microenvironment, for example, via an immune invasion mechanism enforced by tumor cells. Therefore, T lymphocytes play dual roles in glioma immunity, firstly by their anti-tumor responses, and secondly by exploiting gliomas to promote immune invasion. As an immunosuppression strategy, glioma induces T-cell exhaustion and suppression of effector T cells by regulatory T cells (Tregs) or by altering their signaling pathways. Further, the expression of immune checkpoint inhibitors on the glioma cell surface leads to T cell anergy and dysfunction. Overall, this dynamic interplay between T lymphocytes and glioma is crucial for designing more effective immunotherapies. The current review provides detailed knowledge on the roles of T lymphocytes in the glioma immune microenvironment and helps to explore novel therapeutic approaches to reinvigorate T lymphocytes.
Collapse
Affiliation(s)
- Laiba Noor
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Vibhuti Joshi
- Department of Biotechnology, Bennett University, Greater Noida 201310, Uttar Pradesh, India
| |
Collapse
|
5
|
Nussinov R, Yavuz BR, Jang H. Single cell spatial biology over developmental time can decipher pediatric brain pathologies. Neurobiol Dis 2024; 199:106597. [PMID: 38992777 DOI: 10.1016/j.nbd.2024.106597] [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: 03/27/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric low grade brain tumors and neurodevelopmental disorders share proteins, signaling pathways, and networks. They also share germline mutations and an impaired prenatal differentiation origin. They may differ in the timing of the events and proliferation. We suggest that their pivotal distinct, albeit partially overlapping, outcomes relate to the cell states, which depend on their spatial location, and timing of gene expression during brain development. These attributes are crucial as the brain develops sequentially, and single-cell spatial organization influences cell state, thus function. Our underlying premise is that the root cause in neurodevelopmental disorders and pediatric tumors is impaired prenatal differentiation. Data related to pediatric brain tumors, neurodevelopmental disorders, brain cell (sub)types, locations, and timing of expression in the developing brain are scant. However, emerging single cell technologies, including transcriptomic, spatial biology, spatial high-resolution imaging performed over the brain developmental time, could be transformational in deciphering brain pathologies thereby pharmacology.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
6
|
Tamo AK, Djouonkep LDW, Selabi NBS. 3D Printing of Polysaccharide-Based Hydrogel Scaffolds for Tissue Engineering Applications: A Review. Int J Biol Macromol 2024; 270:132123. [PMID: 38761909 DOI: 10.1016/j.ijbiomac.2024.132123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
In tissue engineering, 3D printing represents a versatile technology employing inks to construct three-dimensional living structures, mimicking natural biological systems. This technology efficiently translates digital blueprints into highly reproducible 3D objects. Recent advances have expanded 3D printing applications, allowing for the fabrication of diverse anatomical components, including engineered functional tissues and organs. The development of printable inks, which incorporate macromolecules, enzymes, cells, and growth factors, is advancing with the aim of restoring damaged tissues and organs. Polysaccharides, recognized for their intrinsic resemblance to components of the extracellular matrix have garnered significant attention in the field of tissue engineering. This review explores diverse 3D printing techniques, outlining distinctive features that should characterize scaffolds used as ideal matrices in tissue engineering. A detailed investigation into the properties and roles of polysaccharides in tissue engineering is highlighted. The review also culminates in a profound exploration of 3D polysaccharide-based hydrogel applications, focusing on recent breakthroughs in regenerating different tissues such as skin, bone, cartilage, heart, nerve, vasculature, and skeletal muscle. It further addresses challenges and prospective directions in 3D printing hydrogels based on polysaccharides, paving the way for innovative research to fabricate functional tissues, enhancing patient care, and improving quality of life.
Collapse
Affiliation(s)
- Arnaud Kamdem Tamo
- Institute of Microsystems Engineering IMTEK, University of Freiburg, 79110 Freiburg, Germany; Freiburg Center for Interactive Materials and Bioinspired Technologies FIT, University of Freiburg, 79110 Freiburg, Germany; Freiburg Materials Research Center FMF, University of Freiburg, 79104 Freiburg, Germany; Ingénierie des Matériaux Polymères (IMP), Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, CNRS, UMR 5223, 69622 Villeurbanne CEDEX, France.
| | - Lesly Dasilva Wandji Djouonkep
- College of Petroleum Engineering, Yangtze University, Wuhan 430100, China; Key Laboratory of Drilling and Production Engineering for Oil and Gas, Wuhan 430100, China
| | - Naomie Beolle Songwe Selabi
- Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
7
|
Sharma M, Aggarwal N, Mishra J, Panda JJ. Neuroglia targeting nano-therapeutic approaches to rescue aging and neurodegenerating brain. Int J Pharm 2024; 654:123950. [PMID: 38430951 DOI: 10.1016/j.ijpharm.2024.123950] [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: 09/11/2023] [Revised: 02/12/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Despite intense efforts at the bench, the development of successful brain-targeting therapeutics to relieve malicious neural diseases remains primitive. The brain, being a beautifully intricate organ, consists of heterogeneous arrays of neuronal and glial cells. Primarily acting as the support system for neuronal functioning and maturation, glial cells have been observed to be engaged more apparently in the progression and worsening of various neural pathologies. The diseased state is often related to metabolic alterations in glial cells, thereby modulating their physiological homeostasis in conjunction with neuronal dysfunction. A plethora of data indicates the effect of oxidative stress, protein aggregation, and DNA damage in neuroglia impairments. Still, a deeper insight is needed to gain a conflict-free understanding in this arena. As a consequence, glial cells hold the potential to be identified as promising targets for novel therapeutic approaches aimed at brain protection. In this review, we describe the recent strides taken in the direction of understanding the impact of oxidative stress, protein aggregation, and DNA damage on neuroglia impairment and neuroglia-directed nanotherapeutic approaches to mitigate the burden of various neural disorders.
Collapse
Affiliation(s)
- Manju Sharma
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| | - Nidhi Aggarwal
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India
| | - Jibanananda Mishra
- School of Biosciences, RIMT University, Mandi Gobindgarh, Punjab 147301, India.
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Mohali, Punjab 140306, India.
| |
Collapse
|
8
|
Cresto N, Courret M, Génin A, Martin CMP, Bourret J, Sakkaki S, de Bock F, Janvier A, Polizzi A, Payrastre L, Ellero-Simatos S, Audinat E, Perroy J, Marchi N. Continuous low-level dietary exposure to glyphosate elicits dose and sex-dependent synaptic and microglial adaptations in the rodent brain. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123477. [PMID: 38307239 DOI: 10.1016/j.envpol.2024.123477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Prolonged exposure to low levels of dietary contaminants is a context in modern life that could alter organ physiology gradually. Here, we aimed to investigate the impact of continuous exposure to acceptable daily intake (ADI) and non-observable adverse effect level (NOAEL) of glyphosate from gestation to adulthood using C57BL/6J mice and incorporating these levels into their food pellets. From adulthood, we analyzed neurophysiological and neuro-glia cellular adaptations in male and female animals. Using ex-vivo hippocampal slice electrophysiology, we found a reduced efficacy of Schaffer collateral-to-CA1 excitatory synapses in glyphosate-exposed dietary conditions, with ADI and NOAEL dose-dependent effects. Short-term facilitation of excitatory synaptic transmission was specifically increased in NOAEL conditions, with a predominant influence in males, suggesting a reduced probability of neurotransmitter release. Long-term synaptic potentiation (LTP) was decreased in NOAEL-exposed mice. Next, we explore whether these neurophysiological modifications are associated with neuro-glia changes in the somatosensory cortex and hippocampus. High-resolution confocal microscopy analyses unveil a dose-dependent increased density of excitatory Vglut1+ Homer1+ synapses. Microglial Iba1+ cells displayed a shortening of their ramifications, a sign of cellular reactivity that was more pronounced in males at NOAEL levels. The morphology of GFAP+ astrocytes was generally not modified. Finally, we asked whether mouse-specific cross-correlations exist among all data sets generated. This examination included the novel object recognition (NOR) test performed before ex vivo functional and immunohistochemical examinations. We report a negative linear regression between the number of synapses and NOR or LTP maintenance when plotting ADI and NOAEL datasets. These results outline synaptic and microglial cell adaptations resulting from prenatal and continuous dietary low levels of glyphosate, discernible in, but not limited to, adult males exposed to the NOAEL. We discuss the potential significance of these findings to real-world consumer situations and long-term brain resilience.
Collapse
Affiliation(s)
- Noemie Cresto
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Margot Courret
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Athénaïs Génin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Céline Marie Pauline Martin
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Julie Bourret
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Sophie Sakkaki
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Frederic de Bock
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Alicia Janvier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Arnaud Polizzi
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Laurence Payrastre
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, Toulouse, France
| | - Etienne Audinat
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicola Marchi
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
9
|
Izhiman Y, Esfandiari L. Emerging role of extracellular vesicles and exogenous stimuli in molecular mechanisms of peripheral nerve regeneration. Front Cell Neurosci 2024; 18:1368630. [PMID: 38572074 PMCID: PMC10989355 DOI: 10.3389/fncel.2024.1368630] [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: 01/10/2024] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Peripheral nerve injuries lead to significant morbidity and adversely affect quality of life. The peripheral nervous system harbors the unique trait of autonomous regeneration; however, achieving successful regeneration remains uncertain. Research continues to augment and expedite successful peripheral nerve recovery, offering promising strategies for promoting peripheral nerve regeneration (PNR). These include leveraging extracellular vesicle (EV) communication and harnessing cellular activation through electrical and mechanical stimulation. Small extracellular vesicles (sEVs), 30-150 nm in diameter, play a pivotal role in regulating intercellular communication within the regenerative cascade, specifically among nerve cells, Schwann cells, macrophages, and fibroblasts. Furthermore, the utilization of exogenous stimuli, including electrical stimulation (ES), ultrasound stimulation (US), and extracorporeal shock wave therapy (ESWT), offers remarkable advantages in accelerating and augmenting PNR. Moreover, the application of mechanical and electrical stimuli can potentially affect the biogenesis and secretion of sEVs, consequently leading to potential improvements in PNR. In this review article, we comprehensively delve into the intricacies of cell-to-cell communication facilitated by sEVs and the key regulatory signaling pathways governing PNR. Additionally, we investigated the broad-ranging impacts of ES, US, and ESWT on PNR.
Collapse
Affiliation(s)
- Yara Izhiman
- Esfandiari Laboratory, Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Leyla Esfandiari
- Esfandiari Laboratory, Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Electrical and Computer Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
10
|
Raha S, Paidi RK, Dutta D, Pahan K. Cinnamic acid, a natural plant compound, exhibits neuroprotection in a mouse model of Sandhoff disease via PPARα. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:17-32. [PMID: 38532783 PMCID: PMC10961485 DOI: 10.1515/nipt-2023-0027] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Tay-Sachs disease (TSD) and its severe form Sandhoff disease (SD) are autosomal recessive lysosomal storage metabolic disorders, which often result into excessive GM2 ganglioside accumulation predominantly in lysosomes of nerve cells. Although patients with these diseases appear normal at birth, the progressive accumulation of undegraded GM2 gangliosides in neurons leads to early death accompanied by manifestation of motor difficulties and gradual loss of behavioral skills. Unfortunately, there is still no effective treatment available for TSD/SD. The present study highlights the importance of cinnamic acid (CA), a naturally occurring aromatic fatty acid present in a number of plants, in inhibiting the disease process in a transgenic mouse model of SD. Oral administration of CA significantly attenuated glial activation and inflammation and reduced the accumulation of GM2 gangliosides/glycoconjugates in the cerebral cortex of Sandhoff mice. Besides, oral CA also improved behavioral performance and increased the survival of Sandhoff mice. While assessing the mechanism, we found that oral administration of CA increased the level of peroxisome proliferator-activated receptor α (PPARα) in the brain of Sandhoff mice and that oral CA remained unable to reduce glycoconjugates, improve behavior and increase survival in Sandhoff mice lacking PPARα. Our results indicate a beneficial function of CA that utilizes a PPARα-dependent mechanism to halt the progression of SD and thereby increase the longevity of Sandhoff mice.
Collapse
Affiliation(s)
- Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
11
|
Paciello F, Pisani A, Rolesi R, Montuoro R, Mohamed-Hizam V, Boni G, Ripoli C, Galli J, Sisto R, Fetoni AR, Grassi C. Oxidative stress and inflammation cause auditory system damage via glial cell activation and dysregulated expression of gap junction proteins in an experimental model of styrene-induced oto/neurotoxicity. J Neuroinflammation 2024; 21:4. [PMID: 38178142 PMCID: PMC10765700 DOI: 10.1186/s12974-023-02996-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/13/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Redox imbalance and inflammation have been proposed as the principal mechanisms of damage in the auditory system, resulting in functional alterations and hearing loss. Microglia and astrocytes play a crucial role in mediating oxidative/inflammatory injury in the central nervous system; however, the role of glial cells in the auditory damage is still elusive. OBJECTIVES Here we investigated glial-mediated responses to toxic injury in peripheral and central structures of the auditory pathway, i.e., the cochlea and the auditory cortex (ACx), in rats exposed to styrene, a volatile compound with well-known oto/neurotoxic properties. METHODS Male adult Wistar rats were treated with styrene (400 mg/kg daily for 3 weeks, 5/days a week). Electrophysiological, morphological, immunofluorescence and molecular analyses were performed in both the cochlea and the ACx to evaluate the mechanisms underlying styrene-induced oto/neurotoxicity in the auditory system. RESULTS We showed that the oto/neurotoxic insult induced by styrene increases oxidative stress in both cochlea and ACx. This was associated with macrophages and glial cell activation, increased expression of inflammatory markers (i.e., pro-inflammatory cytokines and chemokine receptors) and alterations in connexin (Cxs) and pannexin (Panx) expression, likely responsible for dysregulation of the microglia/astrocyte network. Specifically, we found downregulation of Cx26 and Cx30 in the cochlea, and high level of Cx43 and Panx1 in the ACx. CONCLUSIONS Collectively, our results provide novel evidence on the role of immune and glial cell activation in the oxidative/inflammatory damage induced by styrene in the auditory system at both peripheral and central levels, also involving alterations of gap junction networks. Our data suggest that targeting glial cells and connexin/pannexin expression might be useful to attenuate oxidative/inflammatory damage in the auditory system.
Collapse
Affiliation(s)
- Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Anna Pisani
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rolando Rolesi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Raffaele Montuoro
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Giammarco Boni
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Jacopo Galli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Renata Sisto
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, Rome, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Unit of Audiology, Università Degli Studi di Napoli Federico II, Naples, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| |
Collapse
|
12
|
Raha S, Dutta D, Paidi RK, Pahan K. Lipid-Lowering Drug Gemfibrozil Protects Mice from Tay-Sachs Disease via Peroxisome Proliferator-Activated Receptor α. Cells 2023; 12:2791. [PMID: 38132111 PMCID: PMC10741479 DOI: 10.3390/cells12242791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/16/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Tay-Sachs disease (TSD) is a progressive heritable neurodegenerative disorder characterized by the deficiency of the lysosomal β-hexosaminidase enzyme (Hex-/-) and the storage of GM2 ganglioside, as well as other related glycoconjugates. Along with motor difficulties, TSD patients also manifest a gradual loss of skills and behavioral problems, followed by early death. Unfortunately, there is no cure for TSD; however, research on treatments and therapeutic approaches is ongoing. This study underlines the importance of gemfibrozil (GFB), an FDA-approved lipid-lowering drug, in inhibiting the disease process in a transgenic mouse model of Tay-Sachs. Oral administration of GFB significantly suppressed glial activation and inflammation, while also reducing the accumulation of GM2 gangliosides/glycoconjugates in the motor cortex of Tay-Sachs mice. Furthermore, oral GFB improved behavioral performance and increased the life expectancy of Tay-Sachs mice. While investigating the mechanism, we found that oral administration of GFB increased the level of peroxisome proliferator-activated receptor α (PPARα) in the brain of Tay-Sachs mice, and that GFB remained unable to reduce glycoconjugates and improve behavior and survival in Tay-Sachs mice lacking PPARα. Our results indicate a beneficial function of GFB that employs a PPARα-dependent mechanism to halt the progression of TSD and increase longevity in Tay-Sachs mice.
Collapse
Affiliation(s)
- Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; (S.R.); (D.D.); (R.K.P.)
| | - Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; (S.R.); (D.D.); (R.K.P.)
| | - Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; (S.R.); (D.D.); (R.K.P.)
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA; (S.R.); (D.D.); (R.K.P.)
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Plesa AM, Shadpour M, Boyden E, Church GM. Transcriptomic reprogramming for neuronal age reversal. Hum Genet 2023; 142:1293-1302. [PMID: 37004545 PMCID: PMC10066999 DOI: 10.1007/s00439-023-02529-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/24/2023] [Indexed: 04/04/2023]
Abstract
Aging is a progressive multifaceted functional decline of a biological system. Chronic age-related conditions such as neurodegenerative diseases are leading causes of death worldwide, and they are becoming a pressing problem for our society. To address this global challenge, there is a need for novel, safe, and effective rejuvenation therapies aimed at reversing age-related phenotypes and improving human health. With gene expression being a key determinant of cell identity and function, and in light of recent studies reporting rejuvenation effects through genetic perturbations, we propose an age reversal strategy focused on reprogramming the cell transcriptome to a youthful state. To this end, we suggest using transcriptomic data from primary human cells to predict rejuvenation targets and develop high-throughput aging assays, which can be used in large perturbation screens. We propose neural cells as particularly relevant targets for rejuvenation due to substantial impact of neurodegeneration on human frailty. Of all cell types in the brain, we argue that glutamatergic neurons, neuronal stem cells, and oligodendrocytes represent the most impactful and tractable targets. Lastly, we provide experimental designs for anti-aging reprogramming screens that will likely enable the development of neuronal age reversal therapies, which hold promise for dramatically improving human health.
Collapse
Affiliation(s)
- Alexandru M. Plesa
- Department of Genetics, Harvard Medical School, Boston, MA USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA USA
| | - Michael Shadpour
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA USA
- Department of Biological Engineering, MIT, Cambridge, MA USA
| | - Ed Boyden
- Department of Biological Engineering, MIT, Cambridge, MA USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA USA
- Howard Hughes Medical Institute, MIT, Cambridge, MA USA
| | - George M. Church
- Department of Genetics, Harvard Medical School, Boston, MA USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA USA
| |
Collapse
|
14
|
Puty B, Bittencourt LO, Lima LAO, Plaça JR, Dionizio A, Buzalaf MAR, Gomes BD, de Oliveira EHC, Lima RR. Unraveling molecular characteristic of fluoride neurotoxicity on U87 glial-like cells: insights from transcriptomic and proteomic approach. Front Cell Neurosci 2023; 17:1153198. [PMID: 37362003 PMCID: PMC10289037 DOI: 10.3389/fncel.2023.1153198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
The potential of fluoride (F) as a neurotoxicant in humans is still controversial in the literature. However, recent studies have raised the debate by showing different mechanism of F-induced neurotoxicity, as oxidative stress, energy metabolism and inflammation in the central nervous system (CNS). In the present study, we investigated the mechanistic action of two F concentration (0.095 and 0.22 μg/ml) on gene and protein profile network using a human glial cell in vitro model over 10 days of exposure. A total of 823 genes and 2,084 genes were modulated after exposure to 0.095 and 0.22 μg/ml F, respectively. Among them, 168 were found to be modulated by both concentrations. The number of changes in protein expression induced by F were 20 and 10, respectively. Gene ontology annotations showed that the main terms were related to cellular metabolism, protein modification and cell death regulation pathways, such as the MAP kinase (MAPK) cascade, in a concentration independent manner. Proteomics confirmed the changes in energy metabolism and also provided evidence of F-induced changes in cytoskeleton components of glial cells. Our results not only reveal that F has the potential to modulate gene and protein profiles in human U87 glial-like cells overexposed to F, but also identify a possible role of this ion in cytoskeleton disorganization.
Collapse
Affiliation(s)
- Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Cell Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leidiane Alencar Oliveira Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Jéssica Rodrigues Plaça
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, Centro de Pesquisa, Inovacão e Desenvolvimento/Fundacão de Amparo á Pesuisa do Estado de São Paulo (CEPID/FAPESP), Ribeirão Preto, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | - Bruno Duarte Gomes
- Laboratory of Neurophysiology Eduardo Oswaldo Cruz, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | | | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| |
Collapse
|
15
|
Fabbri R, Cacopardo L, Ahluwalia A, Magliaro C. Advanced 3D Models of Human Brain Tissue Using Neural Cell Lines: State-of-the-Art and Future Prospects. Cells 2023; 12:1181. [PMID: 37190089 PMCID: PMC10136913 DOI: 10.3390/cells12081181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Human-relevant three-dimensional (3D) models of cerebral tissue can be invaluable tools to boost our understanding of the cellular mechanisms underlying brain pathophysiology. Nowadays, the accessibility, isolation and harvesting of human neural cells represents a bottleneck for obtaining reproducible and accurate models and gaining insights in the fields of oncology, neurodegenerative diseases and toxicology. In this scenario, given their low cost, ease of culture and reproducibility, neural cell lines constitute a key tool for developing usable and reliable models of the human brain. Here, we review the most recent advances in 3D constructs laden with neural cell lines, highlighting their advantages and limitations and their possible future applications.
Collapse
Affiliation(s)
- Rachele Fabbri
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
| | - Ludovica Cacopardo
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of 3R Principles in Teaching and Research (Centro 3R), Italy
| | - Arti Ahluwalia
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of 3R Principles in Teaching and Research (Centro 3R), Italy
| | - Chiara Magliaro
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of 3R Principles in Teaching and Research (Centro 3R), Italy
| |
Collapse
|
16
|
SAJI R, UCHIO R, FUWA A, OKUDA-HANAFUSA C, KAWASAKI K, MUROYAMA K, MUROSAKI S, YAMAMOTO Y, HIROSE Y. Turmeronols (A and B) from Curcuma longa have anti-inflammatory effects in lipopolysaccharide-stimulated BV-2 microglial cells by reducing NF-κB signaling. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 42:172-179. [PMID: 37404570 PMCID: PMC10315188 DOI: 10.12938/bmfh.2022-071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/10/2023] [Indexed: 07/06/2023]
Abstract
Turmeronols (A and B), bisabolane-type sesquiterpenoids found in turmeric, reduce inflammation outside the brain in animals; however, their effects on neuroinflammation, a common pathology of various neurodegenerative diseases, are not understood. Inflammatory mediators produced by microglial cells play a key role in neuroinflammation, so this study evaluated the anti-inflammatory effects of turmeronols in BV-2 microglial cells stimulated with lipopolysaccharide (LPS). Pretreatment with turmeronol A or B significantly inhibited LPS-induced nitric oxide (NO) production; mRNA expression of inducible NO synthase; production of interleukin (IL)-1β, IL-6, and tumor necrosis factor α and upregulation of their mRNA expression; phosphorylation of nuclear factor-κB (NF-κB) p65 proteins and inhibitor of NF-κB kinase (IKK); and nuclear translocation of NF-κB. These results suggest that these turmeronols may prevent the production of inflammatory mediators by inhibiting the IKK/NF-κB signaling pathway in activated microglial cells and can potentially treat neuroinflammation associated with microglial activation.
Collapse
Affiliation(s)
- Ryosuke SAJI
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Ryusei UCHIO
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Arisa FUWA
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Chinatsu OKUDA-HANAFUSA
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Kengo KAWASAKI
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Koutarou MUROYAMA
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Shinji MUROSAKI
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Yoshihiro YAMAMOTO
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| | - Yoshitaka HIROSE
- Research & Development Institute, House Wellness Foods
Corporation, 3-20 Imoji, Itami-shi, Hyogo 664-0011, Japan
| |
Collapse
|
17
|
Akram MA, Wei Q, Ascoli GA. Machine learning classification reveals robust morphometric biomarker of glial and neuronal arbors. J Neurosci Res 2023; 101:112-129. [PMID: 36196621 PMCID: PMC9828050 DOI: 10.1002/jnr.25131] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/06/2022] [Accepted: 09/20/2022] [Indexed: 01/12/2023]
Abstract
Neurons and glia are the two main cell classes in the nervous systems of most animals. Although functionally distinct, neurons and glia are both characterized by multiple branching arbors stemming from the cell bodies. Glial processes are generally known to form smaller trees than neuronal dendrites. However, the full extent of morphological differences between neurons and glia in multiple species and brain regions has not yet been characterized, nor is it known whether these cells can be reliably distinguished based on geometric features alone. Here, we show that multiple supervised learning algorithms deployed on a large database of morphological reconstructions can systematically classify neuronal and glial arbors with nearly perfect accuracy and precision. Moreover, we report multiple morphometric properties, both size related and size independent, that differ substantially between these cell types. In particular, we newly identify an individual morphometric measurement, Average Branch Euclidean Length that can robustly separate neurons from glia across multiple animal models, a broad diversity of experimental conditions, and anatomical areas, with the notable exception of the cerebellum. We discuss the practical utility and physiological interpretation of this discovery.
Collapse
Affiliation(s)
- Masood A. Akram
- Center for Neural Informatics, Structures & PlasticityKrasnow Institute for Advanced StudyCollege of Engineering and ComputingGeorge Mason UniversityFairfaxVirginiaUSA
- Department of BioengineeringVolgenau School of EngineeringGeorge Mason UniversityFairfaxVirginiaUSA
| | - Qi Wei
- Department of BioengineeringVolgenau School of EngineeringGeorge Mason UniversityFairfaxVirginiaUSA
| | - Giorgio A. Ascoli
- Center for Neural Informatics, Structures & PlasticityKrasnow Institute for Advanced StudyCollege of Engineering and ComputingGeorge Mason UniversityFairfaxVirginiaUSA
- Department of BioengineeringVolgenau School of EngineeringGeorge Mason UniversityFairfaxVirginiaUSA
| |
Collapse
|
18
|
Soligo M, Manni L, Conti G, Chiaretti A. Intranasal nerve growth factor for prevention and recovery of the outcomes of traumatic brain injury. Neural Regen Res 2023; 18:773-778. [DOI: 10.4103/1673-5374.354513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
19
|
Basic Fibroblast Growth Factor Induces Cholinergic Differentiation of Tonsil-Derived Mesenchymal Stem Cells. Tissue Eng Regen Med 2022; 19:1063-1075. [PMID: 35857260 DOI: 10.1007/s13770-022-00474-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are considered a potential tool for regenerating damaged tissues due to their great multipotency into various cell types. Here, we attempted to find the appropriate conditions for neuronal differentiation of tonsil-derived MSCs (TMSCs) and expand the potential application of TMSCs for treating neurological diseases. METHODS The TMSCs were differentiated in DMEM/F-12 (Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12) supplemented with various neurotrophic factors for 7-28 days to determine the optimal neuronal differentiation condition for the TMSCs. The morphologies as well as the levels of the neural markers and neurotransmitters were assessed to determine neuronal differentiation potentials and the neuronal lineages of the differentiated TMSCs. RESULTS Our initial study demonstrated that DMEM/F12 supplemented with 50 ng/mL basic fibroblast growth factor with 10 μM forskolin was the optimal condition for neuronal differentiation for the TMSCs. TMSCs had higher protein expression of neuronal markers, including neuron-specific enolase (NSE), GAP43, postsynaptic density protein 95 (PSD95), and synaptosomal-associated protein of 25 kDa (SNAP25) compared to the undifferentiated TMSCs. Immunofluorescence staining also validated the increased mature neuron markers, NeuN and synaptophysin, in the differentiated TMSCs. The expression of glial fibrillar acidic protein and ionized calcium-binding adaptor molecule 1 the markers of astrocytes and microglia, were also slightly increased. Additionally, the differentiated TMSCs released a significantly higher level of acetylcholine, the cholinergic neurotransmitter, as analyzed by the liquid chromatography-tandem mass spectrometry and showed an enhanced choline acetyltransferase immunoreactivity compared to the undifferentiated cells. CONCLUSION Our study suggests that the optimized condition favors the TMSCs to differentiate into cholinergic neuron-like phenotype, which could be used as a possible therapeutic tool in treating certain neurological disorders such as Alzheimer's disease.
Collapse
|
20
|
Sousa AH, Pereira JPG, Malaquias AC, Sagica FDES, de Oliveira EHC. Intracellular accumulation and DNA damage caused by methylmercury in glial cells. J Biochem Mol Toxicol 2022; 36:e23170. [PMID: 35822649 DOI: 10.1002/jbt.23170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/14/2022] [Accepted: 07/01/2022] [Indexed: 11/10/2022]
Abstract
Mercury is widely used in industrial and extractive processes, and the improper disposal of waste or products containing this metal produces a significant impact on ecosystems, causing adverse effects on living organisms, including humans. Exposure to methylmercury, a highly toxic organic compound, causes important neurological and developmental impairments. Recently, the genotoxicity of mercurial compounds has gained prominence as one of the possible mechanisms associated with the neurological effects of mercury, mostly by disturbing the mitotic spindle and causing chromosome loss. In this sense, it is important to investigate if these compounds can also cause direct damage to DNA, such as single and double-strand breaks. Thus, the aim of this study was to investigate the cytotoxic and genotoxic potential of methylmercury in cell lines derived from neurons (B103) and glia (C6), exposed to methylmercury (MeHg) for 24 h, by analyzing cell viability, metabolic activity, and damage to DNA and chromosomes. We found that in comparison to the neuronal cell line, glial cells showed higher tolerance to MeHg, and therefore a higher LC50 and consequent higher intracellular accumulation of Hg, which led to the occurrence of several genotoxic effects, as evidenced by the presence of micronuclei, bridges, sprouts, and chromosomal aberrations.
Collapse
Affiliation(s)
- Aline H Sousa
- Programa de Pós Graduação em Epidemiologia e vigilância em Saúde, Instituto Evandro Chagas, Ananindeua, Pará, Brazil.,Seção de Bacteriologia, Instituto Evandro Chagas, Ananindeua, Pará, Brazil
| | - João P G Pereira
- Programa de Pós Graduação em Epidemiologia e vigilância em Saúde, Instituto Evandro Chagas, Ananindeua, Pará, Brazil.,Seção de Meio Ambiente, Instituto Evandro Chagas, Ananindeua, Pará, Brazil
| | - Allan C Malaquias
- Faculdade de Medicina, Universidade Federal do Pará, Campus de Altamira, Pará, Brazil
| | | | - Edivaldo H C de Oliveira
- Seção de Meio Ambiente, Instituto Evandro Chagas, Ananindeua, Pará, Brazil.,Faculdade de Ciências Naturais, ICEN, Universidade Federal do Pará, Belém, Pará, Brazil
| |
Collapse
|
21
|
Wang QQ, Yin G, Huang JR, Xi SJ, Qian F, Lee RX, Peng XC, Tang FR. Ionizing Radiation-Induced Brain Cell Aging and the Potential Underlying Molecular Mechanisms. Cells 2021; 10:3570. [PMID: 34944078 PMCID: PMC8700624 DOI: 10.3390/cells10123570] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/10/2023] Open
Abstract
Population aging is occurring rapidly worldwide, challenging the global economy and healthcare services. Brain aging is a significant contributor to various age-related neurological and neuropsychological disorders, including Alzheimer's disease and Parkinson's disease. Several extrinsic factors, such as exposure to ionizing radiation, can accelerate senescence. Multiple human and animal studies have reported that exposure to ionizing radiation can have varied effects on organ aging and lead to the prolongation or shortening of life span depending on the radiation dose or dose rate. This paper reviews the effects of radiation on the aging of different types of brain cells, including neurons, microglia, astrocytes, and cerebral endothelial cells. Further, the relevant molecular mechanisms are discussed. Overall, this review highlights how radiation-induced senescence in different cell types may lead to brain aging, which could result in the development of various neurological and neuropsychological disorders. Therefore, treatment targeting radiation-induced oxidative stress and neuroinflammation may prevent radiation-induced brain aging and the neurological and neuropsychological disorders it may cause.
Collapse
Affiliation(s)
- Qin-Qi Wang
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Gang Yin
- Department of Neurology, Jingzhou Central Hospital, Jingzhou 434023, China;
| | - Jiang-Rong Huang
- Health Science Center, Department of Integrative Medicine, School of Health Sciences, Yangtze University, Jingzhou 434023, China;
| | - Shi-Jun Xi
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Health Science Center, Department of Physiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China;
| | - Rui-Xue Lee
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore;
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, Health Science Center, School of Basic Medicine, Yangtze University, Jingzhou 434023, China; (Q.-Q.W.); (S.-J.X.)
- Health Science Center, Department of Pathophysiology, School of Basic Medicine, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore;
| |
Collapse
|
22
|
Manni L, Conti G, Chiaretti A, Soligo M. Intranasal Delivery of Nerve Growth Factor in Neurodegenerative Diseases and Neurotrauma. Front Pharmacol 2021; 12:754502. [PMID: 34867367 PMCID: PMC8635100 DOI: 10.3389/fphar.2021.754502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/01/2021] [Indexed: 01/01/2023] Open
Abstract
Since the 1980s, the development of a pharmacology based on nerve growth factor (NGF) has been postulated for the therapy of Alzheimer’s disease (AD). This hypothesis was based on the rescuing effect of the neurotrophin on the cholinergic phenotype of the basal forebrain neurons, primarily compromised during the development of AD. Subsequently, the use of NGF was put forward to treat a broader spectrum of neurological conditions affecting the central nervous system, such as Parkinson’s disease, degenerative retinopathies, severe brain traumas and neurodevelopmental dysfunctions. While supported by solid rational assumptions, the progress of a pharmacology founded on these hypotheses has been hampered by the difficulty of conveying NGF towards the brain parenchyma without resorting to invasive and risky delivery methods. At the end of the last century, it was shown that NGF administered intranasally to the olfactory epithelium was able to spread into the brain parenchyma. Notably, after such delivery, pharmacologically relevant concentration of exogenous NGF was found in brain areas located at considerable distances from the injection site along the rostral-caudal axis. These observations paved the way for preclinical characterization and clinical trials on the efficacy of intranasal NGF for the treatment of neurodegenerative diseases and of the consequences of brain trauma. In this review, a summary of the preclinical and clinical studies published to date will be attempted, as well as a discussion about the mechanisms underlying the efficacy and the possible development of the pharmacology based on intranasal conveyance of NGF to the brain.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Giorgio Conti
- Department of Emergency, Intensive Pediatric Therapy and Pediatric Trauma Center, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Department of Woman and Child Health, Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
23
|
Kikel-Coury NL, Brandt JP, Correia IA, O’Dea MR, DeSantis DF, Sterling F, Vaughan K, Ozcebe G, Zorlutuna P, Smith CJ. Identification of astroglia-like cardiac nexus glia that are critical regulators of cardiac development and function. PLoS Biol 2021; 19:e3001444. [PMID: 34793438 PMCID: PMC8601506 DOI: 10.1371/journal.pbio.3001444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Glial cells are essential for functionality of the nervous system. Growing evidence underscores the importance of astrocytes; however, analogous astroglia in peripheral organs are poorly understood. Using confocal time-lapse imaging, fate mapping, and mutant genesis in a zebrafish model, we identify a neural crest-derived glial cell, termed nexus glia, which utilizes Meteorin signaling via Jak/Stat3 to drive differentiation and regulate heart rate and rhythm. Nexus glia are labeled with gfap, glast, and glutamine synthetase, markers that typically denote astroglia cells. Further, analysis of single-cell sequencing datasets of human and murine hearts across ages reveals astrocyte-like cells, which we confirm through a multispecies approach. We show that cardiac nexus glia at the outflow tract are critical regulators of both the sympathetic and parasympathetic system. These data establish the crucial role of glia on cardiac homeostasis and provide a description of nexus glia in the PNS.
Collapse
Affiliation(s)
- Nina L. Kikel-Coury
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Jacob P. Brandt
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Isabel A. Correia
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Michael R. O’Dea
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Dana F. DeSantis
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Felicity Sterling
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Kevin Vaughan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Gulberk Ozcebe
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J. Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
24
|
Bozdemir E, Vigil FA, Chun SH, Espinoza L, Bugay V, Khoury SM, Holstein DM, Stoja A, Lozano D, Tunca C, Sprague SM, Cavazos JE, Brenner R, Liston TE, Shapiro MS, Lechleiter JD. Neuroprotective Roles of the Adenosine A 3 Receptor Agonist AST-004 in Mouse Model of Traumatic Brain Injury. Neurotherapeutics 2021; 18:2707-2721. [PMID: 34608616 PMCID: PMC8804149 DOI: 10.1007/s13311-021-01113-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2021] [Indexed: 10/20/2022] Open
Abstract
Traumatic brain injury (TBI) remains one of the greatest public health concerns with increasing morbidity and mortality rates worldwide. Our group reported that stimulation of astrocyte mitochondrial metabolism by P2Y1 receptor agonists significantly reduced cerebral edema and reactive gliosis in a TBI model. Subsequent data on the pharmacokinetics (PK) and rapid metabolism of these compounds suggested that neuroprotection was likely mediated by a metabolite, AST-004, which binding data indicated was an adenosine A3 receptor (A3R) agonist. The neuroprotective efficacy of AST-004 was tested in a control closed cortical injury (CCCI) model of TBI in mice. Twenty-four (24) hours post-injury, mice subjected to CCCI and treated with AST-004 (0.22 mg/kg, injected 30 min post-trauma) exhibited significantly less secondary brain injury. These effects were quantified with less cell death (PSVue794 fluorescence) and loss of blood brain barrier breakdown (Evans blue extravasation assay), compared to vehicle-treated TBI mice. TBI-treated mice also exhibited significantly reduced neuroinflammatory markers, glial-fibrillary acidic protein (GFAP, astrogliosis) and ionized Ca2+-binding adaptor molecule 1 (Iba1, microgliosis), both at the mRNA (qRT-PCR) and protein (Western blot and immunofluorescence) levels, respectively. Four (4) weeks post-injury, both male and female TBI mice presented a significant reduction in freezing behavior during contextual fear conditioning (after foot shock). AST-004 treatment prevented this TBI-induced impairment in male mice, but did not significantly affect impairment in female mice. Impairment of spatial memory, assessed 24 and 48 h after the initial fear conditioning, was also reduced in AST-004-treated TBI-male mice. Female TBI mice did not exhibit memory impairment 24 and 48 h after contextual fear conditioning and similarly, AST-004-treated female TBI mice were comparable to sham mice. Finally, AST-004 treatments were found to increase in vivo ATP production in astrocytes (GFAP-targeted luciferase activity), consistent with the proposed mechanism of action. These data reveal AST-004 as a novel A3R agonist that increases astrocyte energy production and enhances their neuroprotective efficacy after brain injury.
Collapse
Affiliation(s)
- Eda Bozdemir
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Fabio A. Vigil
- Department of Cellular and Integrative Physiology, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Sang H. Chun
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Liliana Espinoza
- Department of Cellular and Integrative Physiology, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Vladislav Bugay
- Department of Cellular and Integrative Physiology, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Sarah M. Khoury
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Deborah M. Holstein
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Aiola Stoja
- Department of Cellular and Integrative Physiology, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Damian Lozano
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Ceyda Tunca
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Shane M. Sprague
- Department of Neurosurgery, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Jose E. Cavazos
- Department of Neurology, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Robert Brenner
- Department of Cellular and Integrative Physiology, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - Theodore E. Liston
- Astrocyte Pharmaceuticals Inc, 245 First Street, Suite 1800, Cambridge, MA 02142 USA
| | - Mark S. Shapiro
- Department of Cellular and Integrative Physiology, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| | - James D. Lechleiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX 78229-3904 USA
| |
Collapse
|
25
|
Kim JH, Park SH, Han J, Ko PW, Kwon D, Suk K. Gliome database: a comprehensive web-based tool to access and analyze glia secretome data. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2021; 2020:5879255. [PMID: 32743661 PMCID: PMC7396318 DOI: 10.1093/database/baaa057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/12/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022]
Abstract
Glial cells are phenotypically heterogeneous non-neuronal components of the central and peripheral nervous systems. These cells are endowed with diverse functions and molecular machineries to detect and regulate neuronal or their own activities by various secreted mediators, such as proteinaceous factors. In particular, glia-secreted proteins form a basis of a complex network of glia-neuron or glia-glia interactions in health and diseases. In recent years, the analysis and profiling of glial secretomes have raised new expectations for the diagnosis and treatment of neurological disorders due to the vital role of glia in numerous physiological or pathological processes of the nervous system. However, there is no online database of glia-secreted proteins available to facilitate glial research. Here, we developed a user-friendly 'Gliome' database (available at www.gliome.org), a web-based tool to access and analyze glia-secreted proteins. The database provides a vast collection of information on 3293 proteins that are released from glia of multiple species and have been reported to have differential functions under diverse experimental conditions. It contains a web-based interface with the following four key features regarding glia-secreted proteins: (i) fundamental information, such as signal peptide, SecretomeP value, functions and Gene Ontology category; (ii) differential expression patterns under distinct experimental conditions; (iii) disease association; and (iv) interacting proteins. In conclusion, the Gliome database is a comprehensive web-based tool to access and analyze glia-secretome data obtained from diverse experimental settings, whereby it may facilitate the integration of bioinformatics into glial research.
Collapse
Affiliation(s)
- Jong-Heon Kim
- Brain Science and Engineering Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Su-Hyeong Park
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.,D&P BIOTECH, 807 Hoguk-ro, Buk-gu, Daegu, 41404, Republic of Korea
| | - Jin Han
- Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Pan-Woo Ko
- Department of Neurology, Kyungpook National University Chilgok Hospital, 807 Hoguk-ro, Buk-gu, Daegu, 41404, Republic of Korea
| | - Dongseop Kwon
- School of Software Convergence, Myongji University, 34 Geobukgol-ro, Seodaemun-gu, Seoul, 03674, Republic of Korea
| | - Kyoungho Suk
- Brain Science and Engineering Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.,Department of Pharmacology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| |
Collapse
|
26
|
Kohrman D, Borges BC, Cassinotti L, Ji L, Corfas G. Axon-glia interactions in the ascending auditory system. Dev Neurobiol 2021; 81:546-567. [PMID: 33561889 PMCID: PMC9004231 DOI: 10.1002/dneu.22813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 11/09/2022]
Abstract
The auditory system detects and encodes sound information with high precision to provide a high-fidelity representation of the environment and communication. In mammals, detection occurs in the peripheral sensory organ (the cochlea) containing specialized mechanosensory cells (hair cells) that initiate the conversion of sound-generated vibrations into action potentials in the auditory nerve. Neural activity in the auditory nerve encodes information regarding the intensity and frequency of sound stimuli, which is transmitted to the auditory cortex through the ascending neural pathways. Glial cells are critical for precise control of neural conduction and synaptic transmission throughout the pathway, allowing for the precise detection of the timing, frequency, and intensity of sound signals, including the sub-millisecond temporal fidelity is necessary for tasks such as sound localization, and in humans, for processing complex sounds including speech and music. In this review, we focus on glia and glia-like cells that interact with hair cells and neurons in the ascending auditory pathway and contribute to the development, maintenance, and modulation of neural circuits and transmission in the auditory system. We also discuss the molecular mechanisms of these interactions, their impact on hearing and on auditory dysfunction associated with pathologies of each cell type.
Collapse
Affiliation(s)
- David Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Luis Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, 1150 West. Medical Center Dr., Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Puty B, Bittencourt LO, Nogueira IC, Buzalaf MAR, Oliveira EH, Lima RR. Human cultured IMR-32 neuronal-like and U87 glial-like cells have different patterns of toxicity under fluoride exposure. PLoS One 2021; 16:e0251200. [PMID: 34138870 PMCID: PMC8211231 DOI: 10.1371/journal.pone.0251200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/21/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Fluoride (F) is a naturally exists in nature but several studies have indicated it as an environmental toxicant to all leaving beings. Human F exposure has increased over the years since this ion has been used by industry on foods, beverages, toothpastes and on water supply. Although F is safe at optimal concentrations in water supply, human exposure to high levels could trigger neurofunctional deficits. MATERIALS AND METHODS In this study, human glial-like (U87) and neuronal-like (IMR-32) cells lineages were used to access F toxicity and CNS cell sensibility on both cell facing the same protocol. Cells were exposed to F over 3, 5 and 10 days on two different F concentrations. Fluoride exposed cells were evaluated by standard toxicity assays to cell viability, apoptosis, necrosis and general cell metabolism. Oxidative stress parameters were evaluated by ATP and ROS levels, lipid peroxidation, GSH/GSSG ratio and comet assay. RESULTS No changes were observed in IMR-32 at any given time while after 10 days of exposure to 0.22μg/mL, U87 glial-like cells showed signs of toxicity such as decreased cell viability by necrosis while general cell metabolism was increased. Oxidative stress parameters were next evaluated only on U87 glial-like cells after 10 days of exposure. F induced a decrease on ATP levels while no changes were observed on reactive oxygen species and lipid peroxidation. GSH/GSSG ratio was decreased followed by DNA damage both on 0.22μg/mL F. CONCLUSIONS Our results suggest an important differential behavior of the distinct types of cells exposed to the different fluoride concentrations, pointing that the U87 glial-like cells as more susceptible to damage triggered by this ion.
Collapse
Affiliation(s)
- Bruna Puty
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- Laboratory of Cell Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Iago Cesar Nogueira
- Laboratory of Cell Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | | | - Edivaldo Herculano Oliveira
- Laboratory of Cell Culture and Cytogenetics, Environmental Section, Evandro Chagas Institute, Ananindeua, Brazil
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| |
Collapse
|
28
|
Oligodendrocyte Development and Regenerative Therapeutics in Multiple Sclerosis. Life (Basel) 2021; 11:life11040327. [PMID: 33918664 PMCID: PMC8069894 DOI: 10.3390/life11040327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/23/2022] Open
Abstract
Myelination by oligodendrocytes (OLs) is an important biological process essential for central nervous system (CNS) development and functions. Oligodendroglial lineage cells undergo several morphological and molecular changes at different stages of their lineage progression into myelinating OLs. The transition steps of the oligodendrocyte progenitor cells (OPCs) to myelinating oligodendrocytes are defined by a specific pattern of regulated gene expression, which is under the control of coordinated signaling pathways. Any abnormal development, loss or failure of oligodendrocytes to myelinate axons can lead to several neurodegenerative diseases like multiple sclerosis (MS). MS is characterized by inflammation and demyelination, and current treatments target only the immune component of the disease, but have little impact on remyelination. Recently, several pharmacological compounds enhancing remyelination have been identified and some of them are in clinical trials. Here, we will review the current knowledge on oligodendrocyte differentiation, myelination and remyelination. We will focus on MS as a pathological condition, the most common chronic inflammatory demyelinating disease of the CNS in young adults.
Collapse
|
29
|
Wang J, Sha Y, Sun T. m 6A Modifications Play Crucial Roles in Glial Cell Development and Brain Tumorigenesis. Front Oncol 2021; 11:611660. [PMID: 33718165 PMCID: PMC7943831 DOI: 10.3389/fonc.2021.611660] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/11/2021] [Indexed: 01/27/2023] Open
Abstract
RNA methylation is a reversible post-transcriptional modification to RNA and has a significant impact on numerous biological processes. N6-methyladenosine (m6A) is known as one of the most common types of eukaryotic mRNA methylation modifications, and exists in a wide variety of organisms, including viruses, yeast, plants, mice, and humans. Widespread and dynamic m6A methylation is identified in distinct developmental stages in the brain, and controls development of neural stem cells and their differentiation into neurons, glial cells such as oligodendrocytes and astrocytes. Here we summarize recent advances in our understanding of RNA methylation regulation in brain development, neurogenesis, gliogenesis, and its dysregulation in brain tumors. This review will highlight biological roles of RNA methylation in development and function of neurons and glial cells, and provide insights into brain tumor formation, and diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Jing Wang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China.,College of Materials Science and Engineering, Huaqiao University, Xiamen, China
| | - Yongqiang Sha
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| |
Collapse
|
30
|
Effects of Pacap on Schwann Cells: Focus on Nerve Injury. Int J Mol Sci 2020; 21:ijms21218233. [PMID: 33153152 PMCID: PMC7663204 DOI: 10.3390/ijms21218233] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 12/27/2022] Open
Abstract
Schwann cells, the most abundant glial cells of the peripheral nervous system, represent the key players able to supply extracellular microenvironment for axonal regrowth and restoration of myelin sheaths on regenerating axons. Following nerve injury, Schwann cells respond adaptively to damage by acquiring a new phenotype. In particular, some of them localize in the distal stump to form the Bungner band, a regeneration track in the distal site of the injured nerve, whereas others produce cytokines involved in recruitment of macrophages infiltrating into the nerve damaged area for axonal and myelin debris clearance. Several neurotrophic factors, including pituitary adenylyl cyclase-activating peptide (PACAP), promote survival and axonal elongation of injured neurons. The present review summarizes the evidence existing in the literature demonstrating the autocrine and/or paracrine action exerted by PACAP to promote remyelination and ameliorate the peripheral nerve inflammatory response following nerve injury.
Collapse
|
31
|
Curry-Hyde A, Gray LG, Chen BJ, Ueberham U, Arendt T, Janitz M. Cell type-specific circular RNA expression in human glial cells. Genomics 2020; 112:5265-5274. [DOI: 10.1016/j.ygeno.2020.09.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/02/2020] [Accepted: 09/19/2020] [Indexed: 12/11/2022]
|
32
|
Demir SA, Timur ZK, Ateş N, Martínez LA, Seyrantepe V. GM2 ganglioside accumulation causes neuroinflammation and behavioral alterations in a mouse model of early onset Tay-Sachs disease. J Neuroinflammation 2020; 17:277. [PMID: 32951593 PMCID: PMC7504627 DOI: 10.1186/s12974-020-01947-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/01/2020] [Indexed: 11/10/2022] Open
Abstract
Background Tay-Sachs disease (TSD), a type of GM2-gangliosidosis, is a progressive neurodegenerative lysosomal storage disorder caused by mutations in the α subunit of the lysosomal β-hexosaminidase enzyme. This disease is characterized by excessive accumulation of GM2 ganglioside, predominantly in the central nervous system. Although Tay-Sachs patients appear normal at birth, the progressive accumulation of undegraded GM2 gangliosides in neurons leads to death. Recently, an early onset Tay-Sachs disease mouse model, with genotype Hexa−/−Neu3−/−, was generated. Progressive accumulation of GM2 led to premature death of the double KO mice. Importantly, this double-deficient mouse model displays typical features of Tay-Sachs patients, such as cytoplasmic vacuolization of nerve cells, deterioration of Purkinje cells, neuronal death, deceleration in movement, ataxia, and tremors. GM2-gangliosidosis is characterized by acute neurodegeneration preceded by activated microglia expansion, macrophage, and astrocyte activation, along with the production of inflammatory mediators. However, the mechanism of disease progression in Hexa−/−Neu3−/− mice, relevant to neuroinflammation is poorly understood. Method In this study, we investigated the onset and progression of neuroinflammatory changes in the cortex, cerebellum, and retina of Hexa−/−Neu3−/− mice and control littermates by using a combination of molecular genetics and immunochemical procedures. Results We found elevated levels of pro-inflammatory cytokine and chemokine transcripts, such as Ccl2, Ccl3, Ccl4, and Cxcl10 and also extensive microglial and astrocyte activation and proliferation, accompanied by peripheral blood mononuclear cell infiltration in the vicinity of neurons and oligodendrocytes. Behavioral tests demonstrated a high level of anxiety, and age-dependent loss in both spatial learning and fear memory in Hexa−/−Neu3−/− mice compared with that in the controls. Conclusion Altogether, our data suggest that Hexa−/−Neu3−/− mice display a phenotype similar to Tay-Sachs patients suffering from chronic neuroinflammation triggered by GM2 accumulation. Furthermore, our work contributes to better understanding of the neuropathology in a mouse model of early onset Tay-Sachs disease.
Collapse
Affiliation(s)
- Seçil Akyıldız Demir
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Zehra Kevser Timur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Nurselin Ateş
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Luis Alarcón Martínez
- Institute of Neurological Science and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey.
| |
Collapse
|
33
|
Tai W, Xu XM, Zhang CL. Regeneration Through in vivo Cell Fate Reprogramming for Neural Repair. Front Cell Neurosci 2020; 14:107. [PMID: 32390804 PMCID: PMC7193690 DOI: 10.3389/fncel.2020.00107] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/07/2020] [Indexed: 12/30/2022] Open
Abstract
The adult mammalian central nervous system (CNS) has very limited regenerative capacity upon neural injuries or under degenerative conditions. In recent years, however, significant progress has been made on in vivo cell fate reprogramming for neural regeneration. Resident glial cells can be reprogrammed into neuronal progenitors and mature neurons in the CNS of adult mammals. In this review article, we briefly summarize the current knowledge on innate adult neurogenesis under pathological conditions and then focus on induced neurogenesis through cell fate reprogramming. We discuss how the reprogramming process can be regulated and raise critical issues requiring careful considerations to move the field forward. With emerging evidence, we envision that fate reprogramming-based regenerative medicine will have a great potential for treating neurological conditions such as brain injury, spinal cord injury (SCI), Alzheimer's disease (AD), Parkinson's disease (PD), and retinopathy.
Collapse
Affiliation(s)
- Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
34
|
Hong C, Jeong B, Park HJ, Chung JY, Lee JE, Kim J, Shin YC, So I. TRP Channels as Emerging Therapeutic Targets for Neurodegenerative Diseases. Front Physiol 2020; 11:238. [PMID: 32351395 PMCID: PMC7174697 DOI: 10.3389/fphys.2020.00238] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
The development of treatment for neurodegenerative diseases (NDs) such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis is facing medical challenges due to the increasingly aging population. However, some pharmaceutical companies have ceased the development of therapeutics for NDs, and no new treatments for NDs have been established during the last decade. The relationship between ND pathogenesis and risk factors has not been completely elucidated. Herein, we review the potential involvement of transient receptor potential (TRP) channels in NDs, where oxidative stress and disrupted Ca2+ homeostasis consequently lead to neuronal apoptosis. Reactive oxygen species (ROS) -sensitive TRP channels can be key risk factors as polymodal sensors, since progressive late onset with secondary pathological damage after initial toxic insult is one of the typical characteristics of NDs. Recent evidence indicates that the dysregulation of TRP channels is a missing link between disruption of Ca2+ homeostasis and neuronal loss in NDs. In this review, we discuss the latest findings regarding TRP channels to provide insights into the research and quests for alternative therapeutic candidates for NDs. As the structures of TRP channels have recently been revealed by cryo-electron microscopy, it is necessary to develop new TRP channel antagonists and reevaluate existing drugs.
Collapse
Affiliation(s)
- Chansik Hong
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Byeongseok Jeong
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Hyung Joon Park
- Department of Physiology, Chosun University School of Medicine, Gwangju, South Korea
| | - Ji Yeon Chung
- Department of Neurology, Chosun University School of Medicine, Gwangju, South Korea
| | - Jung Eun Lee
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jinsung Kim
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Young-Cheul Shin
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
| | - Insuk So
- Department of Physiology and Institute of Dermatological Science, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
35
|
Xia QP, Cheng ZY, He L. The modulatory role of dopamine receptors in brain neuroinflammation. Int Immunopharmacol 2019; 76:105908. [PMID: 31622861 DOI: 10.1016/j.intimp.2019.105908] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 01/11/2023]
Abstract
Neuroinflammation is a general pathological feature of central nervous system (CNS) diseases, primarily caused by activation of astrocytes and microglia, as well as the infiltration of peripheral immune cells. Inhibition of neuroinflammation is an important strategy in the treatment of brain disorders. Dopamine (DA) receptor, a significant G protein-coupled receptor (GPCR), is classified into two families: D1-like (D1 and D5) and D2-like (D2, D3 and D4) receptor families, according to their downstream signaling pathways. Traditionally, DA receptor forms a wide variety of psychological activities and motor functions, such as voluntary movement, working memory and learning. Recently, the role of DA receptor in neuroinflammation has been investigated widely, mainly focusing on nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome, renin-angiotensin system, αB-crystallin, as well as invading peripheral immune cells, including T cells, dendritic cells, macrophages and monocytes. This review briefly outlined the functions and signaling pathways of DA receptor subtypes as well as its role in inflammation-related glial cells, and subsequently summarized the mechanisms of DA receptors affecting neuroinflammation. Meaningfully, this article provided a theoretical basis for drug development targeting DA receptors in inflammation-related brain diseases.
Collapse
Affiliation(s)
- Qing-Peng Xia
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhao-Yan Cheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
36
|
Chen JC, Lee IN, Huang C, Wu YP, Chung CY, Lee MH, Lin MHC, Yang JT. Valproic acid-induced amphiregulin secretion confers resistance to temozolomide treatment in human glioma cells. BMC Cancer 2019; 19:756. [PMID: 31370819 PMCID: PMC6670223 DOI: 10.1186/s12885-019-5843-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/16/2019] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most severe type of primary brain tumor with a high mortality rate. Although extensive treatments for GBM, including resection, irradiation, chemotherapy and immunotherapy, have been tried, the prognosis is still poor. Temozolomide (TMZ), an alkylating agent, is a front-line chemotherapeutic drug for the clinical treatment of GBM; however, its effects are very limited because of the chemoresistance. Valproic acid (VPA), an antiepileptic agent with histone deacetylase inhibitor activity, has been shown to have synergistic effects with TMZ against GBM. The mechanism of action of VPA on TMZ combination therapy is still unclear. Accumulating evidence has shown that secreted proteins are responsible for the cross talking among cells in the tumor microenvironment, which may play a critical role in the regulation of drug responses. METHODS To understand the effect of VPA on secreted proteins in GBM cells, we first used the antibody array to analyze the cell culture supernatant from VPA-treated and untreated GBM cells. The results were further confirmed by lentivirus-mediated knockdown and exogenous recombinant administration. RESULTS Our results showed that amphiregulin (AR) was highly secreted in VPA-treated cells. Knockdown of AR can sensitize GBM cells to TMZ. Furthermore, pretreatment of exogenous recombinant AR significantly increased EGFR activation and conferred resistance to TMZ. To further verify the effect of AR on TMZ resistance, cells pre-treated with AR neutralizing antibody markedly increased sensitivity to TMZ. In addition, we also observed that the expression of AR was positively correlated with the resistance of TMZ in different GBM cell lines. CONCLUSIONS The present study aimed to identify the secreted proteins that contribute to the modulation of drug response. Understanding the full set of secreted proteins present in glial cells might help reveal potential therapeutic opportunities. The results indicated that AR may potentially serve as biomarker and therapeutic approach for chemotherapy regimens in GBM.
Collapse
Affiliation(s)
- Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi City, 60004 Taiwan
| | - I-Neng Lee
- Department of Medical Research, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Cheng Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Earth and Life Sciences, University of Taipei, Taipei, Taiwan
| | - Yu-Ping Wu
- Department of Medical Research, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Chiu-Yen Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Ming-Hsueh Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Martin Hsiu-Chu Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chiayi, 61363 Taiwan
- College of Medicine, Chang Gung University, Tao-Yuan, 33302 Taiwan
| |
Collapse
|
37
|
Kantawong F, Saksiriwisitkul C, Riyapa C, Limpakdee S, Wanachantararak P, Kuboki T. Reprogramming of mouse fibroblasts into neural lineage cells using biomaterials. ACTA ACUST UNITED AC 2018; 8:129-138. [PMID: 29977834 PMCID: PMC6026523 DOI: 10.15171/bi.2018.15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 12/30/2017] [Accepted: 01/04/2018] [Indexed: 11/24/2022]
Abstract
![]()
Introduction: Induced neural stem cells (iNSCs) have the ability of differentiation into neurons, astrocytes and oligodendrocytes. iNSCs are very useful in terms of research and treatment. The present study offers an idea that biomaterials could be one of the tools that could modulate reprogramming process in the fibroblasts.
Methods: Gelatin biomaterials were fabricated into 3 types, including (i) gelatin, (ii) gelatin with 1 mg/mL hydroxyapatite, and (iii) gelatin with hydroxyapatite and pig brain. NIH/3T3 fibroblasts were cultured on each type of biomaterial for 7, 9 and 14 days. RT-PCR was performed to investigate the gene expression of the fibroblasts on biomaterials compared to the fibroblasts on tissue culture plates. PI3K/Akt signaling was performed by flow cytometry after 24 hours seeding on the biomaterials. The biomaterials were also tested with the human APCs and PDL cells.
Results: The fibroblasts exhibited changes in the expression of the reprogramming factor; Klf4 and the neural transcription factors; NFIa, NFIb and Ptbp1 after 9 days culture. The cultivation of fibroblasts on the biomaterials for 7 days showed a higher expression of the transcription factor SOX9. The expression of epigenetic genes; Kat2a and HDAC3 were changed upon the cultivation on the biomaterials for 9 days. The fibroblasts cultured on the biomaterials showed an activation of PI3K/Akt signaling. The human APCs and human PDL cells developed mineralization process on biomaterials
Conclusion: Changes in the expression of Klf4, NFIa, NFIb, Ptbp1 and SOX9 indicated that fibroblasts were differentiated into an astrocytic lineage. It is possible that the well-designed biomaterials could work as powerful tools in the reprogramming process of fibroblasts into iNSCs.
Collapse
Affiliation(s)
- Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Chanidapa Saksiriwisitkul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Chanakan Riyapa
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Suchalinee Limpakdee
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | - Thasaneeya Kuboki
- Laboratory of Biomedical and Biophysical Chemistry, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| |
Collapse
|
38
|
|
39
|
Twiss JL, Fainzilber M. Neuroproteomics: How Many Angels can be Identified in an Extract from the Head of a Pin? Mol Cell Proteomics 2016; 15:341-3. [PMID: 26729708 DOI: 10.1074/mcp.e116.057828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Indexed: 11/06/2022] Open
Affiliation(s)
- Jeffery L Twiss
- From the ‡Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208;
| | - Mike Fainzilber
- §Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|