1
|
Rahman AFMT, Bulbule S, Belayet JB, Benko A, Gottschalk CG, Frick DN, Arnold LA, Hossain MM, Roy A. JRM-28, a Novel HDAC2 Inhibitor, Upregulates Plasticity-Associated Proteins in Hippocampal Neurons and Enhances Morphological Plasticity via Activation of CREB: Implications for Alzheimer's Disease. Cells 2024; 13:1964. [PMID: 39682714 PMCID: PMC11640089 DOI: 10.3390/cells13231964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Enhancement of neuronal plasticity by small-molecule therapeutics protects cognitive skills and also ameliorates progressive neurodegenerative pathologies like Alzheimer's disease (AD) and dementia. One such compound, a novel histone deacetylase 2 (HDAC2) inhibitor named JRM-28, was shown here to enhance dendritic strength, augment spine density, and upregulate post-synaptic neurotransmission in hippocampal neurons. The molecular basis for this effect correlates with JRM-28-induced upregulation of the transcription of cAMP response element-binding protein(CREB), induction of its transcriptional activity, and subsequent stimulation of expressions of CREB-dependent plasticity-associated genes, such as those encoding N-methyl-D-aspartate (NMDA) receptor subunit NR2A and the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluR1. Specifically, JRM-28 stimulated the NMDA- and AMPA-receptor-sensitive ionotropic calcium influx in hippocampal neurons. Interestingly, JRM-28 did not induce NMDA- and AMPA-sensitive calcium influx in hippocampal neurons once the expression of CREB was knocked down by creb siRNA, suggesting the critical role of CREB in JRM-28-mediated upregulation of synaptic plasticity. Finally, JRM-28 upregulated CREB mRNA, CREB-dependent plasticity-associated markers, and ionotropic calcium influx in iPSC-derived AD human neurons, indicating its therapeutic implications in the amelioration of AD pathologies.
Collapse
Affiliation(s)
- A. F. M. Towheedur Rahman
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
| | - Sarojini Bulbule
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| | - Jawad Bin Belayet
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
| | - Anna Benko
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - Carl Gunnar Gottschalk
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| | - David N. Frick
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - M. Mahmun Hossain
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - Avik Roy
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
- Simmaron Research and Development Laboratory, University of Wisconsin-Milwaukee, Chemistry Building, 2000 E Kenwood Blvd, Suite # 320, Milwaukee, WI 53211, USA
| |
Collapse
|
2
|
McKay M, Gorai S, Paidi RK, Mondal S, Pahan K. Identification of Cinnamein, a Component of Balsam of Tolu/Peru, as a New Ligand of PPARα for Plaque Reduction and Memory Protection in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:903-922. [PMID: 38910936 PMCID: PMC11191634 DOI: 10.3233/adr-230179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/15/2024] [Indexed: 06/25/2024] Open
Abstract
Background Despite intense investigations, no effective treatment is yet available to reduce plaques and protect memory and learning in patients with Alzheimer's disease (AD), the most common neurodegenerative disorder. Therefore, it is important to identify a non-toxic, but effective, treatment option for AD. Objective Cinnamein, a nontoxic compound, is naturally available in Balsam of Peru and Tolu Balsam. We examined whether cinnamein treatment could decrease plaques and improve cognitive functions in 5XFAD mouse model of AD. Methods We employed in silico analysis, time-resolved fluorescence energy transfer assay, thermal shift assay, primary neuron isolation, western blot, immunostaining, immunohistochemistry, Barnes maze, T maze, and open field behavior. Results Oral administration of cinnamein led to significant reduction in amyloid-β plaque deposits in the brain and protection of spatial learning and memory in 5XFAD mice. Peroxisome proliferator-activated receptor alpha (PPARα), a nuclear hormone receptor, is involved in plaque lowering and increase in hippocampal plasticity. While investigating underlying mechanisms, we found that cinnamein served as a ligand of PPARα. Accordingly, oral cinnamein upregulated the level of PPARα, but not PPARβ, in the hippocampus, and remained unable to decrease plaques from the hippocampus and improve memory and learning in 5XFAD mice lacking PPARα. While A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) is one of the drivers of nonamyloidogenic pathway, transcription factor EB (TFEB) is considered as the master regulator of autophagy. Cinnamein treatment was found to upregulate both ADAM10 and TFEB in the brain of 5XFAD mice via PPARα. Conclusions Our results suggest that this balsam component may have therapeutic importance in AD.
Collapse
Affiliation(s)
- Mary McKay
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Ramesh K. Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Susanta Mondal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, USA
| |
Collapse
|
3
|
Baena-Caldas GP, Li J, Pedraza L, Ghosh S, Kalmes A, Barone FC, Moreno H, Hernández AI. Neuroprotective effect of the RNS60 in a mouse model of transient focal cerebral ischemia. PLoS One 2024; 19:e0295504. [PMID: 38166102 PMCID: PMC10760892 DOI: 10.1371/journal.pone.0295504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 11/22/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Stroke is a major cause of death, disability, and public health problems. Its intervention is limited to early treatment with thrombolytics and/or endovascular clot removal with mechanical thrombectomy without any available subacute or chronic neuroprotective treatments. RNS60 has reduced neuroinflammation and increased neuronal survival in several animal models of neurodegeneration and trauma. The aim here was to evaluate whether RNS60 protects the brain and cognitive function in a mouse stroke model. METHODS Male C57BL/6J mice were subjected to sham or ischemic stroke surgery using 60-minute transient middle cerebral artery occlusion (tMCAo). In each group, mice received blinded daily administrations of RNS60 or control fluids (PNS60 or normal saline [NS]), beginning 2 hours after surgery over 13 days. Multiple neurobehavioral tests were conducted (Neurological Severity Score [mNSS], Novel Object Recognition [NOR], Active Place Avoidance [APA], and the Conflict Variant of APA [APAc]). On day 14, cortical microvascular perfusion (MVP) was measured, then brains were removed and infarct volume, immunofluorescence of amyloid beta (Aβ), neuronal density, microglial activation, and white matter damage/myelination were measured. SPSS was used for analysis (e.g., ANOVA for parametric data; Kruskal Wallis for non-parametric data; with post-hoc analysis). RESULTS Thirteen days of treatment with RNS60 reduced brain infarction, amyloid pathology, neuronal death, microglial activation, white matter damage, and increased MVP. RNS60 reduced brain pathology and resulted in behavioral improvements in stroke compared to sham surgery mice (increased memory-learning in NOR and APA, improved cognitive flexibility in APAc). CONCLUSION RNS60-treated mice exhibit significant protection of brain tissue and improved neurobehavioral functioning after tMCAo-stroke. Additional work is required to determine mechanisms, time-window of dosing, and multiple dosing volumes durations to support clinical stroke research.
Collapse
Affiliation(s)
- Gloria Patricia Baena-Caldas
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- Health Sciences Division, Department of Morphology, School of Biomedical Sciences, Universidad del Valle, Cali, Colombia
| | - Jie Li
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Lina Pedraza
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Supurna Ghosh
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Andreas Kalmes
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Frank C. Barone
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| | - Herman Moreno
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| | - A. Iván Hernández
- Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| |
Collapse
|
4
|
Paidi RK, Raha S, Roy A, Pahan K. Muscle-building supplement β-hydroxy β-methylbutyrate binds to PPARα to improve hippocampal functions in mice. Cell Rep 2023; 42:112717. [PMID: 37437568 PMCID: PMC10440158 DOI: 10.1016/j.celrep.2023.112717] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 04/09/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023] Open
Abstract
This study underlines the importance of β-hydroxy β-methylbutyrate (HMB), a muscle-building supplement in human, in increasing mouse hippocampal plasticity. Detailed proteomic analyses reveal that HMB serves as a ligand of peroxisome proliferator-activated receptor α (PPARα), a nuclear hormone receptor involved in fat metabolism, via interaction with the Y314 residue. Accordingly, HMB is ineffective in increasing plasticity of PPARα-/- hippocampal neurons. While lentiviral establishment of full-length PPARα restores the plasticity-promoting effect of HMB in PPARα-/- hippocampal neurons, lentiviral transduction of Y314D-PPARα remains unable to do that, highlighting the importance of HMB's interaction with the Y314 residue. Additionally, oral HMB improves spatial learning and memory and reduces plaque load in 5X familial Alzheimer's disease (5XFAD) mice, but not in 5XFADΔPPARα mice (5XFAD lacking PPARα), indicating the involvement of PPARα in HMB-mediated neuroprotection in 5XFAD mice. These results delineate neuroprotective functions of HMB and suggest that this widely used supplement may be repurposed for AD.
Collapse
Affiliation(s)
- Ramesh K Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Avik Roy
- Simmaron Research Institute, Technology Innovation Center, 10437 W Innovation Drive, Wauwatosa, WI, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA.
| |
Collapse
|
5
|
Jana M, Dasarathy S, Ghosh S, Pahan K. Upregulation of DJ-1 in Dopaminergic Neurons by a Physically-Modified Saline: Implications for Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24054652. [PMID: 36902085 PMCID: PMC10002578 DOI: 10.3390/ijms24054652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder in human and loss-of-functions DJ-1 mutations are associated with a familial form of early onset PD. Functionally, DJ-1 (PARK7), a neuroprotective protein, is known to support mitochondria and protect cells from oxidative stress. Mechanisms and agents by which the level of DJ-1 could be increased in the CNS are poorly described. RNS60 is a bioactive aqueous solution created by exposing normal saline to Taylor-Couette-Poiseuille flow under high oxygen pressure. Recently we have described neuroprotective, immunomodulatory and promyelinogenic properties of RNS60. Here we delineate that RNS60 is also capable of increasing the level of DJ-1 in mouse MN9D neuronal cells and primary dopaminergic neurons, highlighting another new neuroprotective effect of RNS60. While investigating the mechanism we found the presence of cAMP response element (CRE) in DJ-1 gene promoter and stimulation of CREB activation in neuronal cells by RNS60. Accordingly, RNS60 treatment increased the recruitment of CREB to the DJ-1 gene promoter in neuronal cells. Interestingly, RNS60 treatment also induced the enrollment of CREB-binding protein (CBP), but not the other histone acetyl transferase p300, to the promoter of DJ-1 gene. Moreover, knockdown of CREB by siRNA led to the inhibition of RNS60-mediated DJ-1 upregulation, indicating an important role of CREB in DJ-1 upregulation by RNS60. Together, these results indicate that RNS60 upregulates DJ-1 in neuronal cells via CREB-CBP pathway. It may be of benefit for PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | | | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
6
|
Beneficial Effects of RNS60 in Cardiac Ischemic Injury. Curr Issues Mol Biol 2022; 44:4877-4887. [PMID: 36286046 PMCID: PMC9600597 DOI: 10.3390/cimb44100331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
RNS60 is a physically modified saline solution hypothesized to contain oxygen nanobubbles. It has been reported to reduce ischemia/reperfusion injury in a pig model of acute myocardial infarction. We investigated the effects of RNS60 during cardiac hypoxia in mice and as an additive to cardioplegic solution in rat hearts. ApoE−/−LDLr−/− mice were treated by intravenous injection of RNS60 or saline as a control while monitoring the ECG and post-hypoxic serum release of troponin T and creatine kinase activity. Hearts infused with Custodiol containing 10% RNS60 or saline as the control were subjected to 4 h of 4 °C preservation, followed by an assessment of myocardial metabolites, purine release, and mechanical function. RNS60 attenuated changes in the ECG STU area during hypoxia, while the troponin T concentration and creatine kinase activity were significantly higher in the serum of the controls. During reperfusion after 4 h of cold ischemia, the Custodiol/RNS60-treated hearts had about 30% lower LVEDP and better dp/dtmax and dp/dtmin together with a decreased release of purine catabolites vs. the controls. The myocardial ATP, total adenine nucleotides, and phosphocreatine concentrations were higher in the RNS60-treated hearts. This study indicates that RNS60 enhances cardioprotection in experimental myocardial hypoxia and under conditions of cardioplegic arrest. Improved cardiac energetics are involved in the protective effect, but complete elucidation of the mechanism requires further study.
Collapse
|
7
|
Jaramillo-Granada AM, Reyes-Figueroa AD, Ruiz-Suárez JC. Xenon and Krypton Dissolved in Water Form Nanoblobs: No Evidence for Nanobubbles. PHYSICAL REVIEW LETTERS 2022; 129:094501. [PMID: 36083645 DOI: 10.1103/physrevlett.129.094501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/07/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
We demonstrate, experimentally and by molecular dynamics simulations, that krypton and xenon form nanostructured water-gas domains. High pressure was applied to force the inert gases to dissolve in water following Henry's law, then the liquid was depressurized, centrifuged, and inspected by dynamic light scattering. The observed objects have similar sizes and electrical properties to nanobubbles, but we found that they have fairly neutral buoyancy even at high gravitational fields. We posit that the formed nano objects are not bubbles but blobs, unique structures conceived as clathrate-hydrate precursors, thus resolving the so-called Laplace pressure bubble catastrophe.
Collapse
Affiliation(s)
- Angela M Jaramillo-Granada
- Centro de Investigación y de Estudios Avanzados-Monterrey, Parque de Investigación e Innovación Tecnológica, 66600 Nuevo León, Mexico
| | - A D Reyes-Figueroa
- Centro de Investigación en Matemáticas Unidad Monterrey, Av. Alianza Centro No. 502, PIIT, Apodaca, 66628 Nuevo León, Mexico
- Consejo Nacional de Ciencia y Tecnología, Av. Insurgentes Sur 1582, Col. Crédito Constructor, Benito Juárez, 03940 CDMX, Mexico
| | - J C Ruiz-Suárez
- Centro de Investigación y de Estudios Avanzados-Monterrey, Parque de Investigación e Innovación Tecnológica, 66600 Nuevo León, Mexico
| |
Collapse
|
8
|
Afshari R, Akhavan O, Hamblin MR, Varma RS. Review of Oxygenation with Nanobubbles: Possible Treatment for Hypoxic COVID-19 Patients. ACS APPLIED NANO MATERIALS 2021; 4:11386-11412. [PMID: 37556289 PMCID: PMC8565459 DOI: 10.1021/acsanm.1c01907] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/12/2021] [Indexed: 05/05/2023]
Abstract
The coronavirus disease (COVID-19) pandemic, which has spread around the world, caused the death of many affected patients, partly because of the lack of oxygen arising from impaired respiration or blood circulation. Thus, maintaining an appropriate level of oxygen in the patients' blood by devising alternatives to ventilator systems is a top priority goal for clinicians. The present review highlights the ever-increasing application of nanobubbles (NBs), miniature gaseous vesicles, for the oxygenation of hypoxic patients. Oxygen-containing NBs can exert a range of beneficial physiologic and pharmacologic effects that include tissue oxygenation, as well as tissue repair mechanisms, antiinflammatory properties, and antibacterial activity. In this review, we provide a comprehensive survey of the application of oxygen-containing NBs, with a primary focus on the development of intravenous platforms. The multimodal functions of oxygen-carrying NBs, including antimicrobial, antiinflammatory, drug carrying, and the promotion of wound healing are discussed, including the benefits and challenges of using NBs as a treatment for patients with acute hypoxemic respiratory failure, particularly due to COVID-19.
Collapse
Affiliation(s)
- Ronak Afshari
- Department of Physics, Sharif University
of Technology, P.O. Box 11155-9161, Tehran 14588-89694,
Iran
| | - Omid Akhavan
- Department of Physics, Sharif University
of Technology, P.O. Box 11155-9161, Tehran 14588-89694,
Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science,
University of Johannesburg, Doornfontein 2028, South
Africa
| | - Rajender S. Varma
- Regional Center of Advanced Technologies and Materials,
Czech Advanced Technology and Research Institute, Palacky
University, Šlechtitelů 27, Olomouc 78371, Czech
Republic
| |
Collapse
|
9
|
Roy A, Kundu M, Chakrabarti S, Patel DR, Pahan K. Oleamide, a Sleep-Inducing Supplement, Upregulates Doublecortin in Hippocampal Progenitor Cells via PPARα. J Alzheimers Dis 2021; 84:1747-1762. [PMID: 34744082 PMCID: PMC10075226 DOI: 10.3233/jad-215124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Doublecortin (DCX), a microtubule associated protein, has emerged as a central biomarker of hippocampal neurogenesis. However, molecular mechanisms by which DCX is regulated are poorly understood. OBJECTIVE Since sleep is involved with the acquisition of memory and oleamide or 9-Octadecenamide (OCT) is a sleep-inducing supplement in human, we examined whether OCT could upregulate DCX in hippocampal progenitor cells (HPCs). METHODS We employed real-time PCR, western blot, immunostaining, chromatin immunoprecipitation, lentiviral transduction in HPCs, and the calcium influx assay. RESULTS OCT directly upregulated the transcription of Dcx in HPCs via activation of peroxisome proliferator-activated receptor α (PPARα), a lipid-lowering transcription factor. We observed that, HPCs of Ppara-null mice displayed significant impairment in DCX expression and neuronal differentiation as compared to that of wild-type mice. Interestingly, treatment with OCT stimulated the differentiation process of HPCs in wild-type, but not Ppara-null mice. Reconstruction of PPARα in mouse Ppara-null HPCs restored the expression of DCX, which was further stimulated with OCT treatment. In contrast, a dominant-negative mutant of PPARα significantly attenuated the stimulatory effect of OCT on DCX expression and suppressed neuronal differentiation of human neural progenitor cells. Furthermore, RNA microarray, STRING, chromatin immunoprecipitation, site-directed mutagenesis, and promoter reporter assay have identified DCX as a new target of PPARα. CONCLUSION These results indicate that OCT, a sleep supplement, directly controls the expression of DCX and suggest that OCT may be repurposed for stimulating the hippocampal neurogenesis.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sudipta Chakrabarti
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Dhruv R Patel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
10
|
Li M, Ma X, Eisener J, Pfeiffer P, Ohl CD, Sun C. How bulk nanobubbles are stable over a wide range of temperatures. J Colloid Interface Sci 2021; 596:184-198. [DOI: 10.1016/j.jcis.2021.03.064] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/04/2021] [Accepted: 03/11/2021] [Indexed: 11/28/2022]
|
11
|
Patel D, Roy A, Raha S, Kundu M, Gonzalez FJ, Pahan K. Upregulation of BDNF and hippocampal functions by a hippocampal ligand of PPARα. JCI Insight 2020; 5:136654. [PMID: 32315292 PMCID: PMC7259538 DOI: 10.1172/jci.insight.136654] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/15/2020] [Indexed: 01/21/2023] Open
Abstract
Discovery strategies commonly focus on the identification of chemical libraries or natural products, but the modulation of endogenous ligands offers a much better therapeutic strategy due to their low adverse potential. Recently, we found that hexadecanamide (Hex) is present in hippocampal nuclei of normal mice as an endogenous ligand of PPARα. This study underlines the importance of Hex in inducing the expression of brain-derived neurotrophic factor (BDNF) from hippocampal neurons via PPARα. The level of Hex was lower in the hippocampi of 5XFAD mice as compared with that in non-Tg mice. Oral administration of Hex increased the level of this molecule in the hippocampus to stimulate BDNF and its downstream plasticity-associated molecules, promote synaptic functions in the hippocampus, and improve memory and learning in 5XFAD mice. However, oral Hex remained unable to stimulate hippocampal plasticity and improve cognitive behaviors in 5XFADPparα-null and 5XFADPparα-ΔHippo mice, indicating an essential role of hippocampal PPARα in Hex-mediated improvement in hippocampal functions. This is the first demonstration to our knowledge of protection of hippocampal functions by oral administration of a hippocampus-based drug, suggesting that Hex may be explored for therapeutic intervention in AD.
Collapse
Affiliation(s)
- Dhruv Patel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
12
|
Tan BH, An H, Ohl CD. How Bulk Nanobubbles Might Survive. PHYSICAL REVIEW LETTERS 2020; 124:134503. [PMID: 32302159 DOI: 10.1103/physrevlett.124.134503] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 05/23/2023]
Abstract
The existence of bulk nanobubbles has long been regarded with scepticism, due to the limitations of experimental techniques and the widespread assumption that spherical bubbles cannot achieve stable equilibrium. We develop a model for the stability of bulk nanobubbles based on the experimental observation that the zeta potential of spherical bubbles abruptly diverges from the planar value below 10 μm. Our calculations recover three persistently reported-but disputed-properties of bulk nanobubbles: that they stabilize at a typical radius of ∼100 nm, that this radius is bounded below 1 μm, and that it increases with ionic concentration.
Collapse
Affiliation(s)
- Beng Hau Tan
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
- Low Energy Electronic Systems, Singapore-MIT Alliance for Research and Technology, 1 Create Way, 138602 Singapore
| | - Hongjie An
- Queensland Micro and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, Queensland 4111, Australia
| | - Claus-Dieter Ohl
- Otto von Guericke University Magdeburg, Institute of Experimental Physics, Universitätsplatz 2, 39016 Magdeburg, Germany
| |
Collapse
|
13
|
Ma Q, Geng Y, Wang HL, Han B, Wang YY, Li XL, Wang L, Wang MW. High Frequency Repetitive Transcranial Magnetic Stimulation Alleviates Cognitive Impairment and Modulates Hippocampal Synaptic Structural Plasticity in Aged Mice. Front Aging Neurosci 2019; 11:235. [PMID: 31619982 PMCID: PMC6759649 DOI: 10.3389/fnagi.2019.00235] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 08/14/2019] [Indexed: 01/11/2023] Open
Abstract
Normal aging is accompanied by hippocampus-dependent cognitive impairment, which is a risk factor of Alzheimer’s disease. This study aims to investigate the effect of high frequency-repetitive transcranial magnetic stimulation (HF-rTMS) on hippocampus-dependent learning and memory in aged mice and explore its underlying mechanisms. Forty-five male Kunming mice (15 months old) were randomly divided into three groups: aged sham, 5 Hz rTMS, and 25 Hz rTMS. Two sessions of 5 Hz or 25 Hz rTMS comprising 1,000 pulses in 10 trains were delivered once a day for 14 consecutive days. The aged sham group was treated by the reverse side of the coil. In the adult sham group, 15 male Kunming mice (3 months old) were treated the same way as the aged sham group. A Morris water maze (MWM) was conducted following the stimulation, and synaptic ultrastructure was observed through a transmission electron microscope. HF-rTMS improved spatial learning and memory impairment in the aged mice, and 5 Hz was more significant than 25 Hz. Synaptic plasticity-associated gene profiles were modified by HF-rTMS, especially neurotrophin signaling pathways and cyclic adenosine monophosphate response element binding protein (CREB) cofactors. Compared to the aged sham group, synaptic plasticity-associated proteins, i.e., synaptophysin (SYN) and postsynaptic density (PSD)-95 were increased; brain-derived neurotrophic factor (BDNF) and phosphorylated CREB (pCREB) significantly increased after the 5 Hz HF-rTMS treatment. Collectively, our results suggest that HF-rTMS ameliorated cognitive deficits in naturally aged mice. The 5 Hz rTMS treatment significantly enhanced synaptic structural plasticity and activated the BDNF/CREB pathway in the hippocampus.
Collapse
Affiliation(s)
- Qinying Ma
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, China
| | - Yuan Geng
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, China
| | - Hua-Long Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, China
| | - Bing Han
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, China
| | - Yan-Yong Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, China
| | - Xiao-Li Li
- Department of Neurology, the First Hospital of Shijiazhuang, Shijiazhuang, China
| | - Lin Wang
- Emergency Department, CNPC Central Hospital, Langfang, China
| | - Ming-Wei Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei, Shijiazhuang, China
| |
Collapse
|
14
|
Del Olmo N, Ruiz-Gayo M. Influence of High-Fat Diets Consumed During the Juvenile Period on Hippocampal Morphology and Function. Front Cell Neurosci 2018; 12:439. [PMID: 30515083 PMCID: PMC6255817 DOI: 10.3389/fncel.2018.00439] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
The negative impact of obesity on neurocognitive functioning is an issue of increasing clinical interest. Over the last decade, a number of studies have analyzed the influence of high-fat diets (HFDs) on cognitive performance, particularly in adolescent individuals. Different approaches, including behavioral, neurochemical, electrophysiological and morphological studies, have been developed to address the effect of HFDs on neural processes interfering with learning and memory skills in rodents. Many of the studies have focused on learning and memory related to the hippocampus and the mechanisms underlying these processes. The goal of the current review article is to highlight the relationship between hippocampal learning/memory deficits and nutritional/endocrine inputs derived from HFDs consumption, with a special emphasis on research showing the effect of HFDs intake during the juvenile period. We have also reviewed recent research regarding the effect of HFDs on hippocampal neurotransmission. An overview of research suggesting the involvement of fatty acid (FA) receptor-mediated signaling pathways in memory deficits triggered by HFDs is also provided. Finally, the role of leptin and HFD-evoked hyperleptinemia is discussed.
Collapse
Affiliation(s)
- Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
15
|
Patel D, Roy A, Kundu M, Jana M, Luan CH, Gonzalez FJ, Pahan K. Aspirin binds to PPARα to stimulate hippocampal plasticity and protect memory. Proc Natl Acad Sci U S A 2018; 115:E7408-E7417. [PMID: 30012602 PMCID: PMC6077698 DOI: 10.1073/pnas.1802021115] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite its long history, until now, no receptor has been identified for aspirin, one of the most widely used medicines worldwide. Here we report that peroxisome proliferator-activated receptor alpha (PPARα), a nuclear hormone receptor involved in fatty acid metabolism, serves as a receptor of aspirin. Detailed proteomic analyses including cheminformatics, thermal shift assays, and TR-FRET revealed that aspirin, but not other structural homologs, acts as a PPARα ligand through direct binding at the Tyr314 residue of the PPARα ligand-binding domain. On binding to PPARα, aspirin stimulated hippocampal plasticity via transcriptional activation of cAMP response element-binding protein (CREB). Finally, hippocampus-dependent behavioral analyses, calcium influx assays in hippocampal slices and quantification of dendritic spines demonstrated that low-dose aspirin treatment improved hippocampal plasticity and memory in FAD5X mice, but not in FAD5X/Ppara-null mice. These findings highlight a property of aspirin: stimulating hippocampal plasticity via direct interaction with PPARα.
Collapse
Affiliation(s)
- Dhruv Patel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612
| | - Madhuchhanda Kundu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612
| | - Chi-Hao Luan
- High-Throughput Analysis Laboratory and Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612;
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612
| |
Collapse
|
16
|
Vallarola A, Sironi F, Tortarolo M, Gatto N, De Gioia R, Pasetto L, De Paola M, Mariani A, Ghosh S, Watson R, Kalmes A, Bonetto V, Bendotti C. RNS60 exerts therapeutic effects in the SOD1 ALS mouse model through protective glia and peripheral nerve rescue. J Neuroinflammation 2018; 15:65. [PMID: 29495962 PMCID: PMC5833072 DOI: 10.1186/s12974-018-1101-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/21/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects the motor neuromuscular system leading to complete paralysis and premature death. The multifactorial nature of ALS that involves both cell-autonomous and non-cell-autonomous processes contributes to the lack of effective therapies, usually targeted to a single pathogenic mechanism. RNS60, an experimental drug containing oxygenated nanobubbles generated by modified Taylor-Couette-Poiseuille flow with elevated oxygen pressure, has shown anti-inflammatory and neuroprotective properties in different experimental paradigms. Since RNS60 interferes with multiple cellular mechanisms known to be involved in ALS pathology, we evaluated its effect in in vitro and in vivo models of ALS. METHODS Co-cultures of primary microglia/spinal neurons exposed to LPS and astrocytes/spinal neurons from SOD1G93A mice were used to examine the effect of RNS60 or normal saline (NS) on the selective motor neuron degeneration. Transgenic SOD1G93A mice were treated with RNS60 or NS (300 μl/mouse intraperitoneally every other day) starting at the disease onset and examined for disease progression as well as pathological and biochemical alterations. RESULTS RNS60 protected motor neurons in in vitro paradigms and slowed the disease progression of C57BL/6-SOD1G93A mice through a significant protection of spinal motor neurons and neuromuscular junctions. This was mediated by the (i) activation of an antioxidant response and generation of an anti-inflammatory environment in the spinal cord; (ii) activation of the PI3K-Akt pro-survival pathway in the spinal cord and sciatic nerves; (iii) reduced demyelination of the sciatic nerves; and (iv) elevation of peripheral CD4+/Foxp3+ T regulatory cell numbers. RNS60 did not show the same effects in 129Sv-SOD1G93A mice, which are unable to activate a protective immune response. CONCLUSION RNS60 demonstrated significant therapeutic efficacy in C57BL/6-SOD1G93A mice by virtue of its effects on multiple disease mechanisms in motor neurons, glial cells, and peripheral immune cells. These findings, together with the excellent clinical safety profile, make RNS60 a promising candidate for ALS therapy and support further studies to unravel its molecular mechanism of action. In addition, the differences in efficacy of RNS60 in SOD1G93A mice of different strains may be relevant for identifying potential markers to predict efficacy in clinical trials.
Collapse
Affiliation(s)
- Antonio Vallarola
- Molecular Neurobiology Lab, Department of Neuroscience, IRCCS - Mario Negri Institute, Via La Masa, 19, 20156, Milan, Italy
| | - Francesca Sironi
- Molecular Neurobiology Lab, Department of Neuroscience, IRCCS - Mario Negri Institute, Via La Masa, 19, 20156, Milan, Italy
| | - Massimo Tortarolo
- Molecular Neurobiology Lab, Department of Neuroscience, IRCCS - Mario Negri Institute, Via La Masa, 19, 20156, Milan, Italy
| | - Noemi Gatto
- Molecular Neurobiology Lab, Department of Neuroscience, IRCCS - Mario Negri Institute, Via La Masa, 19, 20156, Milan, Italy
| | - Roberta De Gioia
- Molecular Neurobiology Lab, Department of Neuroscience, IRCCS - Mario Negri Institute, Via La Masa, 19, 20156, Milan, Italy
| | - Laura Pasetto
- Translational Biomarkers Lab, Department of Molecular Biochemistry and Pharmacology, IRCCS - Mario Negri, Milan, Italy
| | - Massimiliano De Paola
- Analytical Biochemistry Lab, Department of Environmental Health Sciences, IRCCS- Mario Negri Institute, Milan, Italy
| | - Alessandro Mariani
- Analytical Biochemistry Lab, Department of Environmental Health Sciences, IRCCS- Mario Negri Institute, Milan, Italy
| | | | | | | | - Valentina Bonetto
- Translational Biomarkers Lab, Department of Molecular Biochemistry and Pharmacology, IRCCS - Mario Negri, Milan, Italy
| | - Caterina Bendotti
- Molecular Neurobiology Lab, Department of Neuroscience, IRCCS - Mario Negri Institute, Via La Masa, 19, 20156, Milan, Italy.
| |
Collapse
|
17
|
Increase in Mitochondrial Biogenesis in Neuronal Cells by RNS60, a Physically-Modified Saline, via Phosphatidylinositol 3-Kinase-Mediated Upregulation of PGC1α. J Neuroimmune Pharmacol 2017; 13:143-162. [PMID: 29188424 PMCID: PMC5928179 DOI: 10.1007/s11481-017-9771-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/10/2017] [Indexed: 11/25/2022]
Abstract
This study highlights a novel approach to upregulate mitochondrial biogenesis in neuronal cells. RNS60 is a 0.9% saline solution containing oxygenated nanobubbles that is generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not NS (normal saline), PNS60 (saline containing a comparable level of oxygen without the TCP modification), or RNS10.3 (TCP-modified normal saline without excess oxygen), increased the expression of Nrf1, Tfam, Mcu, and Tom20 (genes associated with mitochondrial biogenesis) and upregulated mitochondrial biogenesis in MN9D dopaminergic neuronal cells. Similarly RNS60 also increased mitochondrial biogenesis in primary dopaminergic neurons and in the nigra of MPTP-intoxicated mice. However, RNS60 had no effect on lysosomal biogenesis. Interestingly, we found that RNS60 upregulated PGC1α and siRNA knockdown of PGC1α abrogated the ability of RNS60 to increase mitochondrial biogenesis. Furthermore, we delineated that RNS60 increased the transcription of Pgc1a via type IA phosphatidylinositol (PI) 3-kinase-mediated activation of cAMP-response element-binding protein (CREB). Accordingly, knockdown of the PI3K – CREB pathway suppressed RNS60-mediated mitochondrial biogenesis. These results describe a novel property of RNS60 of enhancing mitochondrial biogenesis via PI 3-kinase-CREB-mediated up-regulation of PGC1α, which may be of therapeutic benefit in different neurodegenerative disorders.
Collapse
|
18
|
Jana M, Ghosh S, Pahan K. Upregulation of Myelin Gene Expression by a Physically-Modified Saline via Phosphatidylinositol 3-Kinase-Mediated Activation of CREB: Implications for Multiple Sclerosis. Neurochem Res 2017; 43:407-419. [PMID: 29143164 PMCID: PMC5799355 DOI: 10.1007/s11064-017-2435-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/03/2017] [Accepted: 11/09/2017] [Indexed: 12/29/2022]
Abstract
An increase in central nervous system (CNS) remyelination and a decrease in CNS inflammation are important steps to halt the progression of multiple sclerosis (MS). RNS60 is a bioactive aqueous solution generated by subjecting normal saline to Taylor–Couette–Poiseuille flow under elevated oxygen pressure. Recently we have demonstrated that RNS60 exhibits anti-inflammatory properties. Here, we describe promyelinating property of RNS60. RNS60, but not normal saline (NS), RNS10.3 (TCP-modified saline without excess oxygen) or PNS60 (saline containing excess oxygen without TCP modification), stimulated the expression of myelin-specific genes and proteins (myelin basic protein, MBP; myelin oligodendrocyte glycoprotein, MOG and proteolipid protein, PLP) in primary mouse oligodendroglia and mixed glial cells. While investigating the mechanisms, we found that RNS60 treatment induced the activation of cAMP response element binding protein (CREB) in oligodendrocytes, ultimately leading to the recruitment of CREB to the promoters of myelin-specific genes. Furthermore, activation of type 1A p110β/α, but not type 1B p110γ, phosphatidylinositol-3 (PI-3) kinase by RNS60 together with abrogation of RNS60-mediated activation of CREB and upregulation of myelin genes by LY294002 (a specific inhibitor of PI-3 kinase) suggest that RNS60 upregulates the activation of CREB and the expression of myelin-specific molecules in oligodendrocytes via activation of PI3 kinase. These results highlight a novel promyelinating property of RNS60, which may be of benefit for MS and other demyelinating disorders.
Collapse
Affiliation(s)
- Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA
| | - Supurna Ghosh
- Revalesio Corporation, 1200 East D Street, Tacoma, WA, 98421, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite 310, Chicago, IL, 60612, USA.
| |
Collapse
|
19
|
Therapeutic Strategies Under Development Targeting Inflammatory Mechanisms in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2017; 55:2789-2813. [PMID: 28455693 DOI: 10.1007/s12035-017-0532-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
|
20
|
Ivannikov MV, Sugimori M, Llinás RR. Neuromuscular transmission and muscle fatigue changes by nanostructured oxygen. Muscle Nerve 2017; 55:555-563. [PMID: 27422738 DOI: 10.1002/mus.25248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/29/2016] [Accepted: 07/12/2016] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Oxygen (O2 ) nanobubbles offer a new method for tissue oxygenation. The effects of O2 nanobubbles on transmission at neuromuscular junctions (NMJs) and muscle function were explored in murine diaphragm. METHODS Electrophysiological parameters, NMJ ultrastructure, muscle force, and muscle fatigue were studied during superfusion with solutions with different oxygen levels or oxygen nanobubbles. RESULTS High frequency nerve stimulation of muscles superfused with O2 nanobubble solution slowed neurotransmission decline over those with either control or hyperoxic solution. O2 nanobubble solution increased the amplitude of evoked end plate potentials and quantal content but did not affect spontaneous activity. Electron microscopy of stimulated O2 nanobubble treated NMJs showed accumulation of large synaptic vesicles and endosome-like structures. O2 nanobubble solution had no effects on isometric muscle force, but it significantly decreased fatigability and maximum force recovery time in nerve stimulated muscles. CONCLUSIONS O2 nanobubbles increase neurotransmission and reduce the probability of neurotransmission failure in muscle fatigue. Muscle Nerve 55: 555-563, 2017.
Collapse
Affiliation(s)
- Maxim V Ivannikov
- Department of Neuroscience and Physiology, NYU School of Medicine, 550 First Avenue, New York, New York, 10016, USA
| | - Mutsuyuki Sugimori
- Department of Neuroscience and Physiology, NYU School of Medicine, 550 First Avenue, New York, New York, 10016, USA
| | - Rodolfo R Llinás
- Department of Neuroscience and Physiology, NYU School of Medicine, 550 First Avenue, New York, New York, 10016, USA
| |
Collapse
|
21
|
Li M, Tonggu L, Zhan X, Mega TL, Wang L. Cryo-EM Visualization of Nanobubbles in Aqueous Solutions. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:11111-11115. [PMID: 27022010 DOI: 10.1021/acs.langmuir.6b00261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The detection of nanobubbles on surfaces is well established (e.g., AFM and optical microscopy methods), but currently no methods exist for the direct detection of bulk nanobubbles. Here, cryo-electron microscopy (cryo-EM) has been employed to observe bubbles in aqueous solutions for the first time. Nitrogen bubbles generated by a chemical reaction were observed in amorphous ice trapped between two carbon films. The cryo-EM images of bubbles showed the same features as predicted by theory. The fact that no bubbles were observed near an air-water interface suggests that bubbles may diffuse to the nearby air-water interface and escape. The estimate of the bubble diffusion coefficient is about 30-250 μm2/s.
Collapse
Affiliation(s)
- Mo Li
- Department of Biological Structure, University of Washington , Seattle, Washington 98195, United States
| | - Lige Tonggu
- Department of Biological Structure, University of Washington , Seattle, Washington 98195, United States
| | - Xi Zhan
- Department of Biological Structure, University of Washington , Seattle, Washington 98195, United States
| | - Tony L Mega
- Revalesio Corporation, 1200 East D Street, Tacoma, Washington 98421, United States
| | - Liguo Wang
- Department of Biological Structure, University of Washington , Seattle, Washington 98195, United States
| |
Collapse
|
22
|
Identification and characterization of PPARα ligands in the hippocampus. Nat Chem Biol 2016; 12:1075-1083. [PMID: 27748752 PMCID: PMC5110367 DOI: 10.1038/nchembio.2204] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/08/2016] [Indexed: 02/01/2023]
Abstract
Peroxisome proliferator-activated receptor-α (PPARα) regulates hepatic fatty acid catabolism and mediates the metabolic response to starvation. Recently we found that PPARα is constitutively activated in nuclei of hippocampal neurons and controls plasticity via direct transcriptional activation of CREB. Here we report the discovery of three endogenous PPARα ligands-3-hydroxy-(2,2)-dimethyl butyrate, hexadecanamide, and 9-octadecenamide-in mouse brain hippocampus. Mass spectrometric detection of these compounds in mouse hippocampal nuclear extracts, in silico interaction studies, time-resolved FRET analyses, and thermal shift assay results clearly indicated that these three compounds served as ligands of PPARα. Site-directed mutagenesis studies further revealed that PPARα Y464 and Y314 are involved in binding these hippocampal ligands. Moreover, these ligands activated PPARα and upregulated the synaptic function of hippocampal neurons. These results highlight the discovery of hippocampal ligands of PPARα capable of modulating synaptic functions.
Collapse
|
23
|
Modi KK, Rangasamy SB, Dasarathi S, Roy A, Pahan K. Cinnamon Converts Poor Learning Mice to Good Learners: Implications for Memory Improvement. J Neuroimmune Pharmacol 2016; 11:693-707. [PMID: 27342118 DOI: 10.1007/s11481-016-9693-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/15/2016] [Indexed: 11/28/2022]
Abstract
This study underlines the importance of cinnamon, a commonly used natural spice and flavoring material, and its metabolite sodium benzoate (NaB) in converting poor learning mice to good learning ones. NaB, but not sodium formate, was found to upregulate plasticity-related molecules, stimulate NMDA- and AMPA-sensitive calcium influx and increase of spine density in cultured hippocampal neurons. NaB induced the activation of CREB in hippocampal neurons via protein kinase A (PKA), which was responsible for the upregulation of plasticity-related molecules. Finally, spatial memory consolidation-induced activation of CREB and expression of different plasticity-related molecules were less in the hippocampus of poor learning mice as compared to good learning ones. However, oral treatment of cinnamon and NaB increased spatial memory consolidation-induced activation of CREB and expression of plasticity-related molecules in the hippocampus of poor-learning mice and converted poor learners into good learners. These results describe a novel property of cinnamon in switching poor learners to good learners via stimulating hippocampal plasticity.
Collapse
Affiliation(s)
- Khushbu K Modi
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite, Chicago, IL, 310, USA
| | - Suresh B Rangasamy
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite, Chicago, IL, 310, USA
| | - Sridevi Dasarathi
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite, Chicago, IL, 310, USA
| | - Avik Roy
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite, Chicago, IL, 310, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison St, Suite, Chicago, IL, 310, USA. .,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, 820 South Damen Avenue, Chicago, IL, USA.
| |
Collapse
|