1
|
Ergul Erkec O, Huyut Z, Acikgoz E, Huyut MT. Effects of exogenous ghrelin treatment on oxidative stress, inflammation and histological parameters in a fat-fed streptozotocin rat model. Arch Physiol Biochem 2024:1-11. [PMID: 39324977 DOI: 10.1080/13813455.2024.2407551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/17/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
In this study, the anti-inflammatory, antioxidative, and protective effects of ghrelin were investigated in a fat-fed streptozotocin (STZ) rat model and compared with metformin, diabetes and the healthy control groups. Histopathological evaluations were performed on H&E-stained pancreas and brain sections. Biochemical parameters were investigated by enzyme-linked immunosorbent assay. Blood glucose levels were significantly decreased with ghrelin or metformin treatments than the diabetes group. STZ administration increased brain, renal and pancreatic IL-1β, TNF-α and MDA while decreasing GPX, CAT, SOD, and NGF levels. Ghrelin increased renal GPX, CAT, NGF pancreatic GPX, SOD, CAT, NGF and brain SOD, NGF while it decreased renal, pancreatic and brain IL-1β, TNF-α and MDA levels. Ghrelin reduced neuronal loss and degeneration in the cerebral cortex and hippocampus and greatly ameliorated diabetes-related damage in pancreas. In conclusion, the data suggested that ghrelin is an effective candidate as a protectant for reducing the adverse effects of diabetes.
Collapse
Affiliation(s)
- Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mehmet Tahir Huyut
- Department of Biostatistics, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| |
Collapse
|
2
|
Maytalman E, Nemutlu Samur D. Neuroendocrine modulation by metamizole and indomethacin: investigating the impact on neuronal markers and GnRH release. Endocrine 2024; 85:1327-1336. [PMID: 38625503 DOI: 10.1007/s12020-024-03822-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/02/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE Some evidence that non-steroidal anti-inflammatory drugs have neuroprotective effects indicates their potential for use in a new field. However, their effects on hormone secretion have yet to be adequately discovered. Therefore, we aimed to evaluate the effects of metamizole and indomethacin on neuronal markers as well as the GnRH expression in the GT1-7 cell line. METHODS The effects of these drugs on proliferation were evaluated by MTT analysis. The effect of 10-50-250 µM concentrations of the drugs also on the expression of neuronal factors and markers, including NGF, nestin and βIII Tubulin, and additionally GnRH, was determined by the RT-qPCR method. RESULTS NGF and nestin mRNA expressions were increased in all concentrations of both metamizole and indomethacin. No changes were detected in βIII Tubulin. While metamizole showed an increase in GnRH mRNA expression, there was no change at 10 and 50 µM concentrations of indomethacin, but a remarkable decrease was observed at 250 µM concentrations. CONCLUSIONS The results of our study showing an increase in the expression of neuronal factors reveal that metamizole and indomethacin may have possible neuroprotective effects. Moreover, the effects on the GnRH expression appear to be different. Animal models are required to confirm these effects of NSAIDs on neurons.
Collapse
Affiliation(s)
- Erkan Maytalman
- Department of Pharmacology, School of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey.
| | - Dilara Nemutlu Samur
- Department of Pharmacology, School of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| |
Collapse
|
3
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [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: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
4
|
Li J, Yang M, Dai Y, Guo X, Ding Y, Li X, Zhang S, Xu W, Chen L, Tao J, Liu W. Electroacupuncture regulates Rab5a-mediating NGF transduction to improve learning and memory ability in the early stage of AD mice. CNS Neurosci Ther 2024; 30:e14743. [PMID: 38780008 PMCID: PMC11112630 DOI: 10.1111/cns.14743] [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: 08/20/2023] [Revised: 03/22/2024] [Accepted: 04/11/2024] [Indexed: 05/25/2024] Open
Abstract
AIMS Nerve growth factor (NGF) loss is a potential factor for the degeneration of basal forebrain cholinergic neurons (BFCNs) in Alzheimer's disease (AD), and Rab5a is a key regulatory molecule of NGF signaling transduction. Here, we investigated the changes of Rab5a in 5 × FAD mice and further explored the mechanism of Electroacupuncture (EA) treatment in improving cognition in the early stage of AD. METHODS The total Rab5a and Rab5a-GTP in 5-month-old 5 × FAD mice and wild-type mice were detected using WB and IP technologies. 5 × FAD mice were treated with EA at the Bai hui (DU20) and Shen ting (DU24) acupoints for 4 weeks and CRE/LOXP technology was used to confirm the role of Rab5a in AD mediated by EA stimulation. The Novel Object Recognition and Morris water maze tests were used to evaluate the cognitive function of 5 × FAD mice. The Nissl, immunohistochemistry, and Thioflavin S staining were used to observe pathological morphological changes in the basal forebrain circuit. The Golgi staining was used to investigate the synaptic plasticity of the basal forebrain circuit and WB technology was used to detect the expression levels of cholinergic-related and NGF signal-related proteins. RESULTS The total Rab5a was unaltered, but Rab5a-GTP increased and the rab5a-positive early endosomes appeared enlarged in the hippocampus of 5 × FAD mice. Notably, EA reduced Rab5a-GTP in the hippocampus in the early stage of 5 × FAD mice. EA could improve object recognition memory and spatial learning memory by reducing Rab5a activity in the early stage of 5 × FAD mice. Moreover, EA could reduce Rab5a activity to increase NGF transduction and increase the levels of phosphorylated TrkA, AKT, and ERK in the basal forebrain and hippocampus, and increase the expression of cholinergic-related proteins, such as ChAT, vAchT, ChT1, m1AchR, and m2AchR in the basal forebrain and ChAT, m1AchR, and m2AchR in the hippocampus, improving synaptic plasticity in the basal forebrain hippocampal circuit in the early stage of 5 × FAD mice. CONCLUSIONS Rab5a hyperactivation is an early pathological manifestation of 5 × FAD mice. EA could suppress Rab5a-GTP to promote the transduction of NGF signaling, and enhance the synaptic plasticity of the basal forebrain hippocampal circuit improving cognitive impairment in the early stage of 5 × FAD mice.
Collapse
Affiliation(s)
- Jianhong Li
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
- Fujian Key Laboratory of Aptamers Technology900TH hospital of Joint Logistics Support ForceFuzhouChina
| | - Minguang Yang
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
| | - Yaling Dai
- National‐Local Joint Engineering Research Center of Rehabilitation Medicine TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Xiaoqin Guo
- National‐Local Joint Engineering Research Center of Rehabilitation Medicine TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Yanyi Ding
- National‐Local Joint Engineering Research Center of Rehabilitation Medicine TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Xiaoling Li
- Provincial and Ministerial Co‐founded Collaborative Innovation Center of Rehabilitation TechnologyFujian University of Traditional Chinese MedicineFuzhouChina
| | - Shenghang Zhang
- Fujian Key Laboratory of Aptamers Technology900TH hospital of Joint Logistics Support ForceFuzhouChina
| | - Wenshan Xu
- Fujian Key Laboratory of Cognitive RehabilitationAffiliated Rehabilitation Hospital of Fujian University of Traditional Chinese MedicineFuzhouChina
| | - Lidian Chen
- Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese MedicineFujian University of Traditional Chinese MedicineFuzhouChina
| | - Jing Tao
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
| | - Weilin Liu
- The Institute of Rehabilitation IndustryFujian University of Traditional Chinese MedicineFuzhouChina
| |
Collapse
|
5
|
Bove M, Palmieri MA, Santoro M, Agosti LP, Gaetani S, Romano A, Dimonte S, Costantino G, Sikora V, Tucci P, Schiavone S, Morgese MG, Trabace L. Amygdalar neurotransmission alterations in the BTBR mice model of idiopathic autism. Transl Psychiatry 2024; 14:193. [PMID: 38632257 PMCID: PMC11024334 DOI: 10.1038/s41398-024-02905-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Autism Spectrum Disorders (ASD) are principally diagnosed by three core behavioural symptoms, such as stereotyped repertoire, communication impairments and social dysfunctions. This complex pathology has been linked to abnormalities of corticostriatal and limbic circuits. Despite experimental efforts in elucidating the molecular mechanisms behind these abnormalities, a clear etiopathogenic hypothesis is still lacking. To this aim, preclinical studies can be really helpful to longitudinally study behavioural alterations resembling human symptoms and to investigate the underlying neurobiological correlates. In this regard, the BTBR T+ Itpr3tf/J (BTBR) mice are an inbred mouse strain that exhibits a pattern of behaviours well resembling human ASD-like behavioural features. In this study, the BTBR mice model was used to investigate neurochemical and biomolecular alterations, regarding Nerve Growth Factor (NGF) and Brain-Derived Neurotrophic Factor (BDNF), together with GABAergic, glutamatergic, cholinergic, dopaminergic and noradrenergic neurotransmissions and their metabolites in four different brain areas, i.e. prefrontal cortex, hippocampus, amygdala and hypothalamus. In our results, BTBR strain reported decreased noradrenaline, acetylcholine and GABA levels in prefrontal cortex, while hippocampal measurements showed reduced NGF and BDNF expression levels, together with GABA levels. Concerning hypothalamus, no differences were retrieved. As regarding amygdala, we found reduced dopamine levels, accompanied by increased dopamine metabolites in BTBR mice, together with decreased acetylcholine, NGF and GABA levels and enhanced glutamate content. Taken together, our data showed that the BTBR ASD model, beyond its face validity, is a useful tool to untangle neurotransmission alterations that could be underpinned to the heterogeneous ASD-like behaviours, highlighting the crucial role played by amygdala.
Collapse
Affiliation(s)
- Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Maria Adelaide Palmieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Martina Santoro
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Lisa Pia Agosti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Giuseppe Costantino
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
- Department of Pathology, Sumy State University, 40007, Sumy, Ukraine
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy.
| |
Collapse
|
6
|
Kumro J, Tripathi A, Terry AV, Pillai A, Blake DT. α7 nicotinic acetylcholine receptors are necessary for basal forebrain activation to increase expression of the nerve growth factor receptor TrkA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582932. [PMID: 38463995 PMCID: PMC10925259 DOI: 10.1101/2024.03.01.582932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Activation of the basal forebrain leads to increases in the expression of the nerve growth factor receptor, Tropomyosin receptor kinase A (TrkA) and decreases in expression of the beta amyloid cleavage enzyme 1 (BACE1) in the cerebral cortex of both sexes of 5xFAD mice. The studies described in this report were designed to determine if these changes were dependent on acetylcholine receptors. Mice were stimulated unilaterally in the basal forebrain for two weeks. Animals were administered a cholinergic antagonist, or saline, 30 minutes prior to stimulation. Animals administered saline exhibited significant increases in TrkA expression and decreases in BACE1 in the stimulated hemisphere relative to the unstimulated. While both nonselective nicotinic and muscarinic acetylcholine receptor blockade attenuated the BACE1 decline, only the nicotinic receptor antagonism blocked the TrkA increase. Next, we applied selective nicotinic antagonists, and the α7 antagonist blocked the TrkA increases, but the α4β2 antagonist did not. BACE1 declines were not blocked by either intervention. Mice with a loxP conditional knockout of the gene for the α7 nicotinic receptor were also employed in these studies. Animals were either stimulated bilaterally for two weeks, or left unstimulated. With or without stimulation, the expression of TrkA receptors was lower in the cortical region with the α7 nicotinic receptor knockdown. We thus conclude that α7 nicotinic receptor activation is necessary for normal expression of TrkA and increases caused by basal forebrain activation, while BACE1 declines caused by stimulation have dependency on a broader array of receptor subtypes.
Collapse
Affiliation(s)
- Jacob Kumro
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Ashutosh Tripathi
- Dept Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX
| | - Alvin V Terry
- Dept Pharmacology/Toxicology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Anilkumar Pillai
- Dept Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA
- Dept Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA
| | - David T Blake
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
7
|
Xiong HY, Hendrix J, Schabrun S, Wyns A, Campenhout JV, Nijs J, Polli A. The Role of the Brain-Derived Neurotrophic Factor in Chronic Pain: Links to Central Sensitization and Neuroinflammation. Biomolecules 2024; 14:71. [PMID: 38254671 PMCID: PMC10813479 DOI: 10.3390/biom14010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Chronic pain is sustained, in part, through the intricate process of central sensitization (CS), marked by maladaptive neuroplasticity and neuronal hyperexcitability within central pain pathways. Accumulating evidence suggests that CS is also driven by neuroinflammation in the peripheral and central nervous system. In any chronic disease, the search for perpetuating factors is crucial in identifying therapeutic targets and developing primary preventive strategies. The brain-derived neurotrophic factor (BDNF) emerges as a critical regulator of synaptic plasticity, serving as both a neurotransmitter and neuromodulator. Mounting evidence supports BDNF's pro-nociceptive role, spanning from its pain-sensitizing capacity across multiple levels of nociceptive pathways to its intricate involvement in CS and neuroinflammation. Moreover, consistently elevated BDNF levels are observed in various chronic pain disorders. To comprehensively understand the profound impact of BDNF in chronic pain, we delve into its key characteristics, focusing on its role in underlying molecular mechanisms contributing to chronic pain. Additionally, we also explore the potential utility of BDNF as an objective biomarker for chronic pain. This discussion encompasses emerging therapeutic approaches aimed at modulating BDNF expression, offering insights into addressing the intricate complexities of chronic pain.
Collapse
Affiliation(s)
- Huan-Yu Xiong
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
| | - Jolien Hendrix
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, 3000 Leuven, Belgium
- Research Foundation—Flanders (FWO), 1000 Brussels, Belgium
| | - Siobhan Schabrun
- The School of Physical Therapy, University of Western Ontario, London, ON N6A 3K7, Canada;
- The Gray Centre for Mobility and Activity, Parkwood Institute, London, ON N6A 4V2, Canada
| | - Arne Wyns
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
| | - Jente Van Campenhout
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
| | - Jo Nijs
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
- Chronic Pain Rehabilitation, Department of Physical Medicine and Physiotherapy, University Hospital Brussels, 1090 Brussels, Belgium
- Department of Health and Rehabilitation, Unit of Physiotherapy, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Göterbog, Sweden
| | - Andrea Polli
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (H.-Y.X.); (J.H.); (A.W.); (J.V.C.); (A.P.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, 3000 Leuven, Belgium
- Research Foundation—Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
8
|
Bonomini F, Favero G, Castrezzati S, Borsani E. Role of Neurotrophins in Orofacial Pain Modulation: A Review of the Latest Discoveries. Int J Mol Sci 2023; 24:12438. [PMID: 37569811 PMCID: PMC10419393 DOI: 10.3390/ijms241512438] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Orofacial pain represents a multidisciplinary biomedical challenge involving basic and clinical research for which no satisfactory solution has been found. In this regard, trigeminal pain is described as one of the worst pains perceived, leaving the patient with no hope for the future. The aim of this review is to evaluate the latest discoveries on the involvement of neurotrophins in orofacial nociception, describing their role and expression in peripheral tissues, trigeminal ganglion, and trigeminal nucleus considering their double nature as "supporters" of the nervous system and as "promoters" of nociceptive transmission. In order to scan recent literature (last ten years), three independent researchers referred to databases PubMed, Embase, Google Scholar, Scopus, and Web of Science to find original research articles and clinical trials. The researchers selected 33 papers: 29 original research articles and 4 clinical trials. The results obtained by the screening of the selected articles show an interesting trend, in which the precise modulation of neurotrophin signaling could switch neurotrophins from being a "promoter" of pain to their beneficial neurotrophic role of supporting the nerves in their recovery, especially when a structural alteration is present, as in neuropathic pain. In conclusion, neurotrophins could be interesting targets for orofacial pain modulation but more studies are necessary to clarify their role for future application in clinical practice.
Collapse
Affiliation(s)
- Francesca Bonomini
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.B.); (G.F.); (S.C.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.B.); (G.F.); (S.C.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Stefania Castrezzati
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.B.); (G.F.); (S.C.)
| | - Elisa Borsani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.B.); (G.F.); (S.C.)
- Interdepartmental University Center of Research “Adaptation and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| |
Collapse
|
9
|
Ananth MR, Rajebhosale P, Kim R, Talmage DA, Role LW. Basal forebrain cholinergic signalling: development, connectivity and roles in cognition. Nat Rev Neurosci 2023; 24:233-251. [PMID: 36823458 PMCID: PMC10439770 DOI: 10.1038/s41583-023-00677-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 01/18/2023] [Indexed: 02/25/2023]
Abstract
Acetylcholine plays an essential role in fundamental aspects of cognition. Studies that have mapped the activity and functional connectivity of cholinergic neurons have shown that the axons of basal forebrain cholinergic neurons innervate the pallium with far more topographical and functional organization than was historically appreciated. Together with the results of studies using new probes that allow release of acetylcholine to be detected with high spatial and temporal resolution, these findings have implicated cholinergic networks in 'binding' diverse behaviours that contribute to cognition. Here, we review recent findings on the developmental origins, connectivity and function of cholinergic neurons, and explore the participation of cholinergic signalling in the encoding of cognition-related behaviours.
Collapse
Affiliation(s)
- Mala R Ananth
- Section on Circuits, Synapses, and Molecular Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Prithviraj Rajebhosale
- Section on Genetics of Neuronal Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ronald Kim
- Section on Genetics of Neuronal Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - David A Talmage
- Section on Genetics of Neuronal Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Lorna W Role
- Section on Circuits, Synapses, and Molecular Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
10
|
Chavushyan VA, Simonyan KV, Danielyan MH, Avetisyan LG, Darbinyan LV, Isoyan AS, Lorikyan AG, Hovhannisyan LE, Babakhanyan MA, Sukiasyan LM. Pathology and prevention of brain microvascular and neuronal dysfunction induced by a high-fructose diet in rats. Metab Brain Dis 2023; 38:269-286. [PMID: 36271967 DOI: 10.1007/s11011-022-01098-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 10/08/2022] [Indexed: 02/03/2023]
Abstract
A high-fructose diet causes metabolic abnormalities in rats, and the cluster of complications points to microvascular and neuronal disorders of the brain. The aim of this study was to evaluate i) the involvement of microvascular disorders and neuronal plasticity in the deleterious effects of a high-fructose diet on the rat brain and ii) a comparative assessment of the effectiveness of Phytocollection therapy (with antidiabetic, antioxidant, and acetylcholinesterase inhibitory activities) compared to Galantamine as first-line therapy for dementia and Diabeton as first-line therapy for hyperglycemia. The calcium adenosine triphosphate non-injection histoangiological method was used to assess capillary network diameter and density. A high-fructose diet resulted in a significant decrease in the diameter and density of the capillary bed, and pharmacological manipulations had a modulatory effect on microcirculatory adaptive mechanisms. In vivo single-unit extracellular recording was used to investigate short-term plasticity in the medial prefrontal cortex. Differences in the parameters of spike background activity and expression of excitatory and inhibitory responses of cortical neurons have been discovered, allowing for flexibility and neuronal function stabilization in pathology and pharmacological prevention. Integration of the coupling mechanism between microvascular function and neuronal spike activity could delay the progressive decline in cognitive function in rats fed a high fructose diet.
Collapse
Affiliation(s)
- V A Chavushyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - K V Simonyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia.
| | - M H Danielyan
- Histochemistry and Electron Microscopy Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - L G Avetisyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - L V Darbinyan
- Sensorimotor Integration Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - A S Isoyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - A G Lorikyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - L E Hovhannisyan
- G.S. Davtyan Institute of Hydroponics Problems NAS RA, 0082, Yerevan, Armenia
| | - M A Babakhanyan
- G.S. Davtyan Institute of Hydroponics Problems NAS RA, 0082, Yerevan, Armenia
| | - L M Sukiasyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
- Yerevan State Medical University After M. Heratsi, 0025, Yerevan, Armenia
| |
Collapse
|
11
|
Hussein A, Tielemans A, Baxter MG, Benson DL, Huntley GW. Cognitive deficits and altered cholinergic innervation in young adult male mice carrying a Parkinson's disease Lrrk2 G2019S knockin mutation. Exp Neurol 2022; 355:114145. [PMID: 35732218 PMCID: PMC9338764 DOI: 10.1016/j.expneurol.2022.114145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 06/14/2022] [Indexed: 01/29/2023]
Abstract
Impaired executive function is a common and debilitating non-motor symptom of idiopathic and hereditary Parkinson's disease (PD), but there is little understanding of the underlying pathophysiological mechanisms and circuits. The G2019S mutation in the kinase domain of leucine-rich repeat kinase 2 (LRRK2) greatly increases risk for late-onset PD, and non-manifesting LRRK2G2019S carriers can also exhibit early and significant cognitive impairment. Here, we subjected young adult male mice carrying a Lrrk2G2019S knockin mutation to touchscreen-based operant tasks that measure attention, goal-directed learning and cognitive flexibility, all of which rely on frontal-striatal connectivity and are strongly modulated by cholinergic innervation. In a visuospatial attention task, mutant mice exhibited significantly more omissions and longer response latencies than controls that could not be attributed to deficits in motivation, visual sensory perception per se or locomotion, thereby suggesting impairments in divided attention and/or action-selection as well as generally slower information processing speed. Pretreating mice with the acetylcholinesterase inhibitor donepezil normalized both higher omission rates and longer response latencies in the mutants, but did not affect any performance metric in controls. Strikingly, cholinergic fiber density in cortical areas PL/IL and DMS (dorsomedial striatum) was significantly sparser in mutants than in controls, while further behavioral interrogation of the mutants revealed significant impairments in action-outcome associations but preserved cognitive flexibility. These data suggest that the Lrrk2G2019S mutation negatively impacts cholinergic innervation anatomically and functionally by young adulthood, impairing corticostriatal network function in ways that may contribute to early PD-associated executive function deficits.
Collapse
|
12
|
A Microglial Function for the Nerve Growth Factor: Predictions of the Unpredictable. Cells 2022; 11:cells11111835. [PMID: 35681529 PMCID: PMC9180430 DOI: 10.3390/cells11111835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/29/2022] [Accepted: 06/02/2022] [Indexed: 12/10/2022] Open
Abstract
Microglia are the only immune cell population present in the brain parenchyma. Their vantage position in the central nervous system (CNS) enables these myeloid cells to perform the most disparate of tasks: from the classical immune functions of fighting infections and surveilling the extracellular space for pathogens and damage, to sculpting the neuronal circuitry by pruning unnecessary synapses and assisting neurons in spine formation, aiding in the maintenance of brain homeostasis. The neurotrophin field has always been dominated by the neurocentric view that the primary target of these molecules must be neurons: this holds true even for the Nerve Growth Factor (NGF), which owes its popularity in the neuroscience community to its trophic and tropic activity towards sensory and sympathetic neurons in the peripheral nervous system, and cholinergic neurons in the CNS. The increasing evidence that microglia are an integral part of neuronal computation calls for a closer look as to whether these glial cells are capable of responding directly to NGF. In this review, we will first outline evidence in support of a role for NGF as a molecule mediating neuroimmune communication. Then, we will illustrate some of those non-immune features that have made microglial cells one of the hottest topics of this last decade. In conclusion, we will discuss evidence in support of a microglial function for NGF.
Collapse
|
13
|
Zavalin K, Hassan A, Fu C, Delpire E, Lagrange AH. Loss of KCC2 in GABAergic Neurons Causes Seizures and an Imbalance of Cortical Interneurons. Front Mol Neurosci 2022; 15:826427. [PMID: 35370549 PMCID: PMC8966887 DOI: 10.3389/fnmol.2022.826427] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
K-Cl transporter KCC2 is an important regulator of neuronal development and neuronal function at maturity. Through its canonical transporter role, KCC2 maintains inhibitory responses mediated by γ-aminobutyric acid (GABA) type A receptors. During development, late onset of KCC2 transporter activity defines the period when depolarizing GABAergic signals promote a wealth of developmental processes. In addition to its transporter function, KCC2 directly interacts with a number of proteins to regulate dendritic spine formation, cell survival, synaptic plasticity, neuronal excitability, and other processes. Either overexpression or loss of KCC2 can lead to abnormal circuit formation, seizures, or even perinatal death. GABA has been reported to be especially important for driving migration and development of cortical interneurons (IN), and we hypothesized that properly timed onset of KCC2 expression is vital to this process. To test this hypothesis, we created a mouse with conditional knockout of KCC2 in Dlx5-lineage neurons (Dlx5 KCC2 cKO), which targets INs and other post-mitotic GABAergic neurons in the forebrain starting during embryonic development. While KCC2 was first expressed in the INs of layer 5 cortex, perinatal IN migrations and laminar localization appeared to be unaffected by the loss of KCC2. Nonetheless, the mice had early seizures, failure to thrive, and premature death in the second and third weeks of life. At this age, we found an underlying change in IN distribution, including an excess number of somatostatin neurons in layer 5 and a decrease in parvalbumin-expressing neurons in layer 2/3 and layer 6. Our research suggests that while KCC2 expression may not be entirely necessary for early IN migration, loss of KCC2 causes an imbalance in cortical interneuron subtypes, seizures, and early death. More work will be needed to define the specific cellular basis for these findings, including whether they are due to abnormal circuit formation versus the sequela of defective IN inhibition.
Collapse
Affiliation(s)
- Kirill Zavalin
- Department of Neurology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Anjana Hassan
- Department of Neurology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Cary Fu
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eric Delpire
- Department of Anesthesiology, School of Medicine, Vanderbilt University, Nashville, TN, United States
| | - Andre H. Lagrange
- Department of Neurology, School of Medicine, Vanderbilt University, Nashville, TN, United States,Department of Neurology, Tennessee Valley Healthcare – Veterans Affairs (TVH VA), Medical Center, Nashville, TN, United States,*Correspondence: Andre H. Lagrange,
| |
Collapse
|
14
|
Ju IG, Hong SM, Yun SW, Huh E, Kim DH, Kim SY, Oh MS. CCL01, a novel formulation composed of Cuscuta seeds and Lactobacillus paracasei NK112, enhances memory function via nerve growth factor-mediated neurogenesis. Food Funct 2021; 12:10690-10699. [PMID: 34605514 DOI: 10.1039/d1fo01403j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Memory decline occurs due to various factors, including stress, depression, and aging, and lowers the quality of life. Several nutritional supplements and probiotics have been used to enhance memory function, and efforts have been made to develop mixed supplements with maximized efficacy. In this study, we aimed to examine whether a novel formulation composed of Cuscuta seeds and Lactobacillus paracasei NK112, CCL01, enhances memory function and induces neurogenesis via nerve growth factor (NGF) induction. Firstly, we orally administered CCL01 to normal mice and assessed their memory function 4 weeks after the first administration by performing a step-through passive avoidance test. We found that CCL01 at 100 mg kg-1 treatment enhanced the fear-based memory function. By analyzing the expression of Ki-67 and doublecortin, which are the markers of proliferating cells and immature neurons, respectively, we observed that CCL01 induced neuronal proliferation and differentiation in the hippocampus of the mice. Additionally, we found that the expression of synaptic markers increased in the hippocampus of CCL01-treated mice. We measured the NGF expression in the supernatant of C6 cells after CCL01 treatment and found that CCL01 increased NGF release. Furthermore, treatment of CCL01-conditioned glial media on N2a cells increased neuronal differentiation via the TrkA/ERK/CREB signaling pathway and neurotrophic factor expression. Moreover, when CCL01 was administered and scopolamine was injected, CCL01 ameliorated memory decline. These results suggest that CCL01 is an effective enhancer of memory function and can be applied to various age groups requiring memory improvement.
Collapse
Affiliation(s)
- In Gyoung Ju
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Seong Min Hong
- Department of Pharmacy, College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea.
| | - Soo-Won Yun
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eugene Huh
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Sun Yeou Kim
- Department of Pharmacy, College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, Republic of Korea.
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea. .,Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.,Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
NGF Eye Administration Recovers the TrkB and Glutamate/GABA Marker Deficit in the Adult Visual Cortex Following Optic Nerve Crush. Int J Mol Sci 2021; 22:ijms221810014. [PMID: 34576177 PMCID: PMC8471133 DOI: 10.3390/ijms221810014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 12/21/2022] Open
Abstract
Eye-drop recombinant human nerve growth factor (ed-rhNGF) has proved to recover the retina and optic nerve damage in animal models, including the unilateral optic nerve crush (ONC), and to improve visual acuity in humans. These data, associated with evidence that ed-rhNGF stimulates the brain derived neurotrophic factor (BDNF) in retina and cortex, suggests that NGF might exert retino-fugal effects by affecting BDNF and its receptor TrkB. To address these questions, their expression and relationship with the GABAergic and glutamatergic transmission markers, GAD65 and GAD67, vesicular inhibitory amino acid transporter (VGAT), and vesicular glutamate transporters 1 and 2 (VGLUT-1 and VGLUT-2) were investigated in adult ONC rats contralateral and ipsilateral visual cortex (VCx). Ed-rhNGF recovers the ONC-induced alteration of GABAergic and glutamatergic markers in contralateral VCx, induces an upregulation of TrkB, which is positively correlated with BDNF precursor (proBDNF) decrease in both VCx sides, and strongly enhances TrkB+ cell soma and neuronal endings surrounded by GAD65 immuno-reactive afferents. These findings contribute to enlarging the knowledge on the mechanism of actions and cellular targets of exogenously administrated NGF, and suggest that ed-rhNGF might act by potentiating the activity-dependent TrkB expression in GAD+ cells in VCx following retina damage and/or ONC.
Collapse
|
16
|
Soligo M, Felsani FM, Da Ros T, Bosi S, Pellizzoni E, Bruni S, Isopi J, Marcaccio M, Manni L, Fiorito S. Distribution in the brain and possible neuroprotective effects of intranasally delivered multi-walled carbon nanotubes. NANOSCALE ADVANCES 2021; 3:418-431. [PMID: 36131737 PMCID: PMC9418508 DOI: 10.1039/d0na00869a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 11/12/2020] [Indexed: 06/15/2023]
Abstract
Carbon nanotubes (CNTs) are currently under active investigation for their use in several biomedical applications, especially in neurological diseases and nervous system injury due to their electrochemical properties. Nowadays, no CNT-based therapeutic products for internal use appear to be close to the market, due to the still limited knowledge on their fate after delivery to living organisms and, in particular, on their toxicological profile. The purpose of the present work was to address the distribution in the brain parenchyma of two intranasally delivered MWCNTs (MWCNTs 1 and a-MWCNTs 2), different from each other, the first being non electroconductive while the second results in being electroconductive. After intranasal delivery, the presence of CNTs was investigated in several brain areas, discriminating the specific cell types involved in the CNT uptake. We also aimed to verify the neuroprotective potential of the two types of CNTs, delivering them in rats affected by early diabetic encephalopathy and analysing the modulation of nerve growth factor metabolism and the effects of CNTs on the neuronal and glial phenotypes. Our findings showed that both CNT types, when intranasally delivered, reached numerous brain areas and, in particular, the limbic area that plays a crucial role in the development and progression of major neurodegenerative diseases. Furthermore, we demonstrated that electroconductive MWCNTs were able to exert neuroprotective effects through the modulation of a key neurotrophic factor and probably the improvement of neurodegeneration-related gliosis.
Collapse
Affiliation(s)
- Marzia Soligo
- Institute of Translational Pharmacology, CNR Via Fosso del Cavaliere 100 00133 Rome Italy
| | - Fausto Maria Felsani
- Institute of Translational Pharmacology, CNR Via Fosso del Cavaliere 100 00133 Rome Italy
| | - Tatiana Da Ros
- INSTM - Trieste Unit, Department of Chemical and Pharmaceutical Sciences, University of Trieste Via Licio Giorgieri 1 34127 Trieste Italy
| | - Susanna Bosi
- INSTM - Trieste Unit, Department of Chemical and Pharmaceutical Sciences, University of Trieste Via Licio Giorgieri 1 34127 Trieste Italy
| | - Elena Pellizzoni
- INSTM - Trieste Unit, Department of Chemical and Pharmaceutical Sciences, University of Trieste Via Licio Giorgieri 1 34127 Trieste Italy
| | - Stefano Bruni
- Former Medical Director Sanofi - Genzyme, Italy, currently Orchard Therapeutics 108 Cannon Street London UK
| | - Jacopo Isopi
- Department of Chemistry "G. Ciamician", University of Bologna Via Selmi, 2 40126 Bologna Italy
| | - Massimo Marcaccio
- Department of Chemistry "G. Ciamician", University of Bologna Via Selmi, 2 40126 Bologna Italy
| | - Luigi Manni
- Institute of Translational Pharmacology, CNR Via Fosso del Cavaliere 100 00133 Rome Italy
| | - Silvana Fiorito
- Institute of Translational Pharmacology, CNR Via Fosso del Cavaliere 100 00133 Rome Italy
| |
Collapse
|
17
|
Eriksdotter M, Mitra S. Gene and cell therapy for the nucleus basalis of Meynert with NGF in Alzheimer's disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:219-229. [PMID: 34225964 DOI: 10.1016/b978-0-12-819975-6.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is currently no effective treatment for the most common of the dementia disorders, Alzheimer's disease (AD). It has been known for decades that the central cholinergic system is important for memory. The cholinergic neurons in the basal forebrain with its cortical and hippocampal projections degenerate in AD and thus contribute to the cognitive decline characteristic of AD. This knowledge led to the development of the currently approved treatment for AD, with inhibitors of acetylcholine-esterase targeting the cholinergic system with beneficial but mild effects. In recent years, other approaches to influence the degenerating cholinergic system in AD focusing on nerve growth factor (NGF) have been undertaken. NGF is required for the survival and function of the basal forebrain cholinergic neurons, the most important being the nucleus basalis of Meynert (nbM). Since there is a lack of NGF and its receptors in the AD forebrain, the hypothesis is that local delivery of NGF to the nbM could revive the cholinergic circuitry and thereby restore cognitive functions. Since NGF does not pass through the blood-brain barrier, approaches involving cerebral injections of genetically modified cells or viral vectors or implantation of encapsulated cells in the nbM in AD patients have been used. These attempts have been partially successful but also have limitations, which are presented and discussed here. In conclusion, these trials point to the importance of further development of NGF-related therapies in AD.
Collapse
Affiliation(s)
- Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden; Theme Aging, Karolinska University Hospital, Huddinge, Sweden.
| | - Sumonto Mitra
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Moss DE, Perez RG. Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer's Disease: Nexus of Cholinergic and Nerve Growth Factor Dysfunction. Curr Alzheimer Res 2021; 18:1010-1022. [PMID: 34911424 PMCID: PMC8855657 DOI: 10.2174/1567205018666211215150547] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/04/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is increasingly viewed as a complex multi-dimensional disease without effective treatments. Recent randomized, placebo-controlled studies have shown volume losses of ~0.7% and ~3.5% per year, respectively, in the basal cholinergic forebrain (CBF) and hippocampus in untreated suspected prodromal AD. One year of donepezil treatment reduced these annualized rates of atrophy to about half of untreated rates. Similar positive although variable results have also been found in volumetric measurements of the cortex and whole brain in patients with mild cognitive impairment as well as more advanced AD stages after treatments with all three currently available acetylcholinesterase (AChE) inhibitors (donepezil, rivastigmine, and galantamine). Here we review the anti-neurodegenerative benefits of AChE inhibitors and the expected parallel disease-accelerating impairments caused by anticholinergics, within a framework of the cholinergic hypothesis of AD and AD-associated loss of nerve growth factor (NGF). Consistent with the "loss of trophic factor hypothesis of AD," we propose that AChE inhibitors enhance acetylcholine-dependent release and uptake of NGF, thereby sustaining cholinergic neuronal viability and thus slowing AD-associated degeneration of the CBF, to ultimately delay dementia progression. We propose that improved cholinergic therapies for AD started early in asymptomatic persons, especially those with risk factors, will delay the onset, progression, or emergence of dementia. The currently available competitive and pseudo- irreversible AChE inhibitors are not CNS-selective and thus induce gastrointestinal toxicity that limits cortical AChE inhibition to ~30% (ranges from 19% to 41%) as measured by in vivo PET studies in patients undergoing therapy. These levels of inhibition are marginal relative to what is required for effective symptomatic treatment of dementia or slowing AD-associated neurodegeneration. In contrast, because of the inherently slow de novo synthesis of AChE in the CNS (about one-- tenth the rate of synthesis in peripheral tissues), irreversible AChE inhibitors produce significantly higher levels of inhibition in the CNS than in peripheral tissues. For example, methanesulfonyl fluoride, an irreversible inhibitor reduces CNS AChE activity by ~68% in patients undergoing therapy and ~80% in cortical biopsies of non-human primates. The full therapeutic benefits of AChE inhibitors, whether for symptomatic treatment of dementia or disease-slowing, thus would benefit by producing high levels of CNS inhibition. One way to obtain such higher levels of CNS AChE inhibition would be by using irreversible inhibitors.
Collapse
Affiliation(s)
- Donald E. Moss
- Department of Psychology, University of Texas at El Paso, El Paso, Texas, 79968 USA
| | - Ruth G. Perez
- Department of Molecular and Translational Medicine, Center of Emphasis in Neurosciences, Graduate School of Biomedical Sciences Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, 79905 USA
| |
Collapse
|
19
|
Fair SR, Julian D, Hartlaub AM, Pusuluri ST, Malik G, Summerfied TL, Zhao G, Hester AB, Ackerman WE, Hollingsworth EW, Ali M, McElroy CA, Buhimschi IA, Imitola J, Maitre NL, Bedrosian TA, Hester ME. Electrophysiological Maturation of Cerebral Organoids Correlates with Dynamic Morphological and Cellular Development. Stem Cell Reports 2020; 15:855-868. [PMID: 32976764 PMCID: PMC7562943 DOI: 10.1016/j.stemcr.2020.08.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 12/22/2022] Open
Abstract
Cerebral organoids (COs) are rapidly accelerating the rate of translational neuroscience based on their potential to model complex features of the developing human brain. Several studies have examined the electrophysiological and neural network features of COs; however, no study has comprehensively investigated the developmental trajectory of electrophysiological properties in whole-brain COs and correlated these properties with developmentally linked morphological and cellular features. Here, we profiled the neuroelectrical activities of COs over the span of 5 months with a multi-electrode array platform and observed the emergence and maturation of several electrophysiologic properties, including rapid firing rates and network bursting events. To complement these analyses, we characterized the complex molecular and cellular development that gives rise to these mature neuroelectrical properties with immunohistochemical and single-cell transcriptomic analyses. This integrated approach highlights the value of COs as an emerging model system of human brain development and neurological disease. CO electrophysiology can be quantified with a multi-electrode array method CO electrophysiological trajectories correlate with molecular and cellular development The neurotrophin/TRK signaling pathway is active in COs by 5 months in culture
Collapse
Affiliation(s)
- Summer R Fair
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH 43205-2716, USA
| | - Dominic Julian
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH 43205-2716, USA
| | - Annalisa M Hartlaub
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Sai Teja Pusuluri
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Girik Malik
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Khoury College of Computer Sciences, Northeastern University, Boston, MA 02115, USA
| | - Taryn L Summerfied
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Guomao Zhao
- Department of Obstetrics and Gynecology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Arelis B Hester
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH 43205-2716, USA
| | - William E Ackerman
- Department of Obstetrics and Gynecology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Ethan W Hollingsworth
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH 43205-2716, USA
| | - Mehboob Ali
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Craig A McElroy
- College of Pharmacy, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Irina A Buhimschi
- Department of Obstetrics and Gynecology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Jaime Imitola
- Department of Neurology, Laboratory for Neural Stem Cells and Functional Neurogenetics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Nathalie L Maitre
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Tracy A Bedrosian
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH 43205-2716, USA
| | - Mark E Hester
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, OH 43205-2716, USA; Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
20
|
Abstract
Based on the analysis of literature, the authors describe the neuropathophysiological mechanism of the formation of synapses, synaptic transmission and plasticity, which may underlie the pathogenesis of autism. The results of some studies confirm the involvement of aberrant expression of genes and proteins of synaptic contacts, cell adhesion molecules p120ctn, CNTN5, CNTN6, activation of NMDA glutamate, TrkB, p75 receptors, Ca2+-input, BDNF, serotonin and testosterone. This leads to an imbalance in the exciting, inhibitory synaptic transmission and forms of synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD) at the level of individual neurons and their chains due to suppression of GABA synthesis, expression of its ionotropic and metabotropic receptors, G proteins, NGF, TrkA receptors, a reduction in the number of GABAergic neurons, their contacts and disruption of differentiation. The pathology of the nuclei of the thalamus, especially the reticular nucleus (RN), is associated with a disturbance of the expression of the subunits of metabotropic GABAβ receptors, Ca2+ channels, GABA excretion and the work of chlorine transmitters. These failures do not ensure the inhibitory effect of OC on the exciting associative and ventral nuclei of the thalamus, nor modify the incoming information to the cerebral cortex (CC) from these thalamus nuclei, the dentate gyrus of the hippocampus and the nuclei of the reticular formation. Information propagating into the somatosensory and associative regions of CC is not modified by mirror neurons (MN) when performing arbitrary actions, which prevents the formation of an adequate image in the neural networks of the associative cortex and promotes the development of hyperexcitability, irritability, increased visual and auditory sensitivity, anxiety, and the ability to form a holistic image based on the actions of other people.
Collapse
Affiliation(s)
- A N Chernov
- Almazov National Medical Research Center of the Ministry of Health of Russia, St. Petersburg, Russia
| |
Collapse
|
21
|
Gudasheva TA, Povarnina PY, Tarasiuk AV, Seredenin SB. Low-molecular mimetics of nerve growth factor and brain-derived neurotrophic factor: Design and pharmacological properties. Med Res Rev 2020; 41:2746-2774. [PMID: 32808322 DOI: 10.1002/med.21721] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
To overcome the limitations of the clinical use of neurotrophins nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), scientists have been trying to create their low-molecular-weight mimetics having improved pharmacokinetic properties and lacking side effects of full-sized proteins since the 90s of the last century. The efforts of various research groups have led to the production of peptide and nonpeptide mimetics, being agonists or modulators of the corresponding Trk or p75 receptors that reproduced the therapeutic effects of full-sized proteins. This review discusses different strategies and approaches to the design of such compounds. The relationship between the structure of the mimetics obtained and their action mechanisms and pharmacological properties are analyzed. Special attention is paid to the dipeptide mimetics of individual NGF and BDNF loops having different patterns of activation of Trk receptors signal transduction pathways, phosphoinositide 3-kinase/protein kinase B and mitogen-activated protein kinase/extracellular signal-regulated kinase, which allowed to evaluate the contribution of each pathway to different pharmacological effects. In conclusion, data on therapeutically promising compounds being at different stages of preclinical and clinical studies are summarized.
Collapse
Affiliation(s)
- Tatiana A Gudasheva
- Medicinal Chemistry Department, V. V. Zakusov Research Institute of Pharmacology, Moscow, Russian Federation
| | - Polina Y Povarnina
- Medicinal Chemistry Department, V. V. Zakusov Research Institute of Pharmacology, Moscow, Russian Federation
| | - Aleksey V Tarasiuk
- Medicinal Chemistry Department, V. V. Zakusov Research Institute of Pharmacology, Moscow, Russian Federation
| | - Sergey B Seredenin
- Department of Pharmacogenetics, V. V. Zakusov Research Institute of Pharmacology, Moscow, Russian Federation
| |
Collapse
|
22
|
Hesp C. Beyond connectionism: A neuronal dance of ephaptic and synaptic interactions: Commentary on "The growth of cognition: Free energy minimization and the embryogenesis of cortical computation" by Wright and Bourke (2020). Phys Life Rev 2020; 36:40-43. [PMID: 32807647 DOI: 10.1016/j.plrev.2020.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Casper Hesp
- Department of Psychology, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, Netherlands; Amsterdam Brain and Cognition Centre, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, Netherlands; Institute for Advanced Study, University of Amsterdam, Oude Turfmarkt 147, 1012 GC Amsterdam, Netherlands; Wellcome Centre for Human Neuroimaging, University College London, WC1N 3BG, London, UK.
| |
Collapse
|
23
|
Ding XW, Li R, Geetha T, Tao YX, Babu JR. Nerve growth factor in metabolic complications and Alzheimer's disease: Physiology and therapeutic potential. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165858. [PMID: 32531260 DOI: 10.1016/j.bbadis.2020.165858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/11/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
As the population ages, obesity and metabolic complications as well as neurological disorders are becoming more prevalent, with huge economic burdens on both societies and families. New therapeutics are urgently needed. Nerve growth factor (NGF), first discovered in 1950s, is a neurotrophic factor involved in regulating cell proliferation, growth, survival, and apoptosis in both central and peripheral nervous systems. NGF and its precursor, proNGF, bind to TrkA and p75 receptors and initiate protein phosphorylation cascades, resulting in changes of cellular functions, and are associated with obesity, diabetes and its complications, and Alzheimer's disease. In this article, we summarize changes in NGF levels in metabolic and neuronal disorders, the signal transduction initiated by NGF and proNGF, the physiological and pathophysiological relevance, and therapeutic potential in treating chronic metabolic diseases and cognitive decline.
Collapse
Affiliation(s)
- Xiao-Wen Ding
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA
| | - Rongzi Li
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA
| | - Thangiah Geetha
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.
| | - Jeganathan Ramesh Babu
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; Boshell Metabolic Diseases and Diabetes Program, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
24
|
Li IC, Chang HH, Lin CH, Chen WP, Lu TH, Lee LY, Chen YW, Chen YP, Chen CC, Lin DPC. Prevention of Early Alzheimer's Disease by Erinacine A-Enriched Hericium erinaceus Mycelia Pilot Double-Blind Placebo-Controlled Study. Front Aging Neurosci 2020; 12:155. [PMID: 32581767 PMCID: PMC7283924 DOI: 10.3389/fnagi.2020.00155] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To investigate the efficacy and safety of three H. erinaceus mycelia (EAHE) capsules (350 mg/capsule; containing 5 mg/g erinacine A active ingredient) per day for the treatment of patients with mild Alzheimer's Disease (AD). METHODS This study comprised a 3-week no-drug screening period, followed by a 49-week double-blind treatment period with 2-parallel groups in which eligible patients were randomized to either three 5 mg/g EAHE mycelia capsules per day or identical appearing placebo capsules. Cognitive assessments, ophthalmic examinations, biomarker collection, and neuroimaging were followed throughout the study period. RESULTS After 49 weeks of EAHE intervention, a significant decrease in Cognitive Abilities Screening Instrument score was noted in the placebo group, a significant improvement in Mini-Mental State Examination score was observed in the EAHE group and a significant Instrumental Activities of Daily Living score difference were found between the two groups. In addition, EAHE group achieved a significantly better contrast sensitivity when compared to the placebo group. Moreover, only the placebo group observed significantly lowered biomarkers such as calcium, albumin, apolipoprotein E4, hemoglobin, and brain-derived neurotrophic factor and significantly elevated alpha1-antichymotrypsin and amyloid-beta peptide 1-40 over the study period. Using diffusion tensor imaging, the mean apparent diffusion coefficient (ADC) values from the arcuate fasciculus region in the dominant hemisphere significantly increased in the placebo group while no significant difference was found in the EAHE group in comparison to their baselines. Moreover, ADC values from the parahippocampal cingulum region in the dominant hemisphere significantly decreased in the EAHE group whereas no significant difference was found in the placebo group when compared to their baselines. Lastly, except for four subjects who dropped out of the study due to abdominal discomfort, nausea, and skin rash, no other adverse events were reported. CONCLUSION Three 350 mg/g EAHE capsules intervention for 49 weeks demonstrated higher CASI, MMSE, and IADL scores and achieved a better contrast sensitivity in patients with mild AD when compared to the placebo group, suggesting that EAHE is safe, well-tolerated, and may be important in achieving neurocognitive benefits. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier NCT04065061.
Collapse
Affiliation(s)
- I-Chen Li
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Han-Hsin Chang
- Department of Nutrition, Chung Shan Medical University, Taichung City, Taiwan
| | - Chuan-Han Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
| | - Wan-Ping Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Tsung-Han Lu
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
| | - Li-Ya Lee
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Yu-Wen Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Yen-Po Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei City, Taiwan
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei City, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City, Taiwan
| | - David Pei-Cheng Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
25
|
Wang J, Hu WW, Jiang Z, Feng MJ. Advances in treatment of neurodegenerative diseases: Perspectives for combination of stem cells with neurotrophic factors. World J Stem Cells 2020; 12:323-338. [PMID: 32547681 PMCID: PMC7280867 DOI: 10.4252/wjsc.v12.i5.323] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis, are a group of incurable neurological disorders, characterized by the chronic progressive loss of different neuronal subtypes. However, despite its increasing prevalence among the ever-increasing aging population, little progress has been made in the coincident immense efforts towards development of therapeutic agents. Research interest has recently turned towards stem cells including stem cells-derived exosomes, neurotrophic factors, and their combination as potential therapeutic agents in neurodegenerative diseases. In this review, we summarize the progress in therapeutic strategies based on stem cells combined with neurotrophic factors and mesenchymal stem cells-derived exosomes for neurodegenerative diseases, with an emphasis on the combination therapy.
Collapse
Affiliation(s)
- Jie Wang
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
- Department of Neurology, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu Province, China
| | - Wei-Wei Hu
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Zhi Jiang
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
| | - Mei-Jiang Feng
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China
- Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.
| |
Collapse
|
26
|
Rosso P, Iannitelli A, Pacitti F, Quartini A, Fico E, Fiore M, Greco A, Ralli M, Tirassa P. Vagus nerve stimulation and Neurotrophins: a biological psychiatric perspective. Neurosci Biobehav Rev 2020; 113:338-353. [PMID: 32278791 DOI: 10.1016/j.neubiorev.2020.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023]
Abstract
Since 2004, vagus nerve stimulation (VNS) has been used in treatment-resistant or treatment-intolerant depressive episodes. Today, VNS is suggested as possible therapy for a larger spectrum of psychiatric disorders, including schizophrenia, obsessive compulsive disorders, and panic disorders. Despite a large body of literature supports the application of VNS in patients' treatment, the exact mechanism of action of VNS remains not fully understood. In the present study, the major knowledges on the brain areas and neuronal pathways regulating neuroimmune and autonomic response subserving VNS effects are reviewed. Furthermore, the involvement of the neurotrophins (NTs) Nerve Growth Factor (NGF) and Brain Derived Neurotrophic Factor (BDNF) in vagus nerve (VN) physiology and stimulation is revised. The data on brain NGF/BDNF synthesis and in turn on the activity-dependent plasticity, connectivity rearrangement and neurogenesis, are presented and discussed as potential biomarkers for optimizing stimulatory parameters for VNS. A vagus nerve-neurotrophin interaction model in the brain is finally proposed as a working hypothesis for future studies addressed to understand pathophysiology of psychiatric disturbance.
Collapse
Affiliation(s)
- Pamela Rosso
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Angela Iannitelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesca Pacitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy; Psychiatry Unit San Salvatore Hospital, L'Aquila, Italy
| | - Adele Quartini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elena Fico
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Marco Fiore
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Italy
| | - Paola Tirassa
- National Research Council (CNR), Institute of Biochemistry & Cell Biology (IBBC), Rome, Italy.
| |
Collapse
|
27
|
Kumar A, Kumar P, Pareek V, Faiq MA, Narayan RK, Raza K, Prasoon P, Sharma VK. Neurotrophin mediated HPA axis dysregulation in stress induced genesis of psychiatric disorders: Orchestration by epigenetic modifications. J Chem Neuroanat 2019; 102:101688. [PMID: 31568825 DOI: 10.1016/j.jchemneu.2019.101688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/22/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
Apart from their established role in embryonic development, neurotrophins (NTs) have diverse functions in the nervous system. Their role in the integration of physiological and biochemical aspects of the nervous system is currently attracting much attention. Based on a systematic analysis of the literature, we here propose a new paradigm that, by exploiting a novel role of NTs, may help explain the genesis of stress-related psychiatric disorders, opening new avenues for better management of the same. We hypothesize that NTs as an integrated network play a crucial role in maintaining an indivdual's psychological wellbeing. Given the evidence that stress can induce chronic disruption of the hypothalamic-pituitary-adrenal (HPA) axis which, in turn, is causally linked to several psychiatric disorders, this function may be mediated through the homeostatic mechanisms governing regulation of this axis. In fact, NTs, such as nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are known to participate in neuroendocrine regulation. Recent studies suggest epigenetic modification of NT-HPA axis interplay in the precipitation of psychiatric disorders. Our article highlights why this new knowledge regarding NTs should be considered in the etiogenesis and treatment of stress-induced psychopathology.
Collapse
|
28
|
Savaheli S, Ahmadiani A. Obsessive-compulsive disorder and growth factors: A comparative review. Behav Brain Res 2019; 372:111967. [PMID: 31136772 DOI: 10.1016/j.bbr.2019.111967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/22/2019] [Accepted: 05/20/2019] [Indexed: 01/19/2023]
Abstract
The goal of this article is to clarify the role of various growth factors in the establishment and progression of obsessive-compulsive disorder (OCD). OCD is a chronic mental disorder with recurrent intrusive thoughts and/or repetitive compulsive behaviors that increase during stressful periods. Growth and neurotrophic factors may be contributing factors in the pathophysiology of OCD. Many of them are synthesized and released within the central nervous system and act as trophic agents in neurons; some of them are involved in brain growth, development, neurogenesis, myelination and plasticity, while others take part in the protection of the nervous system following brain injuries. This paper attempts to identify all articles investigating the relationship between OCD and neurotrophic and growth factors, in both animal and human studies, with a focus on adult brain studies. Based on the PubMed and Scopus and Science Direct search tools, the available articles and studies are reviewed. Out of 230 records in total, the ones related to our review topic were taken into account to further understand the pathophysiological mechanism(s) of OCD, providing methods to improve its symptoms via the modification of neurotrophins and growth factor imbalances.
Collapse
Affiliation(s)
- Sara Savaheli
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Cacialli P, Gatta C, D'Angelo L, Leggieri A, Palladino A, de Girolamo P, Pellegrini E, Lucini C. Nerve growth factor is expressed and stored in central neurons of adult zebrafish. J Anat 2019; 235:167-179. [PMID: 30945286 PMCID: PMC6580073 DOI: 10.1111/joa.12986] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2019] [Indexed: 12/16/2022] Open
Abstract
Nerve growth factor (NGF), a member of the neurotrophin family, was initially described as neuronal survival and growth factor, but successively has emerged as an active mediator in many essential functions in the central nervous system of mammals. NGF is synthesized as a precursor pro-NGF and is cleaved intracellularly into mature NGF. However, recent evidence demonstrates that pro-NGF is not a simple inactive precursor, but is also secreted outside the cells and can exert multiple roles. Despite the vast literature present in mammals, studies devoted to NGF in the brain of other vertebrate models are scarce. Zebrafish is a teleost fish widely known for developmental genetic studies and is well established as model for translational neuroscience research. Genomic organization of zebrafish and mouse NGF is highly similar, and zebrafish NGF protein has been reported in mature and two-precursors forms. To add further knowledge on neurotrophic factors in vertebrate brain models, we decided to determine the NGF mRNA and protein distribution in the adult zebrafish brain and to characterize the phenotype of NGF-positive cells. NGF mRNA was visualized by in situ hybridization on whole-mount brains. NGF protein distribution was assessed on microtomic sections by using an antiserum against NGF, able to recognize pro-NGF in adult zebrafish brain as demonstrated also in previous studies. To characterize NGF-positive cells, anti-NGF was employed on microtomic slides of aromatase B transgenic zebrafish (where radial glial cells appeared fluorescent) and by means of double-immunolabeling against NGF/proliferative cell nuclear antigen (PCNA; proliferation marker) and NGF/microtube-associated protein2 (MAP2; neuronal marker). NGF mRNA and protein were widely distributed in the brain of adult zebrafish, and their pattern of distribution of positive perikaryal was overlapping, both in males and females, with few slight differences. Specifically, the immunoreactivity to the protein was observed in fibers over the entire encephalon. MAP2 immunoreactivity was present in the majority of NGF-positive cells, throughout the zebrafish brain. PCNA and aromatase B cells were not positive to NGF, but they were closely intermingled with NGF cells. In conclusion, our study demonstrated that mature neurons in the zebrafish brain express NGF mRNA and store pro-NGF.
Collapse
Affiliation(s)
- Pietro Cacialli
- Dipartimento di Medicina Veterinaria e Produzioni AnimaliUniversità di Napoli Federico IINapoliItaly
- InsermEHESP, Irset (Institut de recherche en santé environnement et travail) ‐ UMR_S 1085Univ RennesRennesFrance
| | - Claudia Gatta
- Dipartimento di Medicina Veterinaria e Produzioni AnimaliUniversità di Napoli Federico IINapoliItaly
| | - Livia D'Angelo
- Dipartimento di Medicina Veterinaria e Produzioni AnimaliUniversità di Napoli Federico IINapoliItaly
- Stazione Zoologica Anton DohrnNapoliItaly
| | - Adele Leggieri
- Dipartimento di Medicina Veterinaria e Produzioni AnimaliUniversità di Napoli Federico IINapoliItaly
| | - Antonio Palladino
- Centro Ricerche Interdipartimentali sui BiomaterialiUniversità di Napoli Federico IINapoliItaly
| | - Paolo de Girolamo
- Dipartimento di Medicina Veterinaria e Produzioni AnimaliUniversità di Napoli Federico IINapoliItaly
| | - Elisabeth Pellegrini
- InsermEHESP, Irset (Institut de recherche en santé environnement et travail) ‐ UMR_S 1085Univ RennesRennesFrance
| | - Carla Lucini
- Dipartimento di Medicina Veterinaria e Produzioni AnimaliUniversità di Napoli Federico IINapoliItaly
| |
Collapse
|
30
|
Biane JS, Takashima Y, Scanziani M, Conner JM, Tuszynski MH. Reorganization of Recurrent Layer 5 Corticospinal Networks Following Adult Motor Training. J Neurosci 2019; 39:4684-4693. [PMID: 30948479 PMCID: PMC6561695 DOI: 10.1523/jneurosci.3442-17.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 01/27/2023] Open
Abstract
Recurrent synaptic connections between neighboring neurons are a key feature of mammalian cortex, accounting for the vast majority of cortical inputs. Although computational models indicate that reorganization of recurrent connectivity is a primary driver of experience-dependent cortical tuning, the true biological features of recurrent network plasticity are not well identified. Indeed, whether rewiring of connections between cortical neurons occurs during behavioral training, as is widely predicted, remains unknown. Here, we probe M1 recurrent circuits following motor training in adult male rats and find robust synaptic reorganization among functionally related layer 5 neurons, resulting in a 2.5-fold increase in recurrent connection probability. This reorganization is specific to the neuronal subpopulation most relevant for executing the trained motor skill, and behavioral performance was impaired following targeted molecular inhibition of this subpopulation. In contrast, recurrent connectivity is unaffected among neighboring layer 5 neurons largely unrelated to the trained behavior. Training-related corticospinal cells also express increased excitability following training. These findings establish the presence of selective modifications in recurrent cortical networks in adulthood following training.SIGNIFICANCE STATEMENT Recurrent synaptic connections between neighboring neurons are characteristic of cortical architecture, and modifications to these circuits are thought to underlie in part learning in the adult brain. We now show that there are robust changes in recurrent connections in the rat motor cortex upon training on a novel motor task. Motor training results in a 2.5-fold increase in recurrent connectivity, but only within the neuronal subpopulation most relevant for executing the new motor behavior; recurrent connectivity is unaffected among adjoining neurons that do not execute the trained behavior. These findings demonstrate selective reorganization of recurrent synaptic connections in the adult neocortex following novel motor experience, and illuminate fundamental properties of cortical function and plasticity.
Collapse
Affiliation(s)
| | | | - Massimo Scanziani
- Neurobiology, University of California, San Diego, California 92093
- Howard Hughes Medical Institute, San Diego, California, 92093, and
| | | | - Mark H Tuszynski
- Departments of Neurosciences,
- Veterans Administration Medical Center, San Diego, California 92161
| |
Collapse
|
31
|
Boskovic Z, Meier S, Wang Y, Milne M, Onraet T, Tedoldi A, Coulson E. Regulation of cholinergic basal forebrain development, connectivity, and function by neurotrophin receptors. Neuronal Signal 2019; 3:NS20180066. [PMID: 32269831 PMCID: PMC7104233 DOI: 10.1042/ns20180066] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
Cholinergic basal forebrain (cBF) neurons are defined by their expression of the p75 neurotrophin receptor (p75NTR) and tropomyosin-related kinase (Trk) neurotrophin receptors in addition to cholinergic markers. It is known that the neurotrophins, particularly nerve growth factor (NGF), mediate cholinergic neuronal development and maintenance. However, the role of neurotrophin signalling in regulating adult cBF function is less clear, although in dementia, trophic signalling is reduced and p75NTR mediates neurodegeneration of cBF neurons. Here we review the current understanding of how cBF neurons are regulated by neurotrophins which activate p75NTR and TrkA, B or C to influence the critical role that these neurons play in normal cortical function, particularly higher order cognition. Specifically, we describe the current evidence that neurotrophins regulate the development of basal forebrain neurons and their role in maintaining and modifying mature basal forebrain synaptic and cortical microcircuit connectivity. Understanding the role neurotrophin signalling plays in regulating the precision of cholinergic connectivity will contribute to the understanding of normal cognitive processes and will likely provide additional ideas for designing improved therapies for the treatment of neurological disease in which cholinergic dysfunction has been demonstrated.
Collapse
Affiliation(s)
- Zoran Boskovic
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Sonja Meier
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Yunpeng Wang
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
- College of Forensic Science, Xi’an Jiaotong University, Shaanxi, China
| | - Michael R. Milne
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Tessa Onraet
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Angelo Tedoldi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth J. Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
32
|
Fodelianaki G, Lansing F, Bhattarai P, Troullinaki M, Zeballos MA, Charalampopoulos I, Gravanis A, Mirtschink P, Chavakis T, Alexaki VI. Nerve Growth Factor modulates LPS - induced microglial glycolysis and inflammatory responses. Exp Cell Res 2019; 377:10-16. [PMID: 30817930 DOI: 10.1016/j.yexcr.2019.02.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 01/09/2023]
Abstract
Microglia, the parenchymal immune cells of the central nervous system, orchestrate neuroinflammation in response to infection or damage, and promote tissue repair. However, aberrant microglial responses are integral to neurodegenerative diseases and critically contribute to disease progression. Thus, it is important to elucidate how microglia - mediated neuroinflammation is regulated by endogenous factors. Here, we explored the effect of Nerve Growth Factor (NGF), an abundant neurotrophin, on microglial inflammatory responses. NGF, via its high affinity receptor TrkA, downregulated LPS - induced production of pro-inflammatory cytokines and NO in primary mouse microglia and inhibited TLR4 - mediated activation of the NF-κB and JNK pathways. Furthermore, NGF attenuated the LPS - enhanced glycolytic activity in microglia, as suggested by reduced glucose uptake and decreased expression of the glycolytic enzymes Pfkβ3 and Ldhα. Consistently, 2DG - mediated glycolysis inhibition strongly downregulated LPS - induced cytokine production in microglial cells. Our findings demonstrate that NGF attenuates pro-inflammatory responses in microglia and may thereby contribute to regulation of microglia - mediated neuroinflammation.
Collapse
Affiliation(s)
- Georgia Fodelianaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Felix Lansing
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Prabesh Bhattarai
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Maria Troullinaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Maria Alejandra Zeballos
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | | | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany.
| |
Collapse
|
33
|
Mitra S, Behbahani H, Eriksdotter M. Innovative Therapy for Alzheimer's Disease-With Focus on Biodelivery of NGF. Front Neurosci 2019; 13:38. [PMID: 30804738 PMCID: PMC6370742 DOI: 10.3389/fnins.2019.00038] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/15/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder associated with abnormal protein modification, inflammation and memory impairment. Aggregated amyloid beta (Aβ) and phosphorylated tau proteins are medical diagnostic features. Loss of memory in AD has been associated with central cholinergic dysfunction in basal forebrain, from where the cholinergic circuitry projects to cerebral cortex and hippocampus. Various reports link AD progression with declining activity of cholinergic neurons in basal forebrain. The neurotrophic molecule, nerve growth factor (NGF), plays a major role in the maintenance of cholinergic neurons integrity and function, both during development and adulthood. Numerous studies have also shown that NGF contributes to the survival and regeneration of neurons during aging and in age-related diseases such as AD. Changes in neurotrophic signaling pathways are involved in the aging process and contribute to cholinergic and cognitive decline as observed in AD. Further, gradual dysregulation of neurotrophic factors like NGF and brain derived neurotrophic factor (BDNF) have been reported during AD development thus intensifying further research in targeting these factors as disease modifying therapies against AD. Today, there is no cure available for AD and the effects of the symptomatic treatment like cholinesterase inhibitors (ChEIs) and memantine are transient and moderate. Although many AD treatment studies are being carried out, there has not been any breakthrough and new therapies are thus highly needed. Long-term effective therapy for alleviating cognitive impairment is a major unmet need. Discussion and summarizing the new advancements of using NGF as a potential therapeutic implication in AD are important. In summary, the intent of this review is describing available experimental and clinical data related to AD therapy, priming to gain additional facts associated with the importance of NGF for AD treatment, and encapsulated cell biodelivery (ECB) as an efficient tool for NGF delivery.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Homira Behbahani
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Aging Theme, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Tirassa P, Rosso P, Iannitelli A. Ocular Nerve Growth Factor (NGF) and NGF Eye Drop Application as Paradigms to Investigate NGF Neuroprotective and Reparative Actions. Methods Mol Biol 2018; 1727:19-38. [PMID: 29222770 DOI: 10.1007/978-1-4939-7571-6_2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The eye is a central nervous system structure that is uniquely accessible to local treatment. Through the ocular surface, it is possible to access the retina, optic nerve, and brain. Animal models of retina degeneration or optic nerve crush could thus serve as tools to investigate whether and how factors, which are anterogradely or retrogradely transported through the optic nerve, might contribute to activate neuroprotection and eventually regeneration. Among these factors, nerve growth factor (NGF) plays a crucial role during development of the visual system, as well as during the entire life span, and in pathological conditions. The ability of NGF to exert survival and trophic actions on the retina and brain cells when applied intraocularly and topically as eye drops is critically reviewed here, together with the effects of ocular neurotrophins on neuronal pathways influencing body rhythm, cognitions, and behavioral functions. The latest data from animal models and humans are presented, and the mechanism of action of ocularly administered NGF is discussed. NGF eye drops are proposed as an experimental strategy to investigate the role and cellular targets of neurotrophins in the mechanism(s) underlying neurodegeneration/regeneration and their involvement in the regulation of neurological and behavioral dysfunctions.
Collapse
Affiliation(s)
- Paola Tirassa
- National Research Council (CNR), Institute of Cell Biology & Neurobiology, Rome, Italy.
| | - Pamela Rosso
- National Research Council (CNR), Institute of Cell Biology & Neurobiology, Rome, Italy.,Department of Science, LIME, University Roma Tre, Rome, Italy
| | - Angela Iannitelli
- Department of Human Sciences, University of L'Aquila, L'Aquila, Italy.,Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
35
|
Fasulo L, Brandi R, Arisi I, La Regina F, Berretta N, Capsoni S, D'Onofrio M, Cattaneo A. ProNGF Drives Localized and Cell Selective Parvalbumin Interneuron and Perineuronal Net Depletion in the Dentate Gyrus of Transgenic Mice. Front Mol Neurosci 2017; 10:20. [PMID: 28232789 PMCID: PMC5299926 DOI: 10.3389/fnmol.2017.00020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/16/2017] [Indexed: 01/12/2023] Open
Abstract
ProNGF, the precursor of mature Nerve Growth Factor (NGF), is the most abundant NGF form in the brain and increases markedly in the cortex in Alzheimer's Disease (AD), relative to mature NGF. A large body of evidence shows that the actions of ProNGF and mature NGF are often conflicting, depending on the receptors expressed in target cells. TgproNGF#3 mice, expressing furin-cleavage resistant proNGF in CNS neurons, directly reveal consequences of increased proNGF levels on brain homeostasis. Their phenotype clearly indicates that proNGF can be a driver of neurodegeneration, including severe learning and memory behavioral deficits, cholinergic deficits, and diffuse immunoreactivity for A-beta and A-beta-oligomers. In aged TgproNGF#3 mice spontaneous epileptic-like events are detected in entorhinal cortex-hippocampal slices, suggesting occurrence of excitatory/inhibitory (E/I) imbalance. In this paper, we investigate the molecular events linking increased proNGF levels to the epileptiform activity detected in hippocampal slices. The occurrence of spontaneous epileptiform discharges in the hippocampal network in TgproNGF#3 mice suggests an impaired inhibitory interneuron homeostasis. In the present study, we detect the onset of hippocampal epileptiform events at 1-month of age. Later, we observe a regional- and cellular-selective Parvalbumin interneuron and perineuronal net (PNN) depletion in the dentate gyrus (DG), but not in other hippocampal regions of TgproNGF#3 mice. These results demonstrate that, in the hippocampus, the DG is selectively vulnerable to altered proNGF/NGF signaling. Parvalbumin interneuron depletion is also observed in the amygdala, a region strongly connected to the hippocampus and likewise receiving cholinergic afferences. Transcriptome analysis of TgproNGF#3 hippocampus reveals a proNGF signature with broad down-regulation of transcription. The most affected mRNAs modulated at early times belong to synaptic transmission and plasticity and extracellular matrix (ECM) gene families. Moreover, alterations in the expression of selected BDNF splice variants were observed. Our results provide further mechanistic insights into the vicious negative cycle linking proNGF and neurodegeneration, confirming the regulation of E/I homeostasis as a crucial mediating mechanism.
Collapse
Affiliation(s)
- Luisa Fasulo
- Bio@SNS Laboratory, Scuola Normale SuperiorePisa, Italy; European Brain Research Institute Rita Levi-MontalciniRome, Italy
| | - Rossella Brandi
- European Brain Research Institute Rita Levi-Montalcini Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute Rita Levi-Montalcini Rome, Italy
| | | | - Nicola Berretta
- Department of Experimental Neurology, Fondazione Santa Lucia IRCCS Rome, Italy
| | - Simona Capsoni
- Bio@SNS Laboratory, Scuola Normale Superiore Pisa, Italy
| | - Mara D'Onofrio
- European Brain Research Institute Rita Levi-Montalcini Rome, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory, Scuola Normale SuperiorePisa, Italy; European Brain Research Institute Rita Levi-MontalciniRome, Italy
| |
Collapse
|
36
|
Leukemia inhibitory factor impairs structural and neurochemical development of rat visual cortex in vivo. Mol Cell Neurosci 2017; 79:81-92. [PMID: 28088609 DOI: 10.1016/j.mcn.2016.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/25/2016] [Accepted: 12/29/2016] [Indexed: 11/23/2022] Open
Abstract
Minipump infusions into visual cortex in vivo at the onset of the critical period have revealed that the proinflammatory cytokine leukemia inhibitory factor (LIF) delays the maturation of thalamocortical projection neurons of the lateral geniculate nucleus, and tecto-thalamic projection neurons of the superior colliculus, and cortical layer IV spiny stellates and layer VI pyramidal neurons. Here, we report that P12-20 LIF infusion inhibits somatic maturation of pyramidal neurons and of all interneuron types in vivo. Likewise, DIV 12-20 LIF treatment in organotypic cultures prevents somatic growth GABA-ergic neurons. Further, while NPY expression is increased in the LIF-infused hemispheres, the expression of parvalbumin mRNA and protein, Kv3.1 mRNA, calbindin D-28k protein, and GAD-65 mRNA, but not of GAD-67 mRNA or calretinin protein is substantially reduced. Also, LIF treatment decreases parvalbumin, Kv3.1, Kv3.2 and GAD-65, but not GAD-67 mRNA expression in OTC. Developing cortical neurons are known to depend on neurotrophins. Indeed, LIF alters neurotrophin mRNA expression, and prevents the growth promoting action of neurotophin-4 in GABA-ergic neurons. The results imply that LIF, by altering neurotrophin expression and/or signaling, could counteract neurotrophin-dependent growth and neurochemical differentiation of cortical neurons.
Collapse
|
37
|
Sampaio TB, Savall AS, Gutierrez MEZ, Pinton S. Neurotrophic factors in Alzheimer's and Parkinson's diseases: implications for pathogenesis and therapy. Neural Regen Res 2017; 12:549-557. [PMID: 28553325 PMCID: PMC5436343 DOI: 10.4103/1673-5374.205084] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neurotrophic factors comprise essential secreted proteins that have several functions in neural and non-neural tissues, mediating the development, survival and maintenance of peripheral and central nervous system. Therefore, neurotrophic factor issue has been extensively investigated into the context of neurodegenerative diseases. Alzheimer's disease and Parkinson's disease show changes in the regulation of specific neurotrophic factors and their receptors, which appear to be critical for neuronal degeneration. Indeed, neurotrophic factors prevent cell death in degenerative processes and can enhance the growth and function of affected neurons in these disorders. Based on recent reports, this review discusses the main findings related to the neurotrophic factor support – mainly brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor – in the survival, proliferation and maturation of affected neurons in Alzheimer's disease and Parkinson's disease as well as their putative application as new therapeutic approach for these diseases management.
Collapse
Affiliation(s)
- Tuane Bazanella Sampaio
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.,Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| | - Anne Suely Savall
- Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| | | | - Simone Pinton
- Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| |
Collapse
|
38
|
Allaway KC, Machold R. Developmental specification of forebrain cholinergic neurons. Dev Biol 2016; 421:1-7. [PMID: 27847324 DOI: 10.1016/j.ydbio.2016.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 01/17/2023]
Abstract
Striatal cholinergic interneurons and basal forebrain cholinergic projection neurons, which together comprise the forebrain cholinergic system, regulate attention, memory, reward pathways, and motor activity through the neuromodulation of multiple brain circuits. The importance of these neurons in the etiology of neurocognitive disorders has been well documented, but our understanding of their specification during embryogenesis is still incomplete. All forebrain cholinergic projection neurons and interneurons appear to share a common developmental origin in the embryonic ventral telencephalon, a region that also gives rise to GABAergic projection neurons and interneurons. Significant progress has been made in identifying the key intrinsic and extrinsic factors that promote a cholinergic fate in this precursor population. However, how cholinergic interneurons and projection neurons differentiate from one another during development, as well as how distinct developmental programs contribute to heterogeneity within those two classes, is not yet well understood. In this review we summarize the transcription factors and signaling molecules known to play a role in the specification and early development of striatal and basal forebrain cholinergic neurons. We also discuss the heterogeneity of these populations and its possible developmental origins.
Collapse
Affiliation(s)
- Kathryn C Allaway
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | - Robert Machold
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
39
|
Lauzon MA, Daviau A, Marcos B, Faucheux N. Growth factor treatment to overcome Alzheimer's dysfunctional signaling. Cell Signal 2015; 27:1025-38. [PMID: 25744541 DOI: 10.1016/j.cellsig.2015.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) will increase as the world population ages, creating a huge socio-economic burden. The three pathophysiological hallmarks of AD are the cholinergic system dysfunction, the β-amyloid peptide deposition and the Tau protein hyperphosphorylation. Current treatments have only transient effects and each tends to concentrate on a single pathophysiological aspect of AD. This review first provides an overall view of AD in terms of its pathophysiological symptoms and signaling dysfunction. We then examine the therapeutic potential of growth factors (GFs) by showing how they can overcome the dysfunctional cell signaling that occurs in AD. Finally, we discuss new alternatives to GFs that help overcome the problem of brain uptake, such as small peptides, with evidence from some of our unpublished data on human neuronal cell line.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Alex Daviau
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Bernard Marcos
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Nathalie Faucheux
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada.
| |
Collapse
|