1
|
Manickam R, Santhana S, Xuan W, Bisht KS, Tipparaju SM. Nampt: a new therapeutic target for modulating NAD + levels in metabolic, cardiovascular, and neurodegenerative diseases. Can J Physiol Pharmacol 2025. [PMID: 40203459 DOI: 10.1139/cjpp-2024-0400] [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: 04/11/2025]
Abstract
NAD+ is an important cofactor involved in regulating many biochemical processes in cells. An imbalance in NAD+/NADH ratio is linked to many diseases. NAD+ is depleted in diabetes, cardiovascular and neurodegenerative diseases, and in aging, and is increased in tumor cells. NAD+ is generated in cells via the de novo, Preiss-Handler, and salvage pathways. Most of the cellular NAD+ is generated through Nampt activation, a key rate-limiting enzyme that is involved in the salvage pathway. Restoration of NAD+/NADH balance offers therapeutic advantages for improving tissue homeostasis and function. NAD+ is known to benefit and restore the body's physiological mechanisms, including DNA replication, chromatin and epigenetic modifications, and gene expression. Recent studies elucidate the role of NAD+ in cells utilizing transgenic mouse models. Translational new therapeutics are positioned to utilize the NAD+ restoration strategies for overcoming the drawbacks that exist in the pharmacological toolkit. The present review highlights the significance of Nampt-NAD+ axis as a major player in energy metabolism and provides an overview with insights into future strategies, providing pharmacological advantages to address current and future medical needs.
Collapse
Affiliation(s)
- Ravikumar Manickam
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Sandhya Santhana
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Wanling Xuan
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Kirpal S Bisht
- Department of Chemistry, College of Arts and Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Bosc L, Pero ME, Balayssac D, Jacquemot N, Allard J, Suzanne P, Vollaire J, Cottet-Rousselle C, Michallet S, Villaret J, Torch S, Marais S, Elena-Herrmann B, Schlattner U, Mercier A, Josserand V, Thibert C, Dallemagne P, Bartolini F, Lafanechère L. Preventing neuropathy and improving anti-cancer chemotherapy with a carbazole-based compound. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642317. [PMID: 40161707 PMCID: PMC11952460 DOI: 10.1101/2025.03.10.642317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Advances in cancer treatment have led to a steady increase in the rate of disease remission. However, while many treatment-related adverse effects gradually resolve after therapy, chemotherapy-induced peripheral neuropathy (CIPN) often persists, with no means of prevention or direct treatment available. Herein, we present Carba1, a novel bi-functional carbazole that mitigates neuropathy through two distinct mechanisms. First, by interacting with tubulin, Carba1 reduces the required dose of taxanes, widely used chemotherapy drugs notorious for their toxic side effects, including CIPN. Second, Carba1 activates nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD salvage pathway, triggering a metabolic rewiring that enhances the resilience of neurons and Schwann cells against chemotherapy-induced toxicity. We demonstrate the neuroprotective efficacy of Carba1 both in vitro, against neurotoxicity induced by paclitaxel (PTX), cisplatin, and bortezomib, and in vivo in a rat model of PTX-induced neuropathy. Importantly, we establish that Carba1 does not compromise the therapeutic efficacy of PTX nor promotes tumor growth. Comparative analyses of Carba1 derivatives further suggest the potential of designing compounds with either dual synergistic and neuroprotective activity or exclusive neuroprotective properties. Altogether, our findings position Carba1 as a promising therapeutic candidate for preventing CIPN, with the potential, if successfully translated to clinical settings, to improve both the quality of life and treatment outcome for cancer patients.
Collapse
Affiliation(s)
- Lauriane Bosc
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Maria Elena Pero
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University; New York, NY 10032, USA
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II; 80137 Naples, Italy
| | - David Balayssac
- Université Clermont Auvergne, U1107, NEURO-DOL, INSERM, CHU Clermont-Ferrand, Direction de la recherche clinique et de l’innovation ; Clermont-Ferrand, France
| | - Nathalie Jacquemot
- Université Clermont Auvergne, U1107, NEURO-DOL, INSERM, Clermont-Ferrand, France
| | - Jordan Allard
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Peggy Suzanne
- Université Normandie, UNICAEN, CERMN ; 14032 Caen, France
| | - Julien Vollaire
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Cécile Cottet-Rousselle
- Laboratory of Fundamental and Applied Bioenergetics, Université Grenoble Alpes, INSERM U1055; Grenoble, France
| | - Sophie Michallet
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Joran Villaret
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Sakina Torch
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Sumari Marais
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria; Pretoria 0028, South Africa
| | - Bénédicte Elena-Herrmann
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics, Université Grenoble Alpes, INSERM U1055; Grenoble, France
- Institut Universitaire de France (IUF) ; 75231 Paris, France
| | - Anne Mercier
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria; Pretoria 0028, South Africa
| | - Véronique Josserand
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | - Chantal Thibert
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
| | | | - Francesca Bartolini
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University; New York, NY 10032, USA
| | - Laurence Lafanechère
- Institute for Advanced Biosciences, Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309; Grenoble, France
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria; Pretoria 0028, South Africa
| |
Collapse
|
3
|
Zou Y, Wu S, Hu Q, Zhou H, Ge Y, Ju Z, Luo S. Sonic hedgehog restrains the ubiquitin-dependent degradation of SP1 to inhibit neuronal/glial senescence associated phenotypes in chemotherapy-induced peripheral neuropathy via the TRIM25-CXCL13 axis. J Adv Res 2025; 68:387-402. [PMID: 38479571 PMCID: PMC11785578 DOI: 10.1016/j.jare.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/22/2024] [Accepted: 03/10/2024] [Indexed: 03/19/2024] Open
Abstract
INTRODUCTION Chemotherapy-induced peripheral neuropathy (CIPN) is a common complication that affects an increasing number of cancer survivors. However, the current treatment options for CIPN are limited. Paclitaxel (PTX) is a widely used chemotherapeutic drug that induces senescence in cancer cells. While previous studies have demonstrated that Sonic hedgehog (Shh) can counteract cellular dysfunction during aging, its role in CIPN remains unknown. OBJECTIVES Herein, the aim of this study was to investigate whether Shh activation could inhibits neuronal/glial senescence and alleviates CIPN. METHODS We treated ND7/23 neuronal cells and RSC96 Schwann cells with two selective Shh activators (purmorphamine [PUR] and smoothened agonist [SAG]) in the presence of PTX. Additionally, we utilized a CIPN mouse model induced by PTX injection. To assess cellular senescence, we performed a senescence-associated β-galactosidase (SA-β-gal) assay, measured reactive oxygen species (ROS) levels, and examined the expression of P16, P21, and γH2AX. To understand the underlying mechanisms, we conducted ubiquitin assays, LC-MS/MS, H&E staining, and assessed protein expression through Western blotting and immunofluorescence staining. RESULTS In vitro, we observed that Shh activation significantly alleviated the senescence-related decline in multiple functions included SA-β-gal activity, expression of P16 and P21, cell viability, and ROS accumulation in DRG sensory neurons and Schwann cells after PTX exposure. Furthermore, our in vivo experiments demonstrated that Shh activation significantly reduced axonal degeneration, demyelination, and improved nerve conduction. Mechanistically, we discovered that PTX reduced the protein level of SP1, which was ubiquitinated by the E3 ligase TRIM25 at the lysine 694 (K694), leading to increased CXCL13 expression, and we found that Shh activation inhibited PTX-induced neuronal/glial senescence and CIPN through the TRIM25-SP1-CXCL13 axis. CONCLUSION These findings provide evidence for the role of PTX-induced senescence in DRG sensory neurons and Schwann cells, suggesting that Shh could be a potential therapeutic target for CIPN.
Collapse
Affiliation(s)
- Ying Zou
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Biology, School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qian Hu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Haoxian Zhou
- Department of Cardiology, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuanlong Ge
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Zhenyu Ju
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Biology, School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Shengkang Luo
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Biology, School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| |
Collapse
|
4
|
Nowicka B, Polkowska I, Zeliszewska-Duk P, Torres A, Duk M. Molecular Assessment of Plasma Concentrations of Selected Adipokines and IL-8 in Horses with Back Pain and Comorbid Asthma-Based on Clinical Cases. Animals (Basel) 2025; 15:310. [PMID: 39943080 PMCID: PMC11815831 DOI: 10.3390/ani15030310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Similarly, in humans and horses, thoracic and lumbosacral back pain cause more disability and work interruptions worldwide than any other disease. Given that there are few effective treatments for back pain in humans and animals, primary prevention strategies and a reduction in pain factors may be crucial. In the analysed data obtained for the horses studied, the pattern of changes in adipocytokine concentrations, including resistin, visfatin and leptin, was noted for those with back pain compared to the control animals. Concentrations of selected adipocytokines in horses from the back pain group were different in animals with a coexisting diagnosis of asthma and back dysfunction. Very few studies are available on adipokine concentrations in horses. No information was found in relation to back pain and asthma in these animals. In humans, correlations of back pain and asthma with concentrations of selected adipokines have been described.
Collapse
Affiliation(s)
- Beata Nowicka
- Department and Clinic of Animal Surgery, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland;
| | - Izabela Polkowska
- Department and Clinic of Animal Surgery, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland;
| | - Paulina Zeliszewska-Duk
- Department of Horse Breeding and Use, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland;
| | - Anna Torres
- Department of Pediatrics and Adolescent Gynecology, Medical University of Lublin, Chodzki 4, 20-093 Lublin, Poland;
| | - Mariusz Duk
- Department of Electronics and Information Technology, Faculty of Electrical Engineering and Computer Science, Lublin University of Technology, 20-618 Lublin, Poland;
| |
Collapse
|
5
|
Im S, Jeong DJ, Kim E, Choi JH, Jang HJ, Kim YY, Um JH, Lee J, Lee YJ, Lee KM, Choi D, Yoo E, Lee HS, Yun J. A novel marine-derived mitophagy inducer ameliorates mitochondrial dysfunction and thermal hypersensitivity in paclitaxel-induced peripheral neuropathy. Br J Pharmacol 2024; 181:4012-4027. [PMID: 38925168 DOI: 10.1111/bph.16476] [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: 06/29/2023] [Revised: 02/07/2024] [Accepted: 04/25/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND AND PURPOSE Mitochondrial dysfunction contributes to the pathogenesis and maintenance of chemotherapy-induced peripheral neuropathy (CIPN), a significant limitation of cancer chemotherapy. Recently, the stimulation of mitophagy, a pivotal process for mitochondrial homeostasis, has emerged as a promising treatment strategy for neurodegenerative diseases, but its therapeutic effect on CIPN has not been explored. Here, we assessed the mitophagy-inducing activity of 3,5-dibromo-2-(2',4'-dibromophenoxy)-phenol (PDE701), a diphenyl ether derivative isolated from the marine sponge Dysidea sp., and investigated its therapeutic effect on a CIPN model. EXPERIMENTAL APPROACH Mitophagy activity was determined by a previously established mitophagy assay using mitochondrial Keima (mt-Keima). Mitophagy induction was further verified by western blotting, immunofluorescence, and electron microscopy. Mitochondrial dysfunction was analysed by measuring mitochondrial superoxide levels in SH-SY5Y cells and Drosophila larvae. A thermal nociception assay was used to evaluate the therapeutic effect of PDE701 on the paclitaxel-induced thermal hyperalgesia phenotype in Drosophila larvae. KEY RESULTS PDE701 specifically induced mitophagy but was not toxic to mitochondria. PDE701 ameliorated paclitaxel-induced mitochondrial dysfunction in both SH-SY5Y cells and Drosophila larvae. Importantly, PDE701 also significantly ameliorated paclitaxel-induced thermal hyperalgesia in Drosophila larvae. Knockdown of ATG5 or ATG7 abolished the effect of PDE701 on thermal hyperalgesia, suggesting that PDE701 exerts its therapeutic effect through mitophagy induction. CONCLUSION AND IMPLICATIONS This study identified PDE701 as a novel mitophagy inducer and a potential therapeutic compound for CIPN. Our results suggest that mitophagy stimulation is a promising strategy for the treatment of CIPN and that marine organisms are a potential source of mitophagy-inducing compounds.
Collapse
Affiliation(s)
- Sangwoo Im
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Dae Jin Jeong
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Eunmi Kim
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Jae-Hyeong Choi
- Korea Institute of Ocean Science & Technology (KIOST), Busan, Republic of Korea
- Department of Applied Ocean Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Hye-Ji Jang
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Young Yeon Kim
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Jee-Hyun Um
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Jihoon Lee
- Korea Institute of Ocean Science & Technology (KIOST), Busan, Republic of Korea
- Department of Applied Ocean Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Yeon-Ju Lee
- Korea Institute of Ocean Science & Technology (KIOST), Busan, Republic of Korea
- Department of Applied Ocean Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Kang-Min Lee
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| | - Dabin Choi
- Altmedical Co., Ltd, Seoul, Republic of Korea
| | - Eunhee Yoo
- Altmedical Co., Ltd, Seoul, Republic of Korea
| | - Hyi-Seung Lee
- Korea Institute of Ocean Science & Technology (KIOST), Busan, Republic of Korea
- Department of Applied Ocean Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Jeanho Yun
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Republic of Korea
| |
Collapse
|
6
|
Naratadam GT, Mecklenburg J, Shein SA, Zou Y, Lai Z, Tumanov AV, Price TJ, Akopian AN. Degenerative and regenerative peripheral processes are associated with persistent painful chemotherapy-induced neuropathies in males and females. Sci Rep 2024; 14:17543. [PMID: 39080341 PMCID: PMC11289433 DOI: 10.1038/s41598-024-68485-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
This study investigated the time course of gene expression changes during the progression of persistent painful neuropathy caused by paclitaxel (PTX) in male and female mouse hindpaws and dorsal root ganglia (DRG). Bulk RNA-seq was used to examine these gene expression changes at 1, 16, and 31 days post-last PTX. At these time points, differentially expressed genes (DEGs) were predominantly related to the reduction or increase in epithelial, skin, bone, and muscle development and to angiogenesis, myelination, axonogenesis, and neurogenesis. These processes are accompanied by the regulation of DEGs related to the cytoskeleton, extracellular matrix organization, and cellular energy production. This gene plasticity during the progression of persistent painful neuropathy could be interpreted as a biological process linked to tissue regeneration/degeneration. In contrast, gene plasticity related to immune processes was minimal at 1-31 days after PTX. It was also noted that despite similarities in biological processes and pain chronicity between males and females, specific DEGs differed dramatically according to sex. The main conclusions of this study are that gene expression plasticity in hindpaw and DRG during PTX neuropathy progression similar to tissue regeneration and degeneration, minimally affects immune system processes and is heavily sex-dependent at the individual gene level.
Collapse
Affiliation(s)
- George T Naratadam
- South Texas Medical Scientist Training Program (STX-MSTP), Integrated Biomedical Sciences (IBMS) Program, The Long School of Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Jennifer Mecklenburg
- Department of Endodontics, School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Sergey A Shein
- Department of Microbiology, Immunology and Molecular Genetics, The Long School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Yi Zou
- Department of Molecular Medicine, The Long School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Zhao Lai
- Department of Molecular Medicine, The Long School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- South Texas Medical Scientist Training Program (STX-MSTP), Integrated Biomedical Sciences (IBMS) Program, The Long School of Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
- Department of Microbiology, Immunology and Molecular Genetics, The Long School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA.
| | - Theodore J Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| | - Armen N Akopian
- South Texas Medical Scientist Training Program (STX-MSTP), Integrated Biomedical Sciences (IBMS) Program, The Long School of Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
- Department of Endodontics, School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
7
|
Chen Y, Zhu Z, Yan Y, Sun H, Wang G, Du X, Li F, Yuan S, Wang W, Wang M, Gu C. P7C3 suppresses astrocytic senescence to protect dopaminergic neurons: Implication in the mouse model of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14819. [PMID: 39056208 PMCID: PMC11273101 DOI: 10.1111/cns.14819] [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: 11/02/2023] [Revised: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
AIMS Astrocytic senescence is inextricably linked to aging and neurodegenerative disorders, including Parkinson's disease (PD). P7C3 is a small, neuroprotective aminopropyl carbazole compound that exhibits anti-inflammatory properties. However, the effects of P7C3 on astrocytic senescence in PD remain to be elucidated. METHODS An in vitro, long culture-induced, replicative senescence cell model and a 1-methyl-4-phenylpyridinium (MPP+)/rotenone-induced premature senescence cell model were used to investigate the effects of P7C3 on astrocytic senescence. An in vivo, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse PD model was used to study the role of P7C3 in astrocytic senescence. Immunoblotting, real-time quantitative RT-PCR (qPCR), immunofluorescence, subcellular fractionation assays, and immunohistochemistry were utilized to confirm the effects of P7C3 on astrocytic senescence and elucidate its underlying mechanisms. RESULTS This study determined that P7C3 suppressed the senescence-associated secretory phenotype (SASP) in both cell models, as demonstrated by the reduction in the critical senescence marker p16 and proinflammatory factors (IL-6, IL-1β, CXCL10, and MMP9) and increased laminB1 levels, implying that P7C3 inhibited replicative astrocytic senescence and MPP+/rotenone-induced premature astrocytic senescence, Most importantly, we demonstrated that P7C3 prevented the death of dopamine (DA) neurons and reduced the behavioral deficits in the MPTP-induced mouse model of PD, which is accompanied by a decrease in senescent astrocytes in the substantia nigra compacta (SNc). Mechanistically, P7C3 promoted Nrf2/Sirt3-mediated mitophagy and reduced mitochondrial reactive oxygen species (mitoROS) generation, which contributed to the suppression of astrocytic senescence. Furthermore, Sirt3 deficiency obviously abolished the inhibitory effects of P7C3 on astrocytic senescence. CONCLUSION This study revealed that P7C3 inhibited astrocytic senescence via increased Nrf2/Sirt3-mediated mitophagy and suppression of mitoROS, which further protected against DA neuronal loss. These observations provide a prospective theoretical basis for P7C3 in the treatment of age-associated neurodegenerative diseases, such as PD.
Collapse
Affiliation(s)
- Yajing Chen
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Zengyan Zhu
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Yinghui Yan
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Hongyang Sun
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsuChina
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders, Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouJiangsuChina
| | - Xiaohuan Du
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Fang Li
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Shuwei Yuan
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Wenjing Wang
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Mei Wang
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| | - Chao Gu
- Department of PharmacyChildren's Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
8
|
Wei F, Hughes M, Omer M, Ngo C, Pugazhendhi AS, Kolanthai E, Aceto M, Ghattas Y, Razavi M, Kean TJ, Seal S, Coathup M. A Multifunctional Therapeutic Strategy Using P7C3 as A Countermeasure Against Bone Loss and Fragility in An Ovariectomized Rat Model of Postmenopausal Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308698. [PMID: 38477537 PMCID: PMC11151083 DOI: 10.1002/advs.202308698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Indexed: 03/14/2024]
Abstract
By 2060, an estimated one in four Americans will be elderly. Consequently, the prevalence of osteoporosis and fragility fractures will also increase. Presently, no available intervention definitively prevents or manages osteoporosis. This study explores whether Pool 7 Compound 3 (P7C3) reduces progressive bone loss and fragility following the onset of ovariectomy (OVX)-induced osteoporosis. Results confirm OVX-induced weakened, osteoporotic bone together with a significant gain in adipogenic body weight. Treatment with P7C3 significantly reduced osteoclastic activity, bone marrow adiposity, whole-body weight gain, and preserved bone area, architecture, and mechanical strength. Analyses reveal significantly upregulated platelet derived growth factor-BB and leukemia inhibitory factor, with downregulation of interleukin-1 R6, and receptor activator of nuclear factor kappa-B (RANK). Together, proteomic data suggest the targeting of several key regulators of inflammation, bone, and adipose turnover, via transforming growth factor-beta/SMAD, and Wingless-related integration site/be-catenin signaling pathways. To the best of the knowledge, this is first evidence of an intervention that drives against bone loss via RANK. Metatranscriptomic analyses of the gut microbiota show P7C3 increased Porphyromonadaceae bacterium, Candidatus Melainabacteria, and Ruminococcaceae bacterium abundance, potentially contributing to the favorable inflammatory, and adipo-osteogenic metabolic regulation observed. The results reveal an undiscovered, and multifunctional therapeutic strategy to prevent the pathological progression of OVX-induced bone loss.
Collapse
Affiliation(s)
- Fei Wei
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
| | - Megan Hughes
- School of BiosciencesCardiff UniversityWalesCF10 3ATUK
| | - Mahmoud Omer
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
| | - Christopher Ngo
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | | | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC)University of Central FloridaOrlandoFL32826USA
| | - Matthew Aceto
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Yasmine Ghattas
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Mehdi Razavi
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Thomas J Kean
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| | - Sudipta Seal
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC)University of Central FloridaOrlandoFL32826USA
| | - Melanie Coathup
- Biionix ClusterUniversity of Central FloridaOrlandoFL82816USA
- College of MedicineUniversity of Central FloridaOrlandoFL32827USA
| |
Collapse
|
9
|
Dai Y, Lin J, Chen X, Ren J, Wu C, Shen H, Li X, Yu J, Jiang B, Yu L. NAMPT/NAD +/PARP1 Pathway Regulates CFA-Induced Inflammatory Pain via NF-κB Signaling in Rodents. Adv Biol (Weinh) 2024; 8:e2400028. [PMID: 38463014 DOI: 10.1002/adbi.202400028] [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: 01/16/2024] [Revised: 02/26/2024] [Indexed: 03/12/2024]
Abstract
Emerging evidence has implicated nicotinamide adenine dinucleotide (NAD+) metabolism in various inflammatory diseases. In the study, the role of NAD+ metabolism in Complete Freund's Adjuvant (CFA)-evoked inflammatory pain and the underlying mechanisms are investigated. The study demonstrated that CFA induced upregulation of nicotinamide phosphoribosyltransferase (NAMPT) in dorsal root ganglia (DRG) without significant changes in the spinal cord. Inhibition of NAMPT expression by intrathecal injection of NAMPT siRNA alleviated CFA-induced pain-like behavior, decreased NAD+ contents in DRG, and lowered poly-(ADP-ribose) polymerase 1 (PARP1) activity levels. These effects are all reversed by the supplement of nicotinamide mononucleotide (NMN). Inhibition of PARP1 expression by intrathecal injection of PARP1 siRNA alleviated CFA-induced pain-like behavior, while elevated NAD+ levels of DRG. The analgesic effect of inhibiting NAMPT/NAD+/PARP1 axis can be attributed to the downregulation of the NF-κB/IL-1β inflammatory pathway. Double immunofluorescence staining showed that the expression of NAMPT/NAD+/PARP1 axis is restricted to DRG neurons. In conclusion, PARP1 activation in response to CFA stimulation, fueled by NAMPT-derived NAD+, mediates CFA-induced inflammatory pain through NF-κB/IL-1β inflammatory pathway.
Collapse
Affiliation(s)
- Yi Dai
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiaqi Lin
- East Hospital Affiliated to Tongji University, Shanghai, 200000, China
| | - Xiangde Chen
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jinxuan Ren
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chengwei Wu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Lishui Municipal Central Hospital, Lishui, 323000, China
| | - Huihui Shen
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Baochun Jiang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Lina Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
10
|
Velma G, Krider IS, Alves ETM, Courey JM, Laham MS, Thatcher GRJ. Channeling Nicotinamide Phosphoribosyltransferase (NAMPT) to Address Life and Death. J Med Chem 2024; 67:5999-6026. [PMID: 38580317 PMCID: PMC11056997 DOI: 10.1021/acs.jmedchem.3c02112] [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: 11/11/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step in NAD+ biosynthesis via salvage of NAM formed from catabolism of NAD+ by proteins with NADase activity (e.g., PARPs, SIRTs, CD38). Depletion of NAD+ in aging, neurodegeneration, and metabolic disorders is addressed by NAD+ supplementation. Conversely, NAMPT inhibitors have been developed for cancer therapy: many discovered by phenotypic screening for cancer cell death have low nanomolar potency in cellular models. No NAMPT inhibitor is yet FDA-approved. The ability of inhibitors to act as NAMPT substrates may be associated with efficacy and toxicity. Some 3-pyridyl inhibitors become 4-pyridyl activators or "NAD+ boosters". NAMPT positive allosteric modulators (N-PAMs) and boosters may increase enzyme activity by relieving substrate/product inhibition. Binding to a "rear channel" extending from the NAMPT active site is key for inhibitors, boosters, and N-PAMs. A deeper understanding may fulfill the potential of NAMPT ligands to regulate cellular life and death.
Collapse
Affiliation(s)
- Ganga
Reddy Velma
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Isabella S. Krider
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Erick T. M. Alves
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Jenna M. Courey
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Megan S. Laham
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Gregory R. J. Thatcher
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
11
|
Li F, Wu C, Wang G. Targeting NAD Metabolism for the Therapy of Age-Related Neurodegenerative Diseases. Neurosci Bull 2024; 40:218-240. [PMID: 37253984 PMCID: PMC10838897 DOI: 10.1007/s12264-023-01072-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/10/2023] [Indexed: 06/01/2023] Open
Abstract
As the aging population continues to grow rapidly, age-related diseases are becoming an increasing burden on the healthcare system and a major concern for the well-being of elderly individuals. While aging is an inevitable process for all humans, it can be slowed down and age-related diseases can be treated or alleviated. Nicotinamide adenine dinucleotide (NAD) is a critical coenzyme or cofactor that plays a central role in metabolism and is involved in various cellular processes including the maintenance of metabolic homeostasis, post-translational protein modifications, DNA repair, and immune responses. As individuals age, their NAD levels decline, and this decrease has been suggested to be a contributing factor to the development of numerous age-related diseases, such as cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. In pursuit of healthy aging, researchers have investigated approaches to boost or maintain NAD levels. Here, we provide an overview of NAD metabolism and the role of NAD in age-related diseases and summarize recent progress in the development of strategies that target NAD metabolism for the treatment of age-related diseases, particularly neurodegenerative diseases.
Collapse
Affiliation(s)
- Feifei Li
- School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Chou Wu
- School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gelin Wang
- School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
12
|
Hovhannisyan AH, Lindquist KA, Belugin S, Mecklenburg J, Ibrahim T, Tram M, Corey TM, Salmon AB, Perez D, Ruparel S, Akopian AN. Sensory innervation of masseter, temporal and lateral pterygoid muscles in common marmosets. Sci Rep 2023; 13:23062. [PMID: 38155190 PMCID: PMC10754842 DOI: 10.1038/s41598-023-49882-9] [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: 04/26/2023] [Accepted: 12/13/2023] [Indexed: 12/30/2023] Open
Abstract
Myogenous temporomandibular disorders is associated with an increased responsiveness of nerves innervating the masseter (MM), temporal (TM), and lateral pterygoid muscles (LPM). This study aimed to examine sensory nerve types innervating MM, TM and LPM of adult non-human primate-common marmosets. Sensory nerves were localized in specific regions of these muscles. Pgp9.5, marker for all nerves, and NFH, a marker for A-fibers, showed that masticatory muscles were primarily innervated with A-fibers. The proportion of C- to A-fibers was highest in LPM, and lowest in MM. All C-fibers (pgp9.5+/NFH-) observed in masticatory muscles were peptidergic (CGRP+) and lacked mrgprD and CHRNA3, a silent nociceptive marker. TrpV1 was register in 17% of LPM nerves. All fibers in masticatory muscles were labeled with GFAP+, a myelin sheath marker. There were substantially more peptidergic A-fibers (CGRP+/NFH+) in TM and LPM compared to MM. MM, TM and LPM NFH+ fibers contained different percentages of trkC+ and parvalbumin+, but not trkB+ fibers. Tyrosine hydroxylase antibodies, which did not label TG, highlighted sympathetic fibers around blood vessels of the masticatory muscles. Overall, masticatory muscle types of marmosets have similarities and differences in innervation patterns.
Collapse
Affiliation(s)
- Anahit H Hovhannisyan
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Karen A Lindquist
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Sergei Belugin
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Jennifer Mecklenburg
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Tarek Ibrahim
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Meilinn Tram
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Tatiana M Corey
- Departments of Laboratory Animal Resources, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Adam B Salmon
- Departments of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, Geriatric Research Education and Clinical Center San Antonio, San Antonio, TX, 78229, USA
| | - Daniel Perez
- Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Shivani Ruparel
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Armen N Akopian
- Departments of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
- Integrated Biomedical Sciences (IBMS) Program, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Departments of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
13
|
Sridharan PS, Miller E, Pieper AA. Application of P7C3 Compounds to Investigating and Treating Acute and Chronic Traumatic Brain Injury. Neurotherapeutics 2023; 20:1616-1628. [PMID: 37651054 PMCID: PMC10684439 DOI: 10.1007/s13311-023-01427-8] [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] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading worldwide cause of disability, and there are currently no medicines that prevent, reduce, or reverse acute or chronic neurodegeneration in TBI patients. Here, we review the target-agnostic discovery of nicotinamide adenine dinucleotide (NAD+)/NADH-stabilizing P7C3 compounds through a phenotypic screen in mice and describe how P7C3 compounds have been applied to advance understanding of the pathophysiology and potential treatment of TBI. We summarize how P7C3 compounds have been shown across multiple laboratories to mitigate disease progression safely and effectively in a broad range of preclinical models of disease related to impaired NAD+/NADH metabolism, including acute and chronic TBI, and note the reported safety and neuroprotective efficacy of P7C3 compounds in nonhuman primates. We also describe how P7C3 compounds facilitated the recent first demonstration that chronic neurodegeneration 1 year after TBI in mice, the equivalent of many decades in people, can be reversed to restore normal neuropsychiatric function. We additionally review how P7C3 compounds have facilitated discovery of new pathophysiologic mechanisms of neurodegeneration after TBI. This includes the role of rapid TBI-induced tau acetylation that drives axonal degeneration, and the discovery of brain-derived acetylated tau as the first blood-based biomarker of neurodegeneration after TBI that directly correlates with the abundance of a therapeutic target in the brain. We additionally review the identification of TBI-induced tau acetylation as a potential mechanistic link between TBI and increased risk of Alzheimer's disease. Lastly, we summarize historical accounts of other successful phenotypic-based drug discoveries that advanced medical care without prior recognition of the specific molecular target needed to achieve the desired therapeutic effect.
Collapse
Affiliation(s)
- Preethy S Sridharan
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Emiko Miller
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA.
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
14
|
Tram M, Ibrahim T, Hovhannisyan A, Akopian A, Ruparel S. Lingual innervation in male and female marmosets. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 14:100134. [PMID: 38099285 PMCID: PMC10719518 DOI: 10.1016/j.ynpai.2023.100134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 12/17/2023]
Abstract
Several gaps in knowledge exists in our understanding of orofacial pain. Some of these include type of peripheral sensory innervation in specific tissues, differences in innervation between sexes and validation of rodent studies in higher order species. The current study addresses these gaps by validating mouse studies for sensory innervation of tongue tissue in non-human primates as well as assesses sex-specific differences. Tongue and trigeminal ganglia were collected from naïve male and female marmosets and tested for nerve fibers using specific markers by immunohistochemistry and number of fibers quantified. We also tested whether specific subgroups of nerve fibers belonged to myelinating or non-myelinating axons. We observed that similar to findings in mice, marmoset tongue was innervated with nerve filaments expressing nociceptor markers like CGRP and TRPV1 as well as non-nociceptor markers like TrkB, parvalbumin (PV) and tyrosine hydroxylase (TH). Furthermore, we found that while portion of TrkB and PV may be sensory fibers, TH-positive fibers were primarily sympathetic nerve fibers. Moreover, number of CGRP, TrkB and TH-positive nerve fibers were similar in both sexes. However, we observed a higher proportion of myelinated TRPV1 positive fibers in females than in males as well as increased number of PV + fibers in females. Taken together, the study for the first time characterizes sensory innervation in non-human primates as well as evaluates sex-differences in innervation of tongue tissue, thereby laying the foundation for future orofacial pain research with new world smaller NHPs like the common marmoset.
Collapse
Affiliation(s)
- Meilinn Tram
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Tarek Ibrahim
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Anahit Hovhannisyan
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Armen Akopian
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Shivani Ruparel
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| |
Collapse
|
15
|
Wang L, Liu M, Zu Y, Yao H, Wu C, Zhang R, Ma W, Lu H, Xi S, Liu Y, Hua L, Wang G, Tang Y. Optimization of NAMPT activators to achieve in vivo neuroprotective efficacy. Eur J Med Chem 2022; 236:114260. [DOI: 10.1016/j.ejmech.2022.114260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/04/2022]
|
16
|
Current and Emerging Pharmacotherapeutic Interventions for the Treatment of Peripheral Nerve Disorders. Pharmaceuticals (Basel) 2022; 15:ph15050607. [PMID: 35631433 PMCID: PMC9144529 DOI: 10.3390/ph15050607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Peripheral nerve disorders are caused by a range of different aetiologies. The range of causes include metabolic conditions such as diabetes, obesity and chronic kidney disease. Diabetic neuropathy may be associated with severe weakness and the loss of sensation, leading to gangrene and amputation in advanced cases. Recent studies have indicated a high prevalence of neuropathy in patients with chronic kidney disease, also known as uraemic neuropathy. Immune-mediated neuropathies including Guillain-Barré syndrome and chronic inflammatory demyelinating polyradiculoneuropathy may cause significant physical disability. As survival rates continue to improve in cancer, the prevalence of treatment complications, such as chemotherapy-induced peripheral neuropathy, has also increased in treated patients and survivors. Notably, peripheral neuropathy associated with these conditions may be chronic and long-lasting, drastically affecting the quality of life of affected individuals, and leading to a large socioeconomic burden. This review article explores some of the major emerging clinical and experimental therapeutic agents that have been investigated for the treatment of peripheral neuropathy due to metabolic, toxic and immune aetiologies.
Collapse
|
17
|
Gao SJ, Liu DQ, Li DY, Sun J, Zhang LQ, Wu JY, Song FH, Zhou YQ, Mei W. Adipocytokines: Emerging therapeutic targets for pain management. Biomed Pharmacother 2022; 149:112813. [PMID: 35279597 DOI: 10.1016/j.biopha.2022.112813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
Although pain has lower mortality rates than cancer, diabetes and stroke, pain is a predominate source of distress and disability. However, the management of pain remains an enormous problem. Many drugs used to pain treatment have more or less side effects. Therefore, the development of novel therapeutic target is critical for the treatment of pain. Notably, studies have shown that adipocytokines have a dual role in pain. Growing shreds of evidence shows that the levels of adipocytokines are upregulated or downregulated in the development of pain. In addition, substantial evidence indicates that regulation of adipocytokines levels in models of pain attenuates or promotes pain behaviors. In this review, we summarized and discussed the effect of adipocytokines in pain. These evidence indicates that adipocytokines attenuate or promote pain behaviors through interacting with their receptors, activating serotonin pathway, interacting with μ-opioid receptor, activating microglia, infiltrating macrophage and so on. Overall, adipocytokines have some potential in treating pain, but the underlying mechanisms remain unclear and need to be further studied.
Collapse
Affiliation(s)
- Shao-Jie Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Dai-Qiang Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Dan-Yang Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jia Sun
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Long-Qing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jia-Yi Wu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Fan-He Song
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ya-Qun Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Wei Mei
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
18
|
Discovery of small-molecule activators of nicotinamide phosphoribosyltransferase (NAMPT) and their preclinical neuroprotective activity. Cell Res 2022; 32:570-584. [PMID: 35459935 DOI: 10.1038/s41422-022-00651-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/19/2022] [Indexed: 01/07/2023] Open
Abstract
The decline of nicotinamide adenine dinucleotide (NAD) occurs in a variety of human pathologies including neurodegeneration. NAD-boosting agents can provide neuroprotective benefits. Here, we report the discovery and development of a class of potent activators (NATs) of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD salvage pathway. We obtained the crystal structure of NAMPT in complex with the NAT, which defined the allosteric action of NAT near the enzyme active site. The optimization of NAT further revealed the critical role of K189 residue in boosting NAMPT activity. NATs effectively increased intracellular levels of NAD and induced subsequent metabolic and transcriptional reprogramming. Importantly, NATs exhibited strong neuroprotective efficacy in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN) without any overt toxicity. These findings demonstrate the potential of NATs in the treatment of neurodegenerative diseases or conditions associated with NAD level decline.
Collapse
|
19
|
Doyle TM, Salvemini D. Mini-Review: Mitochondrial dysfunction and chemotherapy-induced neuropathic pain. Neurosci Lett 2021; 760:136087. [PMID: 34182057 DOI: 10.1016/j.neulet.2021.136087] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a somatosensory axonopathy in cancer patients receiving any of a variety of widely-use antitumor agents. CIPN can lead to long-lasting neuropathic pain that limits the dose or length of otherwise life-saving cancer therapy. Accumulating evidence over the last two decades indicates that many chemotherapeutic agents cause mitochondrial injury in the peripheral sensory nerves by disrupting mitochondrial structure and bioenergetics, increasing nitro-oxidative stress and altering mitochondrial transport, fission, fusion and mitophagy. The accumulation of abnormal and dysfunctional mitochondria in sensory neurons are linked to axonal growth defects resulting in the loss of intraepidermal nerve fibers in the hands and feet, increased spontaneous discharge and the sensitization of peripheral sensory neurons that provoke and promote changes in the central nervous system that establish a chronic neuropathic pain state. This has led to the propose mitotoxicity theory of CIPN. Strategies that improve mitochondrial function have shown success in preventing and reversing CIPN in pre-clinical animal models and have begun to show some progress toward translation to the clinic. In this review, we will review the evidence for, the causes and effects of and current strategies to target mitochondrial dysfunction in CIPN.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO 63104, USA.
| |
Collapse
|
20
|
Boyd JT, LoCoco PM, Furr AR, Bendele MR, Tram M, Li Q, Chang FM, Colley ME, Samenuk GM, Arris DA, Locke EE, Bach SBH, Tobon A, Ruparel SB, Hargreaves KM. Elevated dietary ω-6 polyunsaturated fatty acids induce reversible peripheral nerve dysfunction that exacerbates comorbid pain conditions. Nat Metab 2021; 3:762-773. [PMID: 34140694 PMCID: PMC8287645 DOI: 10.1038/s42255-021-00410-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/12/2021] [Indexed: 02/05/2023]
Abstract
Chronic pain is the leading cause of disability worldwide1 and is commonly associated with comorbid disorders2. However, the role of diet in chronic pain is poorly understood. Of particular interest is the Western-style diet, enriched with ω-6 polyunsaturated fatty acids (PUFAs) that accumulate in membrane phospholipids and oxidise into pronociceptive oxylipins3,4. Here we report that mice administered an ω-6 PUFA-enriched diet develop persistent nociceptive hypersensitivities, spontaneously active and hyper-responsive glabrous afferent fibres and histologic markers of peripheral nerve damage reminiscent of a peripheral neuropathy. Linoleic and arachidonic acids accumulate in lumbar dorsal root ganglia, with increased liberation via elevated phospholipase (PLA)2 activity. Pharmacological and molecular inhibition of PLA2G7 or diet reversal with high levels of ω-3 PUFAs attenuate nociceptive behaviours, neurophysiologic abnormalities and afferent histopathology induced by high ω-6 intake. Additionally, ω-6 PUFA accumulation exacerbates allodynia observed in preclinical inflammatory and neuropathic pain models and is strongly correlated with multiple pain indices of clinical diabetic neuropathy. Collectively, these data reveal dietary enrichment with ω-6 PUFAs as a new aetiology of peripheral neuropathy and risk factor for chronic pain and implicate multiple therapeutic considerations for clinical pain management.
Collapse
Affiliation(s)
- Jacob T Boyd
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Peter M LoCoco
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ashley R Furr
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michelle R Bendele
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Meilinn Tram
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Qun Li
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Fang-Mei Chang
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Madeline E Colley
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Grace M Samenuk
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Dominic A Arris
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Erin E Locke
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stephan B H Bach
- Department of Chemistry, University of Texas San Antonio, San Antonio, TX, USA
| | - Alejandro Tobon
- Department of Neurology, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Shivani B Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kenneth M Hargreaves
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
21
|
Ryu SW, Kim YO, Kim HB, Oh SB, Choi JI, Yoon MH. Antinociceptive effect of intrathecal P7C3 via GABA in a rat model of inflammatory pain. Eur J Pharmacol 2021; 899:174029. [PMID: 33727053 DOI: 10.1016/j.ejphar.2021.174029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
The recently identified molecule P7C3 has been highlighted in the field of pain research. We examined the effect of intrathecal P7C3 in tissue injury pain evoked by formalin injection and determined the role of the GABA system in the activity of P7C3 at the spinal level. Male Sprague-Dawley rats with intrathecal catheters implanted for experimental drug delivery were studied. The effects of intrathecal P7C3 and nicotinamide phosphoribosyltransferase (NAMPT) administered 10 min before the formalin injection were examined. Animals were pretreated with bicuculline, a GABA-A receptor antagonist; saclofen, a GABA-B receptor antagonist; L-allylglycine, a glutamic acid decarboxylase (GAD) blocker; and CHS 828, an NAMPT inhibitor; to observe involvement in the effects of P7C3. The effects of P7C3 alone and the mixture of P7C3 with GABA receptor antagonists on KCl-induced calcium transients were examined in rat dorsal root ganglion (DRG) neurons. The expression of GAD and the concentration of GABA in the spinal cord were evaluated. Intrathecal P7C3 and NAMPT produced an antinociceptive effect in the formalin test. Intrathecal bicuculline, saclofen, L-allylglycine, and CHS 828 reversed the antinociception of P7C3 in both phases. P7C3 decreased the KCl-induced calcium transients in DRG neurons. Both bicuculline and saclofen reversed the blocking effect of P7C3. The levels of GAD expression and GABA concentration decreased after formalin injection and were increased by P7C3. These results suggest that P7C3 increases GAD activity and then increases the GABA concentration in the spinal cord, which in turn may act on GABA receptors causing the antinociceptive effect against pain evoked by formalin injection.
Collapse
Affiliation(s)
- Sang Wan Ryu
- Department of Anesthesiology and Pain Medicine, Chonnam National University, Medical School, Gwangju, Republic of Korea
| | - Yeo Ok Kim
- Department of Anesthesiology and Pain Medicine, Chonnam National University, Medical School, Gwangju, Republic of Korea
| | - Han-Byul Kim
- Department of Neurobiology and Physiology, School of Dentistry Seoul National University, Seoul, Republic of Korea; Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seog Bae Oh
- Department of Neurobiology and Physiology, School of Dentistry Seoul National University, Seoul, Republic of Korea; Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jeong Il Choi
- Department of Anesthesiology and Pain Medicine, Chonnam National University, Medical School, Gwangju, Republic of Korea; The Brain Korea 21 Project, Center for Biomedical Human Resources at Chonnam National University, Gwangju, Republic of Korea
| | - Myung Ha Yoon
- Department of Anesthesiology and Pain Medicine, Chonnam National University, Medical School, Gwangju, Republic of Korea; The Brain Korea 21 Project, Center for Biomedical Human Resources at Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
22
|
Shin MK, Vázquez-Rosa E, Koh Y, Dhar M, Chaubey K, Cintrón-Pérez CJ, Barker S, Miller E, Franke K, Noterman MF, Seth D, Allen RS, Motz CT, Rao SR, Skelton LA, Pardue MT, Fliesler SJ, Wang C, Tracy TE, Gan L, Liebl DJ, Savarraj JPJ, Torres GL, Ahnstedt H, McCullough LD, Kitagawa RS, Choi HA, Zhang P, Hou Y, Chiang CW, Li L, Ortiz F, Kilgore JA, Williams NS, Whitehair VC, Gefen T, Flanagan ME, Stamler JS, Jain MK, Kraus A, Cheng F, Reynolds JD, Pieper AA. Reducing acetylated tau is neuroprotective in brain injury. Cell 2021; 184:2715-2732.e23. [PMID: 33852912 PMCID: PMC8491234 DOI: 10.1016/j.cell.2021.03.032] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/21/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Traumatic brain injury (TBI) is the largest non-genetic, non-aging related risk factor for Alzheimer's disease (AD). We report here that TBI induces tau acetylation (ac-tau) at sites acetylated also in human AD brain. This is mediated by S-nitrosylated-GAPDH, which simultaneously inactivates Sirtuin1 deacetylase and activates p300/CBP acetyltransferase, increasing neuronal ac-tau. Subsequent tau mislocalization causes neurodegeneration and neurobehavioral impairment, and ac-tau accumulates in the blood. Blocking GAPDH S-nitrosylation, inhibiting p300/CBP, or stimulating Sirtuin1 all protect mice from neurodegeneration, neurobehavioral impairment, and blood and brain accumulation of ac-tau after TBI. Ac-tau is thus a therapeutic target and potential blood biomarker of TBI that may represent pathologic convergence between TBI and AD. Increased ac-tau in human AD brain is further augmented in AD patients with history of TBI, and patients receiving the p300/CBP inhibitors salsalate or diflunisal exhibit decreased incidence of AD and clinically diagnosed TBI.
Collapse
Affiliation(s)
- Min-Kyoo Shin
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Edwin Vázquez-Rosa
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yeojung Koh
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Matasha Dhar
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kalyani Chaubey
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Coral J Cintrón-Pérez
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah Barker
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Emiko Miller
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kathryn Franke
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Maria F Noterman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Divya Seth
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Sriganesh Ramachandra Rao
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Lara A Skelton
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jude P J Savarraj
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Glenda L Torres
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hilda Ahnstedt
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan S Kitagawa
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Alex Choi
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pengyue Zhang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chien-Wei Chiang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Francisco Ortiz
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Kilgore
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Victoria C Whitehair
- MetroHealth Rehabilitation Institute, The MetroHealth System, Cleveland, OH; Department of Physical Medicine and Rehabilitation, Case Western Reserve University, School of Medicine, Cleveland, OH USA
| | - Tamar Gefen
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Jonathan S Stamler
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mukesh K Jain
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Allison Kraus
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - James D Reynolds
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Departments of Anesthesiology & Perioperative Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, New York, NY, USA; Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
23
|
Nicotinamide riboside relieves paclitaxel-induced peripheral neuropathy and enhances suppression of tumor growth in tumor-bearing rats. Pain 2021; 161:2364-2375. [PMID: 32433266 DOI: 10.1097/j.pain.0000000000001924] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nicotinamide riboside (NR) is a vitamin B3 precursor of NAD that blunts diabetic and chemotherapy-induced peripheral neuropathy in preclinical models. This study examined whether NR also blunts the loss of intraepidermal nerve fibers induced by paclitaxel, which is associated with peripheral neuropathy. The work was conducted in female rats with N-methyl-nitrosourea (MNU)-induced tumors of the mammary gland to increase its translational relevance, and to assess the interaction of NR with paclitaxel and NR's effect on tumor growth. Once daily oral administration of 200 mg/kg NR p.o. beginning with the first of 3 i.v. injections of 6.6 mg/kg paclitaxel to tumor-bearing rats significantly decreased paclitaxel-induced hypersensitivity to tactile and cool stimuli, as well as place-escape avoidance behaviors. It also blunted the loss of intraepidermal nerve fibers in tumor-bearing rats, as well as a separate cohort of tumor-naive rats. Unexpectedly, concomitant administration of NR during paclitaxel treatment further decreased tumor growth; thereafter, tumor growth resumed at the same rate as vehicle-treated controls. Administration of NR also decreased the percentage of Ki67-positive tumor cells in these rats. Once daily administration of NR did not seem to alter tumor growth or the percentage of Ki67-positive tumor cells in rats that were not treated with paclitaxel and followed for 3 months. These results further support the ability of NR to play a protective role after nerve injury. They also suggest that NR may not only alleviate peripheral neuropathy in patients receiving taxane chemotherapy, but also offer an added benefit by possibly enhancing its tumor-suppressing effects.
Collapse
|
24
|
Yamamoto S, Egashira N. Pathological Mechanisms of Bortezomib-Induced Peripheral Neuropathy. Int J Mol Sci 2021; 22:ijms22020888. [PMID: 33477371 PMCID: PMC7830235 DOI: 10.3390/ijms22020888] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Bortezomib, a first-generation proteasome inhibitor widely used in chemotherapy for hematologic malignancy, has effective anti-cancer activity but often causes severe peripheral neuropathy. Although bortezomib-induced peripheral neuropathy (BIPN) is a dose-limiting toxicity, there are no recommended therapeutics for its prevention or treatment. One of the most critical problems is a lack of knowledge about pathological mechanisms of BIPN. Here, we summarize the known mechanisms of BIPN based on preclinical evidence, including morphological abnormalities, involvement of non-neuronal cells, oxidative stress, and alterations of transcriptional programs in both the peripheral and central nervous systems. Moreover, we describe the necessity of advancing studies that identify the potential efficacy of approved drugs on the basis of pathological mechanisms, as this is a convincing strategy for rapid translation to patients with cancer and BIPN.
Collapse
Affiliation(s)
- Shota Yamamoto
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo 162-8655, Japan;
| | - Nobuaki Egashira
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan
- Correspondence: ; Tel.: +81-92-642-5920
| |
Collapse
|
25
|
Yamamoto S, Egashira N. Drug Repositioning for the Prevention and Treatment of Chemotherapy-Induced Peripheral Neuropathy: A Mechanism- and Screening-Based Strategy. Front Pharmacol 2021; 11:607780. [PMID: 33519471 PMCID: PMC7840493 DOI: 10.3389/fphar.2020.607780] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a severe adverse effect observed in most patients treated with neurotoxic anti-cancer drugs. Currently, there are no therapeutic options available for the prevention of CIPN. Furthermore, few drugs are recommended for the treatment of existing neuropathies because the mechanisms of CIPN remain unclear. Each chemotherapeutic drug induces neuropathy by distinct mechanisms, and thus we need to understand the characteristics of CIPN specific to individual drugs. Here, we review the known pathogenic mechanisms of oxaliplatin- and paclitaxel-induced CIPN, highlighting recent findings. Cancer chemotherapy is performed in a planned manner; therefore, preventive strategies can be planned for CIPN. Drug repositioning studies, which identify the unexpected actions of already approved drugs, have increased in recent years. We have also focused on drug repositioning studies, especially for prevention, because they should be rapidly translated to patients suffering from CIPN.
Collapse
Affiliation(s)
- Shota Yamamoto
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
| | - Nobuaki Egashira
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
26
|
闫 芳, 陈 东, 谢 敬, 曾 维, 李 强. [Escin alleviates chemotherapy-induced peripheral neuropathic pain by inducing autophagy in the spinal cord of rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1634-1638. [PMID: 33243746 PMCID: PMC7704370 DOI: 10.12122/j.issn.1673-4254.2020.11.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To investigate the effect of escin in relieving chemotherapy-induced peripheral neuropathic pain in rats and explore and the underlying mechanism. METHODS Eighteen SD rats were randomly divided into 3 groups (n=6), including an escin preconditioning group (treated with 4 mg/kg escin on days 1-7 and then with 2 mg/kg taxol on days 8, 10, 12, and 14), an escin postconditioning group (treated with 2 mg/kg taxol on days 1, 3, 5, and 7 and then with 4 mg/mg escin on days 8-14) and control group (treated with 2 mg/kg taxol on days 1, 3, 5, and 7 and then with saline on days 8-14). Mechanical allodynia and thermal hyperalgesia of the mice were tested on days 4, 7, 10 and 14, and the expression levels of LC3II and p62 in the spinal cord of the rats were detected using Western blotting. RESULTS The rats in both the escin preconditioning group and escin postconditioning group showed obviously increased thresholds of mechanical allodynia and thermal hyperalgesia as compared with those in the control group (P < 0.01). Western blotting showed that the expression level of LC3II was significantly increased while p62 expression was lowered in escin preconditioning group as compared with those in the control group (P < 0.05). The escin postconditioning group also showed significantly higher LC3II expression and lower p62 expression levels than the control group (P < 0.05). CONCLUSIONS Escin can alleviate chemotherapy-induced peripheral neuropathic pain in rats possibly by upregulating the expressions of autophagy-related proteins in the spinal cord.
Collapse
Affiliation(s)
- 芳 闫
- />中山大学肿瘤防治中心麻醉科//华南肿瘤学国家重点实验室/肿瘤医学协同创新中心,广东 广州 510060Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - 东泰 陈
- />中山大学肿瘤防治中心麻醉科//华南肿瘤学国家重点实验室/肿瘤医学协同创新中心,广东 广州 510060Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - 敬敦 谢
- />中山大学肿瘤防治中心麻醉科//华南肿瘤学国家重点实验室/肿瘤医学协同创新中心,广东 广州 510060Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - 维安 曾
- />中山大学肿瘤防治中心麻醉科//华南肿瘤学国家重点实验室/肿瘤医学协同创新中心,广东 广州 510060Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - 强 李
- />中山大学肿瘤防治中心麻醉科//华南肿瘤学国家重点实验室/肿瘤医学协同创新中心,广东 广州 510060Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
27
|
Robbins J, Busquets O, Tong M, de la Monte SM. Dysregulation of Insulin-Linked Metabolic Pathways in Alzheimer's Disease: Co-Factor Role of Apolipoprotein E ɛ4. J Alzheimers Dis Rep 2020; 4:479-493. [PMID: 33344887 PMCID: PMC7739986 DOI: 10.3233/adr-200238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Brain insulin resistance and deficiency are well-recognized abnormalities in Alzheimer's disease (AD) and likely mediators of impaired energy metabolism. Since apolipoprotein E (APOE) is a major risk factor for late-onset AD, it was of interest to examine its potential contribution to altered insulin-linked signaling networks in the brain. OBJECTIVE The main goal was to evaluate the independent and interactive contributions of AD severity and APOE ɛ4 dose on brain expression of insulin-related polypeptides and inflammatory mediators of metabolic dysfunction. METHODS Postmortem fresh frozen frontal lobe tissue from banked cases with known APOE genotypes and different AD Braak stages were used to measure insulin network polypeptide immunoreactivity with a commercial multiplex enzyme-linked immunosorbent assay (ELISA). RESULTS Significant AD Braak stage and APOE genotype-related abnormalities in insulin, C-peptide, gastric inhibitory polypeptide (GIP), glucaton-like peptide-1 (GLP-1), leptin, ghrelin, glucagon, resistin, and plasminogen activator inhibitor-1 (PAI-1) were detected. The main factors inhibiting polypeptide expression and promoting neuro-inflammatory responses included AD Braak stage and APOE ɛ4/ɛ4 rather than ɛ3/ɛ4. CONCLUSION This study demonstrates an expanded role for impaired expression of insulin-related network polypeptides as well as neuroinflammatory mediators of brain insulin resistance in AD pathogenesis and progression. In addition, the findings show that APOE has independent and additive effects on these aberrations in brain polypeptide expression, but the impact is decidedly greater for APOE ɛ4/ɛ4 than ɛ3/ɛ4.
Collapse
Affiliation(s)
- James Robbins
- Alpert Medical School of Brown University, Providence, RI, USA
| | - Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Ming Tong
- Alpert Medical School of Brown University, Providence, RI, USA,Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Suzanne M. de la Monte
- Alpert Medical School of Brown University, Providence, RI, USA,Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Departments of Pathology and Laboratory Medicine Providence VA Medical Center, Rhode Island Hospital, and the Women and Infants Hospital of Rhode Island, Providence, RI, USA,Correspondence to: Dr. Suzanne M. de la Monte, MD, MPH, Rhode Island Hospital, 55 Claverick Street, Room 419, Providence, RI 02903, USA. Tel.: +1 401 444 7364; Fax: +1 401 444 2939; E-mail:
| |
Collapse
|
28
|
Mecklenburg J, Zou Y, Wangzhou A, Garcia D, Lai Z, Tumanov AV, Dussor G, Price TJ, Akopian AN. Transcriptomic sex differences in sensory neuronal populations of mice. Sci Rep 2020; 10:15278. [PMID: 32943709 PMCID: PMC7499251 DOI: 10.1038/s41598-020-72285-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
Many chronic pain conditions show sex differences in their epidemiology. This could be attributed to sex-dependent differential expression of genes (DEGs) involved in nociceptive pathways, including sensory neurons. This study aimed to identify sex-dependent DEGs in estrous female versus male sensory neurons, which were prepared by using different approaches and ganglion types. RNA-seq on non-purified sensory neuronal preparations, such as whole dorsal root ganglion (DRG) and hindpaw tissues, revealed only a few sex-dependent DEGs. Sensory neuron purification increased numbers of sex-dependent DEGs. These DEG sets were substantially influenced by preparation approaches and ganglion types [DRG vs trigeminal ganglia (TG)]. Percoll-gradient enriched DRG and TG neuronal fractions produced distinct sex-dependent DEG groups. We next isolated a subset of sensory neurons by sorting DRG neurons back-labeled from paw and thigh muscle. These neurons have a unique sex-dependent DEG set, yet there is similarity in biological processes linked to these different groups of sex-dependent DEGs. Female-predominant DEGs in sensory neurons relate to inflammatory, synaptic transmission and extracellular matrix reorganization processes that could exacerbate neuro-inflammation severity, especially in TG. Male-selective DEGs were linked to oxidative phosphorylation and protein/molecule metabolism and production. Our findings catalog preparation-dependent sex differences in neuronal gene expressions in sensory ganglia.
Collapse
Affiliation(s)
- Jennifer Mecklenburg
- Department of Endodontics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Dawn Garcia
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- Departments of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA.
- Department of Pharmacology, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
29
|
Two Antagonistic Microtubule Targeting Drugs Act Synergistically to Kill Cancer Cells. Cancers (Basel) 2020; 12:cancers12082196. [PMID: 32781579 PMCID: PMC7463452 DOI: 10.3390/cancers12082196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Paclitaxel is a microtubule stabilizing agent and a successful drug for cancer chemotherapy inducing, however, adverse effects. To reduce the effective dose of paclitaxel, we searched for pharmaceutics which could potentiate its therapeutic effect. We screened a chemical library and selected Carba1, a carbazole, which exerts synergistic cytotoxic effects on tumor cells grown in vitro, when co-administrated with a low dose of paclitaxel. Carba1 targets the colchicine binding-site of tubulin and is a microtubule-destabilizing agent. Catastrophe induction by Carba1 promotes paclitaxel binding to microtubule ends, providing a mechanistic explanation of the observed synergy. The synergistic effect of Carba1 with paclitaxel on tumor cell viability was also observed in vivo in xenografted mice. Thus, a new mechanism favoring paclitaxel binding to dynamic microtubules can be transposed to in vivo mouse cancer treatments, paving the way for new therapeutic strategies combining low doses of microtubule targeting agents with opposite mechanisms of action.
Collapse
|
30
|
Hua X, Sun D, Zhang W, Fu J, Tong J, Sun S, Zeng F, Ouyang S, Zhang G, Wang S, Li D, Miao C, Wang P. P7C3‐A20 alleviates fatty liver by shaping gut microbiota and inducing FGF21/FGF1, via the AMP‐activated protein kinase/CREB regulated transcription coactivator 2 pathway. Br J Pharmacol 2020; 178:2111-2130. [PMID: 32037512 DOI: 10.1111/bph.15008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/30/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Affiliation(s)
- Xia Hua
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| | - Di‐Yang Sun
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| | - Wen‐Jie Zhang
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| | - Jiang‐Tao Fu
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| | - Jie Tong
- Department of Pharmacy Shanghai Tenth People's Hospital affiliated to School of Medicine, Tongji University Shanghai China
| | - Si‐Jia Sun
- Department of Pharmacy Shanghai Tenth People's Hospital affiliated to School of Medicine, Tongji University Shanghai China
| | - Fei‐Yan Zeng
- Department of Pharmacy Shanghai Tenth People's Hospital affiliated to School of Medicine, Tongji University Shanghai China
| | - Shen‐Xi Ouyang
- Department of Pharmacy Shanghai Tenth People's Hospital affiliated to School of Medicine, Tongji University Shanghai China
| | - Guo‐Yan Zhang
- Department of Pharmacy Shanghai Tenth People's Hospital affiliated to School of Medicine, Tongji University Shanghai China
| | - Shu‐Na Wang
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| | - Dong‐Jie Li
- Department of Pharmacy Shanghai Tenth People's Hospital affiliated to School of Medicine, Tongji University Shanghai China
| | - Chao‐Yu Miao
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| | - Pei Wang
- Department of Pharmacology, School of Pharmacy Second Military Medical University/Naval Medical University Shanghai China
| |
Collapse
|
31
|
Xing S, Hu Y, Huang X, Shen D, Chen C. Nicotinamide phosphoribosyltransferase‑related signaling pathway in early Alzheimer's disease mouse models. Mol Med Rep 2019; 20:5163-5171. [PMID: 31702813 PMCID: PMC6854586 DOI: 10.3892/mmr.2019.10782] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/02/2019] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease of the central nervous system that is characterized by progressive cognitive dysfunction and which ultimately leads to dementia. Studies have shown that energy dysmetabolism contributes significantly to the pathogenesis of a variety of aging-associated diseases and degenerative diseases of the nervous system, including AD. One focus of research thus has been how to regulate the expression of nicotinamide phosphoribosyltransferase (NAMPT) to prevent against neurodegenerative diseases. Therefore, the present study used 6-month-old APPswe/PS1ΔE9 (APP/PS1) transgenic mice as early AD mouse models and sought to evaluate nicotinamide adenine dinucleotide (NAD+) and FK866 (a NAMPT inhibitor) treatment in APP/PS1 mice to study NAMPT dysmetabolism in the process of AD and elucidate the underlying mechanisms. As a result of this treatment, the expression of NAMPT decreased, the synthesis of ATP and NAD+ became insufficient and the NAD+/NADH ratio was reduced. The administration of NAD+ alleviated the spatial learning and memory of APP/PS1 mice and reduced senile plaques. Administration of NAD+ may also increase the expression of the key protein NAMPT and its related protein sirtuin 1 as well as the synthesis of NAD+. Therefore, increasing NAMPT expression levels may promote NAD+ production. Their regulation could form the basis for a new therapeutic strategy.
Collapse
Affiliation(s)
- Sanli Xing
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Yiran Hu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Xujiao Huang
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Dingzhu Shen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| |
Collapse
|
32
|
Ludman T, Melemedjian OK. Bortezomib-induced aerobic glycolysis contributes to chemotherapy-induced painful peripheral neuropathy. Mol Pain 2019; 15:1744806919837429. [PMID: 30810076 PMCID: PMC6452581 DOI: 10.1177/1744806919837429] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Chemotherapy-induced painful peripheral neuropathy (CIPN) is the most common toxicity associated with widely used chemotherapeutics. CIPN is the major cause of dose reduction or discontinuation of otherwise life-saving treatment. Unfortunately, CIPN can persist in cancer survivors, which adversely affects their quality of life. Moreover, available treatments are vastly inadequate, warranting a better understanding of the biochemical and metabolic mechanisms that occur in response to chemotherapeutics which would be critical for the development of novel therapies for CIPN. Using extracellular flux analysis, this study demonstrated that the proteasome inhibitor, bortezomib, enhanced glycolysis while suppressing oxidative phosphorylation in the sensory neurons of mice. This metabolic phenotype is known as aerobic glycolysis. Bortezomib upregulated lactate dehydrogenase A and pyruvate dehydrogenase kinase 1, which consequently enhanced the production of lactate and repressed pyruvate oxidation, respectively. Moreover, lactate dehydrogenase A- and pyruvate dehydrogenase kinase 1-driven aerobic glycolysis was associated with increased extracellular acidification, augmented calcium responses, and pain in bortezomib-induced CIPN. Remarkably, pharmacological blockade and in vivo knockdown of lactate dehydrogenase A or pyruvate dehydrogenase kinase 1 reversed the metabolic phenotype, attenuated calcium responses, and alleviated pain induced by bortezomib. Collectively, these results elucidate the mechanisms by which bortezomib induces aerobic glycolysis. Moreover, these findings establish aerobic glycolysis as a metabolic phenotype that underpins bortezomib-induced CIPN.
Collapse
Affiliation(s)
- Taylor Ludman
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, USA
| | - Ohannes K Melemedjian
- 1 Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, USA.,2 Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, USA
| |
Collapse
|
33
|
Hikosaka K, Yaku K, Okabe K, Nakagawa T. Implications of NAD metabolism in pathophysiology and therapeutics for neurodegenerative diseases. Nutr Neurosci 2019; 24:371-383. [PMID: 31280708 DOI: 10.1080/1028415x.2019.1637504] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential coenzyme that mediates various redox reactions. Particularly, mitochondrial NAD plays a critical role in energy production pathways, including the tricarboxylic acid (TCA) cycle, fatty acid oxidation, and oxidative phosphorylation. NAD also serves as a substrate for ADP-ribosylation and deacetylation by poly(ADP-ribose) polymerases (PARPs) and sirtuins, respectively. Thus, NAD regulates energy metabolism, DNA damage repair, gene expression, and stress response. Numerous studies have demonstrated the involvement of NAD metabolism in neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and retinal degenerative diseases. Mitochondrial dysfunction is considered crucial pathogenesis for neurodegenerative diseases such as AD and PD. Maintaining appropriate NAD levels is important for mitochondrial function. Indeed, decreased NAD levels are observed in AD and PD, and supplementation of NAD precursors ameliorates disease phenotypes by activating mitochondrial functions. NAD metabolism also plays an important role in axonal degeneration, a characteristic feature of peripheral neuropathy and neurodegenerative diseases. In addition, dysregulated NAD metabolism is implicated in retinal degenerative diseases such as glaucoma and Leber congenital amaurosis, and NAD metabolism is considered a therapeutic target for these diseases. In this review, we summarize the involvement of NAD metabolism in axon degeneration and various neurodegenerative diseases and discuss perspectives of nutritional intervention using NAD precursors.
Collapse
Affiliation(s)
- Keisuke Hikosaka
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Keisuke Yaku
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Keisuke Okabe
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan.,First Department of Internal Medicine, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan.,Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
34
|
Trecarichi A, Flatters SJL. Mitochondrial dysfunction in the pathogenesis of chemotherapy-induced peripheral neuropathy. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 145:83-126. [PMID: 31208528 DOI: 10.1016/bs.irn.2019.05.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Several first-line chemotherapeutic agents, including taxanes, platinum agents and proteasome inhibitors, are associated with the dose-limiting side effect of chemotherapy-induced peripheral neuropathy (CIPN). CIPN predominantly manifests as sensory symptoms, which are likely due to drug accumulation within peripheral nervous tissues rather than the central nervous system. No treatment is currently available to prevent or reverse CIPN. The causal mechanisms underlying CIPN are not yet fully understood. Mitochondrial dysfunction has emerged as a major factor contributing to the development and maintenance of CIPN. This chapter will provide an overview of both clinical and preclinical data supporting this hypothesis. We will review the studies reporting the nature of mitochondrial dysfunction evoked by chemotherapy in terms of changes in mitochondrial morphology, bioenergetics and reactive oxygen species (ROS) generation. Furthermore, we will discuss the in vivo effects of pharmacological interventions that counteract chemotherapy-evoked mitochondrial dysfunction and ameliorate pain-like behavior.
Collapse
Affiliation(s)
- Annalisa Trecarichi
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Sarah J L Flatters
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
35
|
Wang SN, Miao CY. Targeting NAMPT as a therapeutic strategy against stroke. Stroke Vasc Neurol 2019; 4:83-89. [PMID: 31338216 PMCID: PMC6613878 DOI: 10.1136/svn-2018-000199] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/21/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022] Open
Abstract
Stroke is the second and the leading most common cause of death in the world and China, respectively, but with few effective therapies. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme for nicotinamide adenine dinucleotide (NAD) salvage synthesis in mammals, thereby influencing NAD-dependent enzymes and constituting a strong endogenous defence system against various stresses. Accumulating in-vitro and in-vivo studies have demonstrated the neuroprotective effect of NAMPT in stroke. Here, we review the direct evidence of NAMPT as a promising target against stroke from five potential therapeutic strategies, including NAMPT overexpression, recombinant NAMPT, NAMPT activators, NAMPT enzymatic product nicotinamide mononucleotide (NMN), and NMN precursors nicotinamide riboside and nicotinamide, and describe the relevant mechanisms and limitations, providing a promising choice for developing novel and effective therapeutic interventions against ischaemic and haemorrhagic stroke.
Collapse
Affiliation(s)
- Shu-Na Wang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| |
Collapse
|
36
|
de la Monte SM, Tong M, Daiello LA, Ott BR. Early-Stage Alzheimer's Disease Is Associated with Simultaneous Systemic and Central Nervous System Dysregulation of Insulin-Linked Metabolic Pathways. J Alzheimers Dis 2019; 68:657-668. [PMID: 30775986 PMCID: PMC10084886 DOI: 10.3233/jad-180906] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Brain insulin resistance is a well-recognized abnormality in Alzheimer's disease (AD) and the likely mediator of impaired glucose utilization that emerges early and progresses with disease severity. Moreover, the rates of mild cognitive impairment (MCI) or AD are significantly greater in people with diabetes mellitus or obesity. OBJECTIVE This study was designed to determine whether systemic and central nervous system (CNS) insulin resistant disease states emerge together and thus may be integrally related. METHODS Insulin-related molecules were measured in paired human serum and cerebrospinal fluid (CSF) samples from 19 with MCI or early AD, and 21 controls using a multiplex ELISA platform. RESULTS In MCI/AD, both the CSF and serum samples had significantly elevated mean levels of C-peptide and an incretin, and reduced expression of Visfatin, whereas only CSF showed significant reductions in insulin and leptin and only serum had increased glucagon, PAI-1, and ghrelin. Although the overall CSF and serum responses reflected insulin resistance together with insulin deficiency, the specific alterations measured in CSF and serum were different. CONCLUSION In MCI and early-stage AD, CNS and systemic insulin-related metabolic dysfunctions, including insulin resistance, occur simultaneously, suggesting that they are integrally related and possibly mediated similar pathogenic factors.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology and Laboratory Medicine (Neuropathology), Rhode Island Hospital, the Providence VA Medical Center, and the Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Lori A Daiello
- Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,The Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Brian R Ott
- Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,The Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
37
|
Pieper AA, McKnight SL. Benefits of Enhancing Nicotinamide Adenine Dinucleotide Levels in Damaged or Diseased Nerve Cells. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:207-217. [PMID: 30787047 DOI: 10.1101/sqb.2018.83.037622] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Three unbiased lines of research have commonly pointed to the benefits of enhanced levels of nicotinamide adenine dinucleotide (NAD+) to diseased or damaged neurons. Mice carrying a triplication of the gene encoding the culminating enzyme in NAD+ salvage from nicotinamide, NMNAT, are protected from a variety of insults to axons. Protection from Wallerian degeneration of axons is also observed in flies and mice bearing inactivating mutations in the SARM1 gene. Functional studies of the SARM1 gene product have revealed the presence of an enzymatic activity directed toward the hydrolysis of NAD+ Finally, an unbiased drug screen performed in living mice led to the discovery of a neuroprotective chemical designated P7C3. Biochemical studies of the P7C3 chemical show that it can enhance recovery of NAD+ from nicotinamide by activating NAMPT, the first enzyme in the salvage pathway. In combination, these three unrelated research endeavors offer evidence of the benefits of enhanced NAD+ levels to damaged neurons.
Collapse
Affiliation(s)
- Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, Ohio 44106, USA
| | - Steven L McKnight
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
38
|
Wang YH, Liou KT, Tsai KC, Liu HK, Yang LM, Chern CM, Shen YC. GSK-3 inhibition through GLP-1R allosteric activation mediates the neurogenesis promoting effect of P7C3 after cerebral ischemic/reperfusional injury in mice. Toxicol Appl Pharmacol 2018; 357:88-105. [PMID: 30189238 DOI: 10.1016/j.taap.2018.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/22/2022]
Abstract
An aminopropyl carbazole compound, P7C3, has been shown to be a potent neurogenesis promoting agent; however, its fundamental signaling action has yet to be elucidated. A cerebral ischemic/reperfusional (CI/R) injury model in mice was implemented to elucidate the neuronal protective mechanism(s) of P7C3. Treating CI/R mice using P7C3 (50-100 μg/kg, i.v.) significantly improved tracking distance and walking behavior, and reduced brain damage. Specifically, P7C3 promoted the expression of neurogenesis-associated proteins, including doublecortin, beta tubulin III (β-tub3), adam11 and adamts20, near the peri-infarct cortex, accompanied by glycogen synthase kinase 3 (GSK-3) inhibition and β-catenin upregulation. The application of a specific inhibitor against glucagon-like peptide 1 receptor (GLP-1R), exendin(9-39), revealed that the beneficial effects of P7C3 involved triggering the activation of GLP-1R-associated PKA/Akt signaling. P7C3 elicited the GLP-1R-dependent intracellular cAMP increment and the insulin secretion in cellular models. Surface plasmon resonance assay of P7C3 showed a Kd value of 0.53 μM for GLP-1R binding, and the docking of P7C3 to the putative active site on GLP-1R was successfully predicted by molecular modeling. Our findings indicate that P7C3 promotes the expression of neurogenesis proteins by activation of the cAMP/PKA-dependent and Akt/GSK3-associated β-catenin through positive allosteric stimulation of GLP-1R. Within the P7C3 class of neuroprotective molecules, this mechanism appears to be unique to the prototypical P7C3 molecule, as other active derivatives such as P7C2-A20 and P7C3-S243 they do not engage this same pathway and have been shown to work by nicotinamide phosphoribosyltransferase (NAMPT) stimulation.
Collapse
Affiliation(s)
- Yea-Hwey Wang
- National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Kuo-Tong Liou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Chinese Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; Ph.D. Program for Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan
| | - Hui-Kang Liu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Li-Ming Yang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; School of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Chang-Ming Chern
- Division of Neurovascular Disease, Neurological Institute, Taipei Veterans General Hospital, Taiwan; Taipei Municipal Gan-Dau Hospital, Taiwan; Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei City, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan; Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan.
| |
Collapse
|
39
|
Bauman MD, Schumann CM, Carlson EL, Taylor SL, Vázquez-Rosa E, Cintrón-Pérez CJ, Shin MK, Williams NS, Pieper AA. Neuroprotective efficacy of P7C3 compounds in primate hippocampus. Transl Psychiatry 2018; 8:202. [PMID: 30258178 PMCID: PMC6158178 DOI: 10.1038/s41398-018-0244-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/16/2018] [Accepted: 08/03/2018] [Indexed: 01/31/2023] Open
Abstract
There is a critical need for translating basic science discoveries into new therapeutics for patients suffering from difficult to treat neuropsychiatric and neurodegenerative conditions. Previously, a target-agnostic in vivo screen in mice identified P7C3 aminopropyl carbazole as capable of enhancing the net magnitude of postnatal neurogenesis by protecting young neurons from death. Subsequently, neuroprotective efficacy of P7C3 compounds in a broad spectrum of preclinical rodent models has also been observed. An important next step in translating this work to patients is to determine whether P7C3 compounds exhibit similar efficacy in primates. Adult male rhesus monkeys received daily oral P7C3-A20 or vehicle for 38 weeks. During weeks 2-11, monkeys received weekly injection of 5'-bromo-2-deoxyuridine (BrdU) to label newborn cells, the majority of which would normally die over the following 27 weeks. BrdU+ cells were quantified using unbiased stereology. Separately in mice, the proneurogenic efficacy of P7C3-A20 was compared to that of NSI-189, a proneurogenic drug currently in clinical trials for patients with major depression. Orally-administered P7C3-A20 provided sustained plasma exposure, was well-tolerated, and elevated the survival of hippocampal BrdU+ cells in nonhuman primates without adverse central or peripheral tissue effects. In mice, NSI-189 was shown to be pro-proliferative, and P7C3-A20 elevated the net magnitude of hippocampal neurogenesis to a greater degree than NSI-189 through its distinct mechanism of promoting neuronal survival. This pilot study provides evidence that P7C3-A20 safely protects neurons in nonhuman primates, suggesting that the neuroprotective efficacy of P7C3 compounds is likely to translate to humans as well.
Collapse
Affiliation(s)
- Melissa D. Bauman
- 0000 0004 1936 9684grid.27860.3bDepartment of Psychiatry and Behavioral Sciences, University of California, Davis, USA ,0000 0004 1936 9684grid.27860.3bUC Davis MIND Institute, University of California, Davis, USA ,0000 0004 1936 9684grid.27860.3bCalifornia National Primate Research Center, Davis, USA ,0000 0004 1936 9684grid.27860.3bDepartment of Public Health Sciences, University of California, Davis, USA
| | - Cynthia M. Schumann
- 0000 0004 1936 9684grid.27860.3bDepartment of Psychiatry and Behavioral Sciences, University of California, Davis, USA ,0000 0004 1936 9684grid.27860.3bUC Davis MIND Institute, University of California, Davis, USA
| | - Erin L. Carlson
- 0000 0004 1936 9684grid.27860.3bDepartment of Psychiatry and Behavioral Sciences, University of California, Davis, USA
| | - Sandra L. Taylor
- 0000 0004 1936 9684grid.27860.3bDepartment of Public Health Sciences, University of California, Davis, USA
| | - Edwin Vázquez-Rosa
- University Hospital Case Medical Center; Department of Psychiatry Case Western Reserve University; Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Harrington Discovery Institute, Cleveland, OH 44106 USA
| | - Coral J. Cintrón-Pérez
- University Hospital Case Medical Center; Department of Psychiatry Case Western Reserve University; Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Harrington Discovery Institute, Cleveland, OH 44106 USA
| | - Min-Kyoo Shin
- University Hospital Case Medical Center; Department of Psychiatry Case Western Reserve University; Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Harrington Discovery Institute, Cleveland, OH 44106 USA
| | - Noelle S. Williams
- UT Southwestern Medical Center, Department of Biochemistry, Dallas, TX USA
| | - Andrew A. Pieper
- University Hospital Case Medical Center; Department of Psychiatry Case Western Reserve University; Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Harrington Discovery Institute, Cleveland, OH 44106 USA
| |
Collapse
|
40
|
Blaya MO, Wasserman JM, Pieper AA, Sick TJ, Bramlett HM, Dietrich WD. Neurotherapeutic capacity of P7C3 agents for the treatment of Traumatic Brain Injury. Neuropharmacology 2018; 145:268-282. [PMID: 30236963 DOI: 10.1016/j.neuropharm.2018.09.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 09/07/2018] [Accepted: 09/14/2018] [Indexed: 12/25/2022]
Abstract
Traumatic brain injury (TBI) is a significant public health problem around the world. A promising area of research is the characterization of small, drug-like molecules that have potent clinical properties. One pharmacotherapeutic agent in particular, an aminopropyl carbazole called P7C3, was discovered using an in vivo screen to identify new agents that augmented the net magnitude of adult hippocampal neurogenesis. P7C3 greatly enhanced neurogenesis by virtue of increasing survival rates of immature neurons. The potent neuroprotective efficacy of P7C3 is likely due to enhanced nicotinamide phosphoribosyltransferase (NAMPT) activity, which supports critical cellular processes. The scaffold of P7C3 was found to have favorable pharmacokinetic properties, good bioavailability, and was nontoxic. Preclinical studies have shown that administration of the P7C3-series of neuroprotective compounds after TBI can rescue and reverse detrimental cellular events leading to improved functional recovery. In several TBI models and across multiple species, P7C3 and its analogues have produced significant neuroprotection, axonal preservation, robust increases in the net magnitude of adult neurogenesis, protection from injury-induced LTP deficits, and improvement in neurological functioning. This review will elucidate the exciting and diverse therapeutic findings of P7C3 administration in the presence of a complex and multifactorial set of cellular and molecular challenges brought forth by experimental TBI. The clinical potential and broad therapeutic applicability of P7C3 warrants much needed investigation into whether these remedial effects can be replicated in the clinic. P7C3 may serve as an important step forward in the design, understanding, and implementation of pharmacotherapies for treating patients with TBI. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Meghan O Blaya
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Joseph M Wasserman
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospital Case Medical Center, Department of Psychiatry Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, OH, 44106, USA
| | - Thomas J Sick
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Helen M Bramlett
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, 33125, USA
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|