1
|
Koh Y, Vázquez-Rosa E, Gao F, Li H, Chakraborty S, Tripathi SJ, Barker S, Bud Z, Bangalore A, Kandjoze UP, León-Alvarado RA, Sridharan PS, Cordova BA, Yu Y, Hyung J, Fang H, Singh S, Katabathula R, LaFramboise T, Kasturi L, Lutterbaugh J, Beard L, Cordova E, Cintrón-Pérez CJ, Franke K, Fragoso MF, Miller E, Indrakumar V, Noel KL, Dhar M, Ajroud K, Zamudio C, Lopes FBTP, Bambakidis E, Zhu X, Wilson B, Flanagan ME, Gefen T, Fujioka H, Fink SP, Desai AB, Dawson D, Williams NS, Kim YK, Ready JM, Paul BD, Shin MK, Markowitz SD, Pieper AA. Inhibiting 15-PGDH blocks blood-brain barrier deterioration and protects mice from Alzheimer's disease and traumatic brain injury. Proc Natl Acad Sci U S A 2025; 122:e2417224122. [PMID: 40397680 DOI: 10.1073/pnas.2417224122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 04/17/2025] [Indexed: 05/23/2025] Open
Abstract
Alzheimer's disease (AD) and traumatic brain injury (TBI) are currently untreatable neurodegenerative disorders afflicting millions of people worldwide. These conditions are pathologically related, and TBI is one of the greatest risk factors for AD. Although blood-brain barrier (BBB) disruption drives progression of both AD and TBI, strategies to preserve BBB integrity have been hindered by lack of actionable targets. Here, we identify 15-hydroxyprostaglandin dehydrogenase (15-PGDH), an enzyme that catabolizes eicosanoids and other anti-inflammatory mediators, as a therapeutic candidate that protects the BBB. We demonstrate that 15-PGDH is enriched in BBB-associated myeloid cells and becomes markedly elevated in human and mouse models of AD and TBI, as well as aging, another major risk factor for AD. Pathological increase in 15-PGDH correlates with pronounced oxidative stress, neuroinflammation, and neurodegeneration, alongside profound BBB structural degeneration characterized by astrocytic endfeet swelling and functional impairment. Pharmacologic inhibition or genetic reduction of 15-PGDH in AD and TBI models strikingly mitigates oxidative damage, suppresses neuroinflammation, and restores BBB integrity. Most notably, inhibiting 15-PGDH not only halts neurodegeneration but also preserves cognitive function at levels indistinguishable from healthy controls. Remarkably, these neuroprotective effects in AD are achieved without affecting amyloid pathology, underscoring a noncanonical mechanism for treating AD. In a murine microglia cell line exposed to amyloid beta oligomer, major protection was demonstrated by multiple anti-inflammatory substrates that 15-PGDH degrades. Thus, our findings position 15-PGDH inhibition as a broad-spectrum strategy to protect the BBB and thereby preserve brain health and cognition in AD and TBI.
Collapse
Affiliation(s)
- Yeojung Koh
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Farrah Gao
- Department of Genetics and Genome Sciences School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Hongyun Li
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Sarah Barker
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Zea Bud
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Frances Payne Bolton School of Nursing, Case Western Reserve University, Cleveland, OH 44106
| | - Anusha Bangalore
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Uapingena P Kandjoze
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Psychology, Neuroscience Program Earlham College, Richmond, IN 47374
| | - Rose A León-Alvarado
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Psychology, Neuroscience Program Earlham College, Richmond, IN 47374
| | - Preethy S Sridharan
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Brittany A Cordova
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Youngmin Yu
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606
| | - Jiwon Hyung
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Hua Fang
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Hathaway Brown School, Shaker Heights, OH 44122
- Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Salendra Singh
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44195
| | | | - Thomas LaFramboise
- Department of Genetics and Genome Sciences School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Lakshmi Kasturi
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - James Lutterbaugh
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Lydia Beard
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Erika Cordova
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Coral J Cintrón-Pérez
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Kathryn Franke
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | | | - Emiko Miller
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Vidya Indrakumar
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Kamryn L Noel
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Matasha Dhar
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Kaouther Ajroud
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Carlos Zamudio
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Filipa Blasco Tavares Pereira Lopes
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Evangeline Bambakidis
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Northwestern University Weinberg College of Arts and Sciences, Chicago, IL 60208
| | - Xiongwei Zhu
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
| | - Brigid Wilson
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Department of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH 44106
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Pathology, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 78229
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core Case, School of Medicine, Western Reserve University, Cleveland, OH 44016
| | - Stephen P Fink
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Amar B Desai
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Dawn Dawson
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Young-Kwang Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21205
- The Solomon H. Snyder Department of Neuroscience, School of Medicine Johns Hopkins University, Baltimore, MD 21205
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Min-Kyoo Shin
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sanford D Markowitz
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
- Department of Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106
| | - Andrew A Pieper
- Department of Psychiatry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals, Cleveland Medical Center, Cleveland, OH 44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Veterans Affairs Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Pathology, School of Medicine Case Western Reserve University, Cleveland, OH 44106
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
2
|
Pathania AS, Badhai K, Singh N, Chopra DS, Singh D. The neuroprotective potential of carbazole in traumatic brain injury. Brain Inj 2025; 39:359-369. [PMID: 39691969 DOI: 10.1080/02699052.2024.2441843] [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: 08/08/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE To review the neuroprotective potential of carbazole and its derivatives in the management of traumatic brain injury (TBI), focusing on their mechanisms of action, preclinical findings, and potential clinical applications. DATA SOURCES Relevant literature was identified through a comprehensive search of PubMed, Embase, Medline, Web of Science, and the Cochrane Library databases. REVIEW METHODS This review includes an analysis of studies investigating the neuroprotective properties of carbazole, emphasizing its anti-apoptotic, antioxidative, neurotrophic, and mitochondrial protective mechanisms. The scope of the review covers preclinical studies and explores future directions for clinical translation. RESULTS Carbazole compounds demonstrate significant neuroprotective effects by reducing oxidative stress, inhibiting apoptosis, and promoting neuroregeneration in preclinical models of TBI. These compounds also potentially preserve blood-brain barrier integrity and modulate inflammatory responses. However, clinical studies evaluating carbazole-based therapies in human subjects are limited, highlighting a critical gap in translational research. CONCLUSION Carbazole and its derivatives are promising therapeutic agents for mitigating secondary damage and enhancing recovery in TBI. Further research is warranted to establish their efficacy and safety in clinical settings and to explore their potential as part of combination therapies for TBI management.
Collapse
Affiliation(s)
| | - Kavita Badhai
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Dimple Sethi Chopra
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
3
|
Nishiguchi T, Yamanishi K, Patel S, Malicoat JR, Phuong NJ, Seki T, Ishii T, Aoyama B, Shimura A, Gorantla N, Yamanashi T, Iwata M, Pieper AA, Shinozaki G. Discovery of novel protective agents for infection-related delirium through bispectral electroencephalography. Transl Psychiatry 2024; 14:413. [PMID: 39358319 PMCID: PMC11447046 DOI: 10.1038/s41398-024-03130-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Delirium is a multifactorial medical condition of waxing and waning impairment across various domains of mental functioning over time. Importantly, delirium is also one of the greatest risk factors for prolonged hospitalization, morbidity, and mortality. Studying this important condition is challenging due to the difficulty in both objective diagnosis in patients and validation of laboratory models. As a result, there is a lack of protective treatments for delirium. Our recent studies report the efficacy of bispectral electroencephalography (BSEEG) in diagnosing delirium in patients and predicting patient outcomes, advancing the concept that this simple measure could represent an additional vital sign for patients. Here, we applied BSEEG to characterize and validate a novel lipopolysaccharide (LPS) mouse model of infection-related delirium. We then applied this model to evaluate the protective efficacy of three putative therapeutic agents: the conventional antipsychotic medication haloperidol, the neuroprotective compound P7C3-A20, and the antibiotic minocycline. Aged mice were more susceptible than young mice to LPS-induced aberration in BSEEG, reminiscent of the greater vulnerability of older adults to delirium. In both young and old mice, P7C3-A20 and minocycline administration prevented LPS-induced BSEEG abnormality. By contrast, haloperidol did not. P7C3-A20 and minocycline have been shown to limit different aspects of LPS toxicity, and our data offers proof of principle that these agents might help protect patients from developing infection-related delirium. Thus, utilization of BSEEG in a mouse model for infection-related delirium can identify putative therapeutic agents for applications in patient clinical trials.
Collapse
Affiliation(s)
- Tsuyoshi Nishiguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Neuropsychiatry, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Kyosuke Yamanishi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Neuropsychiatry, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Shivani Patel
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- University of California, Berkeley, CA, USA
| | - Johnny R Malicoat
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Nathan James Phuong
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tomoteru Seki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Psychiatry, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Takaya Ishii
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- iPS Cell-Based Drug Discovery Group, Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
| | - Bun Aoyama
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- Division of Anesthesiology, National Hospital Organization Kochi Hospital, Kochi, Kochi, Japan
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Nankoku, Kochi, Japan
| | - Akiyoshi Shimura
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Psychiatry, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Nipun Gorantla
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Takehiko Yamanashi
- Department of Neuropsychiatry, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Masaaki Iwata
- Department of Neuropsychiatry, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Gen Shinozaki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
4
|
Sutariya V, Bhatt P, Saini A, Miller A, Badole SL, Tur J, Gittinger M, Kim JW, Manickam R, Tipparaju SM. Development and testing of nanoparticles delivery for P7C3 small molecule using injury models. Mol Cell Biochem 2024; 479:2429-2445. [PMID: 37787834 DOI: 10.1007/s11010-023-04865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
The use of nanoparticles (NPs) has emerged as a potential tool for safe and effective drug delivery. In the present study, we developed small molecule P7C3-based NPs and tested its efficacy and toxicity along with the tissue specific aptamer-modified P7C3 NPs. The P7C3 NPs were prepared using poly (D, L-lactic-co-glycolic acid) carboxylic acid (PLGA-COOH) polymer, were conjugated with skeletal muscle-specific RNA aptamer (A01B P7C3 NPs) and characterized for its cytotoxicity, cellular uptake, and wound healing in vitro. The A01B P7C3 NPs demonstrated an encapsulation efficiency of 30.2 ± 2.6%, with the particle size 255.9 ± 4.3 nm, polydispersity index of 0.335 ± 0.05 and zeta potential of + 10.4 ± 1.8mV. The FTIR spectrum of P7C3 NPs displayed complete encapsulation of the drug in the NPs. The P7C3 NPs and A01B P7C3 NPs displayed sustained drug release in vitro for up to 6 days and qPCR analysis confirmed A01B aptamer binding to P7C3 NPs. The C2C12 cells viability assay displayed no cytotoxic effects of all 3 formulations at 48 and 72 h. In addition, the cellular uptake of A01B P7C3 NPs in C2C12 myoblasts demonstrated higher uptake. In vitro assay mimicking wound healing showed improved wound closure with P7C3 NPs. In addition, P7C3 NPs significantly decreased TNF-α induced NF-κB activity in the C2C12/NF-κB reporter cells after 24-hour treatment. The P7C3 NPs showed 3-4-fold higher efficacy compared to P7C3 solutions in both wound-closure and inflammation assays in C2C12 cells. Furthermore, the P7C3 NPs showed 3-4-fold higher efficacy in reducing the infarct size and protected mouse hearts from ex vivo ischemia-reperfusion injury. Overall, this study demonstrates the safe and effective delivery of P7C3 NPs.
Collapse
Affiliation(s)
- Vijaykumar Sutariya
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Priyanka Bhatt
- Department of Pharmaceutical Sciences, Wegmans School of Pharmacy, St. John Fisher University, Rochester, NY, 14618, USA
| | - Aren Saini
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Abraian Miller
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Sachin L Badole
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Jared Tur
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Mackenzie Gittinger
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Joung Woul Kim
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Ravikumar Manickam
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
5
|
Gao Q, Dai Z, Yang X, Liu C, Liu G. Experimental study on small molecule combinations inducing reprogramming of rat fibroblasts into functional neurons. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:498-508. [PMID: 39183062 PMCID: PMC11375488 DOI: 10.3724/zdxbyxb-2024-0007] [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: 01/07/2024] [Accepted: 04/30/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVES To establish a methodological system for reprogramming rat embryonic fibroblasts (REF) into chemically induced neurons (ciNCs) via small molecule compounds to provide safe and effective donor cells for treatment of neurodegenerative diseases. METHODS Based on the method established by PEI Gang's research group to directly reprogram human fibroblasts into neurons, the induction medium and maturation medium was optimized by replacing the coating solution, mitigating oxidative stress injury, adding neurogenic protective factors, adjusting the concentration of trichothecenes, performing small-molecule removal experiments, and carrying out immunofluorescence and Western blotting on cells at different stages of induction to validate the effect of induction. RESULTS When the original protocol was used for induction, the cell survival rate was (34.24±2.77)%. After replacing the coating solution gelatin with matrigel, the cell survival rate increased to (45.41±4.27)%; after adding melatonin, the cell survival rate increased to (67.95±5.61)% and (23.43±1.42)% were transformed into neural-like cells; after adding the small molecule P7C3-A20, the cell survival rate was further increased to (76.27±1.41)%, and (39.72±4.75)% of the cells were transformed into neural-like cells. When the concentration of trichothecene was increased to 30 μmol/L, the proportion of neural-like cells reached (55.79±1.90)%; after the removal of SP600125, (86.96±2.15)% of the cells survived, and the rate of neural-like cell production increased to (63.43±1.60)%. With the optimized protocol, REF could be successfully induced into ciNC through the neural precursor cell stage, in which the neural precursor cells were able to highly express the neural precursor cell markers SRY-related HMG-box gene 2 (Sox2) and paired box 6 (Pax6) as well as neuron-specific marker tubulin 1 (Tuj1), while the expression of fiber-associated protein vimentin was reduced. After two weeks of induction of neural precursor cells in a maturation medium, most cells displayed neuronal-like cell morphology. The induced ciNCs were able to highly express the mature neuronal surface markers Tuj1 and microtubule-associated protein 2 (MAP2), while the expression of vimentin was reduced. CONCLUSIONS The small molecule combinations optimized in this study can reprogram REF to ciNCs under normoxic conditions.
Collapse
Affiliation(s)
- Qunwei Gao
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China.
| | - Zhenjia Dai
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Xinkang Yang
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Changqing Liu
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Gaofeng Liu
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China. ,
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu 233030, Anhui Province, China. ,
| |
Collapse
|
6
|
Bommanaboina B, Roy D, Baire B. Integrated synthesis of 3,4-carbazoquinone alkaloids N-Me-carbazoquinocin A, B and D-F. RSC Adv 2024; 14:27372-27384. [PMID: 39205936 PMCID: PMC11350554 DOI: 10.1039/d4ra05347h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Carbazole alkaloids carbazoquinocin A-F possessing a 1-alkyl-2-methyl-3,4-ortho-carabazoquinone framework were isolated from the microorganism Streptomyces violaceus 2448-SVT2 in 1995. Furthermore, they were found to exhibit strong inhibitory activity against lipid peroxidation. Herein, we report the integrated synthesis of N-Me-analogues of 5 members of the carbazoquinocin family of natural products, namely, N-Me-carbazoquinocin A, B and D-F. We employed an acid-catalyzed, intramolecular benzannulation of indole-appended Z-enoate propargylic alcohols, which was developed earlier in our laboratory, for the construction of the required carbazole framework. All five natural products were obtained in an overall yield of 50-60%, starting from a commercially available indole.
Collapse
Affiliation(s)
- Basavaiah Bommanaboina
- Department of Chemistry, Indian Institute of Technology Madras Chennai-600036 Tamil Nadu India
| | - Debayan Roy
- Department of Chemistry, Indian Institute of Technology Madras Chennai-600036 Tamil Nadu India
| | - Beeraiah Baire
- Department of Chemistry, Indian Institute of Technology Madras Chennai-600036 Tamil Nadu India
| |
Collapse
|
7
|
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
|
8
|
Amin PM, Wang W, Wang C, Zhou J, Wang Y. Gold-catalyzed benzannulations of 2-alkenylindoles with alkynes: a protecting-group-free regioselective approach to carbazoles. Chem Commun (Camb) 2024; 60:5326-5329. [PMID: 38666614 DOI: 10.1039/d4cc00176a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
A gold(I)-catalyzed protecting-group-free benzannulation approach to functionalized NH-carbazoles was accomplished via the hydroarylation of alkynes with 2-alkenylindoles. A broad spectrum of terminal and internal alkynes and 2-alkenylindoles successfully participated in this annulation reaction. The protocol efficiently enabled the formation of substituted NH-carbazoles with moderate to specific regioselectivities. The synthetic utility of this protocol was demonstrated by a variety of post-functionalizations.
Collapse
Affiliation(s)
- Pathan Mosim Amin
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Key Laboratory of Sustainable Energy Materials Chemistry, Xi'an Jiaotong University (XJTU), Xi'an, 710049, China.
| | - Weilin Wang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Key Laboratory of Sustainable Energy Materials Chemistry, Xi'an Jiaotong University (XJTU), Xi'an, 710049, China.
| | - Chao Wang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Key Laboratory of Sustainable Energy Materials Chemistry, Xi'an Jiaotong University (XJTU), Xi'an, 710049, China.
| | - Junrui Zhou
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Key Laboratory of Sustainable Energy Materials Chemistry, Xi'an Jiaotong University (XJTU), Xi'an, 710049, China.
| | - Youliang Wang
- School of Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, Xi'an Key Laboratory of Sustainable Energy Materials Chemistry, Xi'an Jiaotong University (XJTU), Xi'an, 710049, China.
| |
Collapse
|
9
|
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
|
10
|
Liu SS, Ma CX, Quan ZY, Ding J, Yang L, Liu SM, Zhang HA, Qing H, Liang JH. Discovery of Novel Diphenyl Acrylonitrile Derivatives That Promote Adult Rats' Hippocampal Neurogenesis. Int J Mol Sci 2024; 25:1241. [PMID: 38279241 PMCID: PMC10816640 DOI: 10.3390/ijms25021241] [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: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024] Open
Abstract
We previously discovered WS-6 as a new antidepressant in correlation to its function of stimulating neurogenesis. Herein, several different scaffolds (stilbene, 1,3-diphenyl 1-propene, 1,3-diphenyl 2-propene, 1,2-diphenyl acrylo-1-nitrile, 1,2-diphenyl acrylo-2-nitrile, 1,3-diphenyl trimethylamine), further varied through substitutions of twelve amide substituents plus the addition of a methylene unit and an inverted amide, were examined to elucidate the SARs for promoting adult rat neurogenesis. Most of the compounds could stimulate proliferation of progenitors, but just a few chemicals possessing a specific structural profile, exemplified by diphenyl acrylonitrile 29b, 32a, and 32b, showed better activity than the clinical drug NSI-189 in promoting newborn cells differentiation into mature neurons. The most potent diphenyl acrylonitrile 32b had an excellent brain AUC to plasma AUC ratio (B/P = 1.6), suggesting its potential for further development as a new lead.
Collapse
Affiliation(s)
- Si-Si Liu
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China; (S.-S.L.); (C.-X.M.); (J.D.); (S.-M.L.)
| | - Cong-Xuan Ma
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China; (S.-S.L.); (C.-X.M.); (J.D.); (S.-M.L.)
| | - Zheng-Yang Quan
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Z.-Y.Q.); (L.Y.); (H.-A.Z.)
| | - Jing Ding
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China; (S.-S.L.); (C.-X.M.); (J.D.); (S.-M.L.)
| | - Liang Yang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Z.-Y.Q.); (L.Y.); (H.-A.Z.)
| | - Si-Meng Liu
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China; (S.-S.L.); (C.-X.M.); (J.D.); (S.-M.L.)
| | - He-Ao Zhang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Z.-Y.Q.); (L.Y.); (H.-A.Z.)
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (Z.-Y.Q.); (L.Y.); (H.-A.Z.)
| | - Jian-Hua Liang
- Key Laboratory of Medicinal Molecule Science and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China; (S.-S.L.); (C.-X.M.); (J.D.); (S.-M.L.)
| |
Collapse
|
11
|
Sankara CS, Namboothiri INN. Hauser-Kraus Annulation Initiated Multi-Cascade Reactions for Facile Access to Functionalized and Fused Oxazepines, Carbazoles and Phenanthridinediones. Chemistry 2024; 30:e202303517. [PMID: 37946675 DOI: 10.1002/chem.202303517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023]
Abstract
The Hauser-Kraus (H-K) annulation of N-unsubstituted 3-olefinic oxindoles with 3-nucleophilic phthalides triggers a cascade of ring expansion and ring contraction reactions through several regioselective steps in one pot. While oxazepines were isolated in the presence of stoichiometric amounts of base at room temperature, carbazoles and phenanthridinediones were the products in the presence of excess base and microwave irradiation. Mechanistic studies guided by stepwise reactions and control experiments revealed that the isolable oxazepine intermediate, formed via ring expansion of the H-K adduct, is the key precursor to carbazole and phenanthridinedione via decarboxylative regioselective cyclizations.
Collapse
|
12
|
Shu G, Chen W, Huang C, Shan H, Ye J, Li J, Gui Y. Higher concentration of P7C3 than required for neuroprotection suppresses renal cell carcinoma growth and metastasis. J Cancer 2024; 15:1191-1202. [PMID: 38356717 PMCID: PMC10861821 DOI: 10.7150/jca.90439] [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: 09/23/2023] [Accepted: 11/26/2023] [Indexed: 02/16/2024] Open
Abstract
Background: P7C3 is a novel compound that has been widely applied in neurodegenerative diseases and nerve injury repair. Here, we show that higher concentrations of P7C3 than are required for in vivo neuroprotection have the novel function of suppressing renal cell carcinoma (RCC) proliferation and metastasis. Methods: Colony formation, CCK-8 and EdU assay were applied to evaluate RCC cell proliferation. Wound healing and transwell assay were used to measure RCC cell migration and invasion. Flow cytometry assay was employed to detect RCC cell apoptosis and cell cycle. qRT-PCR assay was carried out to measure ribonucleotide reductase subunit M2 (RRM2) mRNA expression level, while western blot assay was utilized to detect the expression level of target proteins. RCC cell growth in vivo was determined by xenografts in mice. Results: We observed that high concentrations of P7C3 could restrain the proliferation and metastasis of RCC cells and promote cell apoptosis. Mechanistically, this new effect of higher dose of P7C3 was associated with reduced expression of RRM2, and the beneficial efficacy of P7C3 in RCC was blocked when suppression of RRM2 was prevented. When RRM2 suppression was permitted, the cGAS-STING pathway was activated by virtue of RRM2/Bcl-2/Bax signaling. Lastly, intraperitoneal injection of this high level of P7C3 in mice potently inhibited tumor growth. Conclusion: In conclusion, we show here that P7C3 that exerts an anti-cancer effect in RCC. Our study indicated that P7C3 might act as a novel drug for RCC in the future. The regulatory signal pathway RRM2/Bcl-2/BAX/cGAS-STING might present novel insight to the potential mechanism of RCC development.
Collapse
Affiliation(s)
- Ge Shu
- Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenjin Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Chenchen Huang
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Hui Shan
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing Ye
- Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jianfa Li
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaoting Gui
- Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Tian B, Bai J, Sheng L, Chen H, Chang W, Zhang Y, Yao C, Zhou C, Wang X, Shan H, Dong Q, Wang C, Zhou X. P7C3 Ameliorates Bone Loss by Inhibiting Osteoclast Differentiation and Promoting Osteogenesis. JBMR Plus 2023; 7:e10811. [PMID: 38130773 PMCID: PMC10731119 DOI: 10.1002/jbm4.10811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 12/23/2023] Open
Abstract
Bone homeostasis, the equilibrium between bone resorption and formation, is essential for maintaining healthy bone tissue in adult humans. Disruptions of this process can lead to pathological conditions such as osteoporosis. Dual-targeted agents, capable of inhibiting excessive bone resorption and stimulating bone formation, are being explored as a promising strategy for developing new treatments to address osteoporosis. In this study, we investigated the effects of P7C3 on bone remodeling and its potential therapeutic role in osteoporosis treatment in mice. Specifically, P7C3 can remarkably suppress receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation in bone marrow macrophages via the Akt-NF-κB-NFATc1 signaling pathway. Additionally, RNA sequencing (RNAseq) analysis revealed that P7C3 promoted osteoblast differentiation and function through the Wnt/β-catenin signaling pathway, thereby enhancing bone formation. Furthermore, μCT analysis and histological examination of bone tissues from P7C3-treated mice showed attenuation of both Ti-induced bone erosion and ovariectomy (OVX)-induced bone loss. These findings suggest that P7C3 may have a novel function in bone remodeling and may be a promising therapeutic agent for the treatment of osteoporosis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Bo Tian
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Jinyu Bai
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Sheng
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hao Chen
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Wenju Chang
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Chenlu Yao
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Chenmeng Zhou
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Xiaoyu Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Huajian Shan
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Qirong Dong
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Xiaozhong Zhou
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
14
|
Kumar MP, G M, Amaladass P, Manikandan C, Dhayalan V. Recent synthetic strategies for the construction of functionalized carbazoles and their heterocyclic motifs enabled by Lewis acids. RSC Adv 2023; 13:32596-32626. [PMID: 37936643 PMCID: PMC10626344 DOI: 10.1039/d3ra06396h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023] Open
Abstract
This article demonstrates recent innovative cascade annulation methods for preparing functionalized carbazoles and their related polyaromatic heterocyclic compounds enabled by Lewis acid catalysts. Highly substituted carbazole scaffolds were synthesized via Lewis acid mediated Friedel-Crafts arylation, electrocyclization, intramolecular cyclization, cycloaddition, C-N bond-formations, aromatization and cascade domino reactions, metal-catalyzed, iodine catalyzed reactions and multi-component reactions. This review article mainly focuses on Lewis acid-mediated recent synthetic methods to access a variety of electron-rich and electron-poor functional groups substituted carbazole frameworks in one-pot reactions. Polyaromatic carbazole and their related nitrogen-based heterocyclic compounds were found in several synthetic applications in pharma industries, energy devices, and materials sciences. Moreover, the review paper briefly summarised new synthetic strategies of carbazole preparation approaches will assist academic and pharma industries in identifying innovative protocols for producing poly-functionalized carbazoles and related highly complex heterocyclic compounds and discovering active pharmaceutical drugs or carbazole-based alkaloids and natural products.
Collapse
Affiliation(s)
- Marappan Pradeep Kumar
- Department of Chemistry, National Institute of Technology Puducherry Karaikal-609609 Union Territory Puducherry India https://vasudeva49.wixsite.com/catalysislab
| | - Mahantesh G
- Department of Chemistry, National Institute of Technology Puducherry Karaikal-609609 Union Territory Puducherry India https://vasudeva49.wixsite.com/catalysislab
| | - P Amaladass
- Department of Chemistry, Madanapalle Institute of Technology & Science Madanapalle 517325 Andhra Pradesh India
| | - Chitrarasu Manikandan
- Department of Chemistry, National Institute of Technology Puducherry Karaikal-609609 Union Territory Puducherry India https://vasudeva49.wixsite.com/catalysislab
| | - Vasudevan Dhayalan
- Department of Chemistry, National Institute of Technology Puducherry Karaikal-609609 Union Territory Puducherry India https://vasudeva49.wixsite.com/catalysislab
| |
Collapse
|
15
|
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
|
16
|
Wang Z, Gonzalez KM, Cordova LE, Lu J. Nanotechnology-empowered therapeutics targeting neurodegenerative diseases. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1907. [PMID: 37248794 PMCID: PMC10525015 DOI: 10.1002/wnan.1907] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 04/15/2023] [Accepted: 05/01/2023] [Indexed: 05/31/2023]
Abstract
Neurodegenerative diseases are posing pressing health issues due to the high prevalence among aging populations in the 21st century. They are evidenced by the progressive loss of neuronal function, often associated with neuronal necrosis and many related devastating complications. Nevertheless, effective therapeutical strategies to treat neurodegenerative diseases remain a tremendous challenge due to the multisystemic nature and limited drug delivery to the central nervous system. As a result, there is a pressing need to develop effective alternative therapeutics to manage the progression of neurodegenerative diseases. By utilizing the functional reconstructive materials and technologies with specific targeting ability at the nanoscale level, nanotechnology-empowered medicines can transform the therapeutic paradigms of neurodegenerative diseases with minimal systemic side effects. This review outlines the current applications and progresses of the nanotechnology-enabled drug delivery systems to enhance the therapeutic efficacy in treating neurodegenerative diseases. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Leyla Estrella Cordova
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, United States
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, Arizona, 85721, United States
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, United States
| |
Collapse
|
17
|
Hu X, Xie J, Yang Y, Qiu Z, Lu W, Lin X, Xu B. Multi-Target Neural Differentiation (MTND) Therapeutic Cocktail to Suppress Brain Tumor. Int J Mol Sci 2023; 24:12329. [PMID: 37569705 PMCID: PMC10418641 DOI: 10.3390/ijms241512329] [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: 07/13/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Brain tumors have been proved challenging to treat. Here we established a Multi-Target Neural Differentiation (MTND) therapeutic cocktail to achieve effective and safe treatment of brain malignancies by targeting the important hallmarks in brain cancers: poor cell differentiation and compromised cell cycle. In-vitro and in-vivo experiments confirmed the significant therapeutic effect of our MTND therapy. Significantly improved therapeutic effects over current first-line chemo-drugs have been identified in clinical cells, with great inhibition of the growth and migration of tumor cells. Further in-vivo experiments confirmed that sustained MTND treatment showed a 73% reduction of the tumor area. MTND also induced strong expression of phenotypes associated with cell cycle exit/arrest and rapid neural reprograming from clinical glioma cells to glutamatergic and GABAergic expressing cells, which are two key neuronal types involved in many human brain functions, including learning and memory. Collectively, MTND induced multi-targeted genotypic expression changes to achieve direct neural conversion of glioma cells and controlled the cell cycle/tumorigenesis development, helping control tumor cells' malignant proliferation and making it possible to treat brain malignant tumors effectively and safely. These encouraging results open avenues to developing new therapies for brain malignancies beyond cytotoxic agents, providing more effective medication recommendations with reduced toxicity.
Collapse
Affiliation(s)
- Xiaoping Hu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China; (X.H.); (Y.Y.)
| | - Jingdun Xie
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation for Cancer Medicine, Guangzhou 510060, China; (J.X.); (W.L.)
| | - Yilin Yang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China; (X.H.); (Y.Y.)
| | - Ziyi Qiu
- School of Biomedical Engineering (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China;
| | - Weicheng Lu
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation for Cancer Medicine, Guangzhou 510060, China; (J.X.); (W.L.)
| | - Xudong Lin
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China; (X.H.); (Y.Y.)
| | - Bingzhe Xu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510275, China; (X.H.); (Y.Y.)
- School of Biomedical Engineering (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China;
| |
Collapse
|
18
|
Gu X, Yao H, Kwon I, Wang G. Small-molecule activation of NAMPT as a potential neuroprotective strategy. LIFE MEDICINE 2022; 1:270-272. [PMID: 39872755 PMCID: PMC11749627 DOI: 10.1093/lifemedi/lnac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/21/2022] [Indexed: 01/30/2025]
Affiliation(s)
- Xuanyu Gu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Hong Yao
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Ilmin Kwon
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
19
|
Roy D, Tharra P, Baire B. An approach to functionalized carbazoles from Z-enoate propargylic alcohols. A unified total synthesis of N-Me-carazostatin, N-Me-carbazoquinocin C and N-Me-lipocarbazole A4. Chem Commun (Camb) 2022; 58:10210-10213. [PMID: 36000534 DOI: 10.1039/d2cc03526j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Development of an acid catalyzed, intramolecular benzannulation of indoles for the synthesis of functionalized carbazoles has been reported. The indole appended Z-enoate propargylic alcohols have been employed. The N-EDG-indoles involve the 5-exo-dig cyclization followed by 1,2-migration to give the carbazole-butenoates, whereas the N-EWG-indoles undergo the Z-enoate assisted Meyer-Schuster rearrangement to give the dihydrocarbazole-4-oxo-butanoates. Utilizing one of the 2-methyl-carbazole-butyraldehyde (obtained from the corresponding carbazole-butanoate) as the key intermediate, we have developed a simple approach for an efficient synthesis of N-Me-carazostatin, N-Me-carbazoquinocin C and N-Me-lipocarbazole A4.
Collapse
Affiliation(s)
- Debayan Roy
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, India.
| | - Prabhakararao Tharra
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, India.
| | - Beeraiah Baire
- Department of Chemistry, Indian Institute of Technology Madras, Chennai-600036, India.
| |
Collapse
|
20
|
Affiliation(s)
- Li He
- Harbin Institute of Technology School of Chemistry and Chemical Engineering CHINA
| | - Yongjun Xu
- Harbin Institute of Technology School of Chemistry and Chemical Engineering CHINA
| |
Collapse
|
21
|
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]
|
22
|
Xie WS, Shehzadi K, Ma HL, Liang JH. A Potential Strategy for Treatment of Neurodegenerative Disorders by Regulation of Adult Hippocampal Neurogenesis in Human Brain. Curr Med Chem 2022; 29:5315-5347. [DOI: 10.2174/0929867329666220509114232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/13/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Adult hippocampal neurogenesis is a multistage mechanism that continues throughout the lifespan of human and non-human mammals. These adult-born neurons in the central nervous system (CNS) play a significant role in various hippocampus-dependent processes, including learning, mood regulation, pattern recognition, etc. Reduction of adult hippocampal neurogenesis, caused by multiple factors such as neurological disorders and aging, would impair neuronal proliferation and differentiation and result in memory loss. Accumulating studies have indicated that functional neuron impairment could be restored by promoting adult hippocampal neurogenesis. In this review, we summarized the small molecules that could efficiently promote the process of adult neurogenesis, particularly the agents that have the capacity of crossing the blood-brain barrier (BBB), and showed in vivo efficacy in mammalian brains. This may pave the way for the rational design of drugs to treat humnan neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Wei-Song Xie
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Kiran Shehzadi
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Hong-Le Ma
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
| | - Jian-Hua Liang
- Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 102488, China
- Yangtze Delta Region Academy of Beijing Institute of Technology, Jiaxing 314019, China
| |
Collapse
|
23
|
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
|
24
|
Yang X, Qiang Q, Li N, Feng P, Wei W, Hölscher C. Neuroprotective Mechanisms of Glucagon-Like Peptide-1-Based Therapies in Ischemic Stroke: An Update Based on Preclinical Research. Front Neurol 2022; 13:844697. [PMID: 35370875 PMCID: PMC8964641 DOI: 10.3389/fneur.2022.844697] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
The public and social health burdens of ischemic stroke have been increasing worldwide. Hyperglycemia leads to a greater risk of stroke. This increased risk is commonly seen among patients with diabetes and is in connection with worsened clinical conditions and higher mortality in patients with acute ischemic stroke (AIS). Therapy for stroke focuses mainly on restoring cerebral blood flow (CBF) and ameliorating neurological impairment caused by stroke. Although choices of stroke treatment remain limited, much advance have been achieved in assisting patients in recovering from ischemic stroke, along with progress of recanalization therapy through pharmacological and mechanical thrombolysis. However, it is still necessary to develop neuroprotective therapies for AIS to protect the brain against injury before and during reperfusion, prolong the time window for intervention, and consequently improve neurological prognosis. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are broadly regarded as effective drugs in the treatment of type 2 diabetes mellitus (T2DM). Preclinical data on GLP-1 and GLP-1 RAs have displayed an impressive neuroprotective efficacy in stroke, Parkinson's disease (PD), Alzheimer's disease (AD), Amyotrophic lateral sclerosis (ALS), and other neurodegenerative diseases. Based on the preclinical studies in the past decade, we review recent progress in the biological roles of GLP-1 and GLP-1 RAs in ischemic stroke. Emphasis will be placed on their neuroprotective effects in experimental models of cerebral ischemia stroke at cellular and molecular levels.
Collapse
Affiliation(s)
- Xiaoyan Yang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Qiang Qiang
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Nan Li
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Peng Feng
- Department of Neurology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenshi Wei
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Christian Hölscher
- Department of Neurology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China.,Henan University of Chinese Medicine, Academy of Chinese Medical Science, Zhengzhou, China
| |
Collapse
|
25
|
Schroeder R, Nguyen L, Pieper AA, Stevens HE. Maternal treatment with P7C3-A20 protects from impaired maternal care after chronic gestational stress. Behav Brain Res 2022; 416:113558. [PMID: 34453970 PMCID: PMC8573727 DOI: 10.1016/j.bbr.2021.113558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/04/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023]
Abstract
Chronic stress during pregnancy harms both the mother and developing child, and there is an urgent unmet need to understand this process in order to develop protective treatments. Here, we report that chronic gestational stress (CGS) causes aberrant maternal care behavior in the form of increased licking and grooming, decreased nursing, and increased time spent nest building. Treatment of CGS-exposed dams with the NAD+-stabilizing agent P7C3-A20 during pregnancy and postpartum, however, preserved normal maternal care behavior. CGS also caused abnormally low weight gain during gestation and postpartum, which was partially ameliorated by maternal treatment with P7C3-A20. Dams also displayed hyperactive locomotion after CGS, which was not affected by P7C3-A20. Although dams did not display a classic depressive-like phenotype after CGS, some changes in anxiety- and depressive-like behaviors were observed. Our results highlight the need for further characterization of the effects of chronic gestational stress on maternal care behavior and provide clues to possible protective mechanisms.
Collapse
Affiliation(s)
- Rachel Schroeder
- Department of Psychiatry, University of Iowa, Iowa City, IA,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA
| | - Lynn Nguyen
- Department of Psychiatry, University of Iowa, Iowa City, IA
| | - Andrew A. Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA,Department of Psychiatry, Case Western Reserve University,Department of Neuroscience, Case Western Reserve University, Cleveland, OH 44106 USA,University of Texas Southwestern Medical Center, Dallas, Texas USA,Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA,Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, OH 44106 USA,Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, NY, NY, USA
| | - Hanna E. Stevens
- Department of Psychiatry, University of Iowa, Iowa City, IA,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA
| |
Collapse
|
26
|
Necroptosis mediated by receptor interacting protein kinase 3 as critical players in experimental congenital hypothyroidism related neuronal damage. North Clin Istanb 2021; 8:472-478. [PMID: 34909585 PMCID: PMC8630716 DOI: 10.14744/nci.2021.26043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/25/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Congenital hypothyroidism (CH) is literally described as congenital thyroid hormone imperfection. The primary objective of this research was to reveal the possible relation between receptor-acting protein kinase 3 (RIPK3) activity and neuronal damages in rat pups with CH. In addition, we evaluated the favorable impacts of 3.6-dibromo-α-([phenylamino] methyl)-9H-carbazole-9-ethanol (P7C3) reducing RIPK3 activity. METHODS Adult rats were accordingly assigned into four groups: Group 1, which is called congenital hypothyroid; Group 2, which is called congenital hypothyroid administered P7C3; Group 3, called CH administered P7C3 and L-thyroxine; and Group 4, control group. RIPK3 level in plasma concentration and its expression in tissue was determined in all groups. RESULTS Increased RIPK3 expressions were detected as high in the CH group when it is compared to the control group. Furthermore; the expressions in neuronal cytoplasm were found similar among Groups II and III. RIPK3 expressions in those two groups were relatively higher than in the control group. Most reacted parts of the brain were especially Purkinje cells in the cerebellum. CONCLUSION It is concluded that there is excellent parallelism among damaged neurons and high RIPK3 activity in CH pathogenesis. P7C3 compounds may have a safeguarding impact on CH due to decreasing RIPK3 activity.
Collapse
|
27
|
Clanton NA, Spiller TE, Ortiz E, Gao Z, Rodriguez-Poirier JM, DelMonte AJ, Frantz DE. A Metal-Free Reductive N-Alkylation of Indoles with Aldehydes. Org Lett 2021; 23:3233-3236. [PMID: 33630601 DOI: 10.1021/acs.orglett.1c00179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A simple metal-free method has been developed for the reductive N-alkylation of indoles employing aldehydes as the alkylating agent and inexpensive Et3SiH as the reductant. A wide range of aromatic and aliphatic aldehydes are viable substrates along with a variety of substituted indoles. In addition, the method was applied to a one-pot sequential 1,3-alkylation of a substituted indole and successfully demonstrated on a 100 mmol scale.
Collapse
Affiliation(s)
- Nicholas A Clanton
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Taylor E Spiller
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Eliezer Ortiz
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| | - Zhinong Gao
- Chemical Process Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08901, United States
| | | | - Albert J DelMonte
- Chemical Process Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08901, United States
| | - Doug E Frantz
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, Texas 78249, United States
| |
Collapse
|
28
|
Jaromin A, Parapini S, Basilico N, Zaremba-Czogalla M, Lewińska A, Zagórska A, Walczak M, Tyliszczak B, Grzeszczak A, Łukaszewicz M, Kaczmarek Ł, Gubernator J. Azacarbazole n-3 and n-6 polyunsaturated fatty acids ethyl esters nanoemulsion with enhanced efficacy against Plasmodium falciparum. Bioact Mater 2021; 6:1163-1174. [PMID: 33134609 PMCID: PMC7588843 DOI: 10.1016/j.bioactmat.2020.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/27/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022] Open
Abstract
Alternative therapies are necessary for the treatment of malaria due to emerging drug resistance. However, many promising antimalarial compounds have poor water solubility and suffer from the lack of suitable delivery systems, which seriously limits their activity. To address this problem, we synthesized a series of azacarbazoles that were evaluated for antimalarial activity against D10 (chloroquine-sensitive) and W2 (chloroquine-resistant) strains of P. falciparum. The most active compound, 9H-3-azacarbazole (3), was encapsulated in a novel o/w nanoemulsion consisting of ethyl esters of polyunsaturated fatty acids n-3 and n-6 obtained from flax oil as the oil phase, Smix (Tween 80 and Transcutol HP) and water. This formulation was further analyzed using transmission electron microscopy, dynamic light scattering and in vitro and in vivo studies. It was shown that droplets of the 3-loaded nanosystem were spherical, with satisfactory stability, without cytotoxicity towards fibroblasts and intestinal cell lines at concentrations corresponding to twice the IC50 for P. falciparum. Moreover, the nanoemulsion with this type of oil phase was internalized by Caco-2 cells. Additionally, pharmacokinetics demonstrated rapid absorption of compound 3 (tmax = 5.0 min) after intragastric administration of 3-encapsulated nanoemulsion at a dose of 0.02 mg/kg in mice, with penetration of compound 3 to deep compartments. The 3-encapsulated nanoemulsion was found to be 2.8 and 4.2 times more effective in inhibiting the D10 and W2 strains of the parasite, respectively, compared to non-encapsulated 3. Our findings support a role for novel o/w nanoemulsions as delivery vehicles for antimalarial drugs.
Collapse
Affiliation(s)
- Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Nicoletta Basilico
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | | | | | - Agnieszka Zagórska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, Cracow, Poland
| | - Maria Walczak
- Chair and Department of Toxicology, Jagiellonian University Medical College, Faculty of Pharmacy, Cracow, Poland
| | - Bożena Tyliszczak
- Instytute of Materials Science, Cracow University of Technology, Cracow, Poland
| | - Aleksandra Grzeszczak
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Marcin Łukaszewicz
- Department of Biotransformation, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Jerzy Gubernator
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
29
|
Ovchenkova EN, Bichan NG, Gruzdev MS, Ksenofontov AA, Gostev FE, Shelaev IV, Nadtochenko VA, Lomova TN. Carbazole-functionalized cobalt( ii) porphyrin axially bonded with C 60/C 70 derivatives: synthesis and characterization. NEW J CHEM 2021. [DOI: 10.1039/d1nj00980j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Depending on the structure of the fullero[60]/[70]pyrrolidine, the carbazole-functionalized cobalt(ii) porphyrin forms donor–acceptor systems of different compositions.
Collapse
Affiliation(s)
- E. N. Ovchenkova
- G. A. Krestov Institute of Solution Chemistry
- Russian Academy of Sciences Akademicheskaya str
- 1
- Ivanovo
- Russia
| | - N. G. Bichan
- G. A. Krestov Institute of Solution Chemistry
- Russian Academy of Sciences Akademicheskaya str
- 1
- Ivanovo
- Russia
| | - M. S. Gruzdev
- G. A. Krestov Institute of Solution Chemistry
- Russian Academy of Sciences Akademicheskaya str
- 1
- Ivanovo
- Russia
| | - A. A. Ksenofontov
- G. A. Krestov Institute of Solution Chemistry
- Russian Academy of Sciences Akademicheskaya str
- 1
- Ivanovo
- Russia
| | - F. E. Gostev
- N. N. Semenov Federal Research Center for Chemical Physics
- Russian Academy of Sciences
- Moscow
- Russia
| | - I. V. Shelaev
- N. N. Semenov Federal Research Center for Chemical Physics
- Russian Academy of Sciences
- Moscow
- Russia
| | - V. A. Nadtochenko
- N. N. Semenov Federal Research Center for Chemical Physics
- Russian Academy of Sciences
- Moscow
- Russia
| | - T. N. Lomova
- G. A. Krestov Institute of Solution Chemistry
- Russian Academy of Sciences Akademicheskaya str
- 1
- Ivanovo
- Russia
| |
Collapse
|
30
|
Reddy CR, Srinivasu E, Sathish P, Subbarao M, Donthiri RR. One-Pot Arylative Benzannulation of 2-Carbonyl-3-propargyl Indoles with Boronic Acids Leading to Arylated Carbazoles. J Org Chem 2021; 86:1118-1132. [PMID: 33322899 DOI: 10.1021/acs.joc.0c02601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Arylative annulation of 2-carbonyl-3-propargyl indoles with boronic acids under sequential palladium/triflic acid catalysis is described. The present strategy to provide di- and triaryl carbazoles in one pot involves benzannulation through difunctionalization of alkynes. The strategy showed a good substrate scope with respect to boronic acids as well as 2-carbonyl-3-propargyl indoles to afford the corresponding carbazoles in decent yields.
Collapse
Affiliation(s)
- Chada Raji Reddy
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ejjirotu Srinivasu
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Puppala Sathish
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Muppidi Subbarao
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ramachandra Reddy Donthiri
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
| |
Collapse
|
31
|
Banerjee A, Kundu S, Bhattacharyya A, Sahu S, Maji MS. Benzannulation strategies for the synthesis of carbazoles, indolocarbazoles, benzocarbazoles, and carbolines. Org Chem Front 2021. [DOI: 10.1039/d1qo00092f] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review presents a critical and authoritative analysis of several exciting benzannulation approaches developed in the past decade for the construction of carbazoles, indolocarbazoles, benzocarbazoles, and carbolines.
Collapse
Affiliation(s)
- Ankush Banerjee
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Samrat Kundu
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Arya Bhattacharyya
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Samrat Sahu
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Modhu Sudan Maji
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| |
Collapse
|
32
|
P7C3-A20 treatment one year after TBI in mice repairs the blood-brain barrier, arrests chronic neurodegeneration, and restores cognition. Proc Natl Acad Sci U S A 2020; 117:27667-27675. [PMID: 33087571 PMCID: PMC7959512 DOI: 10.1073/pnas.2010430117] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic neurodegeneration, a major cause of the long-term disabilities that afflict survivors of traumatic brain injury (TBI), is linked to an increased risk for late-life neurodegenerative disorders, including Alzheimer’s disease, Parkinson’s disease, vascular dementia, and chronic traumatic encephalopathy. Here, we report on the restoration of blood–brain barrier (BBB) structure and function by P7C3-A20 when administered 12 mo after TBI. This pharmacotherapy was associated with cessation of chronic neurodegeneration and recovery of normal cognitive function, benefits that persisted long after treatment cessation. Pharmacologic renewal of BBB integrity may thus provide a new treatment option for patients who have suffered a remote TBI, or other neurological conditions associated with BBB deterioration. Chronic neurodegeneration in survivors of traumatic brain injury (TBI) is a major cause of morbidity, with no effective therapies to mitigate this progressive and debilitating form of nerve cell death. Here, we report that pharmacologic restoration of the blood–brain barrier (BBB), 12 mo after murine TBI, is associated with arrested axonal neurodegeneration and cognitive recovery, benefits that persisted for months after treatment cessation. Recovery was achieved by 30 d of once-daily administration of P7C3-A20, a compound that stabilizes cellular energy levels. Four months after P7C3-A20, electron microscopy revealed full repair of TBI-induced breaks in cortical and hippocampal BBB endothelium. Immunohistochemical staining identified additional benefits of P7C3-A20, including restoration of normal BBB endothelium length, increased brain capillary pericyte density, increased expression of BBB tight junction proteins, reduced brain infiltration of immunoglobulin, and attenuated neuroinflammation. These changes were accompanied by cessation of TBI-induced chronic axonal degeneration. Specificity for P7C3-A20 action on the endothelium was confirmed by protection of cultured human brain microvascular endothelial cells from hydrogen peroxide-induced cell death, as well as preservation of BBB integrity in mice after exposure to toxic levels of lipopolysaccharide. P7C3-A20 also protected mice from BBB degradation after acute TBI. Collectively, our results provide insights into the pathophysiologic mechanisms behind chronic neurodegeneration after TBI, along with a putative treatment strategy. Because TBI increases the risks of other forms of neurodegeneration involving BBB deterioration (e.g., Alzheimer’s disease, Parkinson’s disease, vascular dementia, chronic traumatic encephalopathy), P7C3-A20 may have widespread clinical utility in the setting of neurodegenerative conditions.
Collapse
|
33
|
Marotta G, Basagni F, Rosini M, Minarini A. Memantine Derivatives as Multitarget Agents in Alzheimer's Disease. Molecules 2020; 25:molecules25174005. [PMID: 32887400 PMCID: PMC7504780 DOI: 10.3390/molecules25174005] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/24/2022] Open
Abstract
Memantine (3,5-dimethyladamantan-1-amine) is an orally active, noncompetitive N-methyl-D-aspartate receptor (NMDAR) antagonist approved for treatment of moderate-to-severe Alzheimer’s disease (AD), a neurodegenerative condition characterized by a progressive cognitive decline. Unfortunately, memantine as well as the other class of drugs licensed for AD treatment acting as acetylcholinesterase inhibitors (AChEIs), provide only symptomatic relief. Thus, the urgent need in AD drug development is for disease-modifying therapies that may require approaching targets from more than one path at once or multiple targets simultaneously. Indeed, increasing evidence suggests that the modulation of a single neurotransmitter system represents a reductive approach to face the complexity of AD. Memantine is viewed as a privileged NMDAR-directed structure, and therefore, represents the driving motif in the design of a variety of multi-target directed ligands (MTDLs). In this review, we present selected examples of small molecules recently designed as MTDLs to contrast AD, by combining in a single entity the amantadine core of memantine with the pharmacophoric features of known neuroprotectants, such as antioxidant agents, AChEIs and Aβ-aggregation inhibitors.
Collapse
|
34
|
Bai J, Zeng S, Zhu J, Fu C, He M, Zhu J, Chen S, Fu X, Li P, Lin Z. The Small Molecule P7C3-A20 Exerts Neuroprotective Effects in a Hypoxic–ischemic Encephalopathy Model via Activation of PI3K/AKT/GSK3β Signaling. Neuroscience 2020; 441:197-208. [DOI: 10.1016/j.neuroscience.2020.05.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 11/27/2022]
|
35
|
Artault M, Mokhtari N, Cantin T, Martin-Mingot A, Thibaudeau S. Superelectrophilic Csp 3-H bond fluorination of aliphatic amines in superacid: the striking role of ammonium-carbenium dications. Chem Commun (Camb) 2020; 56:5905-5908. [PMID: 32342071 DOI: 10.1039/d0cc02081h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The superacid-promoted electrophilic Csp3-H bond activation of aliphatic amines generates superelectrophilic species that can be subsequently fluorinated. Demonstrated by low-temperature in situ NMR experiments, the ammonium-carbenium dications, crucial for this reaction, can also react with C-H bonds opening future synthesis perspectives for this mode of activation.
Collapse
Affiliation(s)
- M Artault
- Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Université de Poitiers, CNRS, Superacid Group in Organic Synthesis Team, F-86073 Poitiers, France.
| | - N Mokhtari
- Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Université de Poitiers, CNRS, Superacid Group in Organic Synthesis Team, F-86073 Poitiers, France.
| | - T Cantin
- Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Université de Poitiers, CNRS, Superacid Group in Organic Synthesis Team, F-86073 Poitiers, France.
| | - A Martin-Mingot
- Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Université de Poitiers, CNRS, Superacid Group in Organic Synthesis Team, F-86073 Poitiers, France.
| | - S Thibaudeau
- Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP), Université de Poitiers, CNRS, Superacid Group in Organic Synthesis Team, F-86073 Poitiers, France.
| |
Collapse
|
36
|
Raji Reddy C, Subbarao M, Sathish P, Kolgave DH, Donthiri RR. One-Pot Assembly of 3-Hydroxycarbazoles via Uninterrupted Propargylation/Hydroxylative Benzannulation Reactions. Org Lett 2020; 22:689-693. [PMID: 31909623 DOI: 10.1021/acs.orglett.9b04472] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel strategy for the synthesis of 3-hydroxycarbazoles involving the consecutive propargylation/palladium-catalyzed hydroxylative benzannulation of indole-2-carbonyls with propargylic alcohols has been exploited. This one-pot procedure leads to a wide range of substituted 3-hydroxycarbazoles in high yield with a broad substrate scope. The method was further extended to access furano-carbazole derivatives from dialkynols via tandem annulations.
Collapse
Affiliation(s)
- Chada Raji Reddy
- Department of Organic Synthesis & Process Chemistry , CSIR-Indian Institute of Chemical Technology , Hyderabad 500007 , India.,Academy of Scientific and Innovative Research (AcSIR) , Ghaziabad 201 002 , India
| | - Muppidi Subbarao
- Department of Organic Synthesis & Process Chemistry , CSIR-Indian Institute of Chemical Technology , Hyderabad 500007 , India.,Academy of Scientific and Innovative Research (AcSIR) , Ghaziabad 201 002 , India
| | - Puppala Sathish
- Department of Organic Synthesis & Process Chemistry , CSIR-Indian Institute of Chemical Technology , Hyderabad 500007 , India.,Academy of Scientific and Innovative Research (AcSIR) , Ghaziabad 201 002 , India
| | - Dattahari H Kolgave
- Department of Organic Synthesis & Process Chemistry , CSIR-Indian Institute of Chemical Technology , Hyderabad 500007 , India.,Academy of Scientific and Innovative Research (AcSIR) , Ghaziabad 201 002 , India
| | - Ramachandra Reddy Donthiri
- Department of Organic Synthesis & Process Chemistry , CSIR-Indian Institute of Chemical Technology , Hyderabad 500007 , India
| |
Collapse
|
37
|
Duan FX, Shi YJ, Chen J, Ding SQ, Wang FC, Tang J, Wang R, Shen L, Xi J, Qi Q, Lü HZ, Hu JG. Neuroprotective effects of P7C3 against spinal cord injury in rats. Exp Biol Med (Maywood) 2019; 244:1680-1687. [PMID: 31718264 DOI: 10.1177/1535370219888620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Fei-Xiang Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - Yu-Jiao Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - Jing Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China.,Department of Immunology, Bengbu Medical College, Bengbu 233030, P.R. China
| | - Shu-Qin Ding
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - Feng-Chao Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China
| | - Jie Tang
- Department of Immunology, Bengbu Medical College, Bengbu 233030, P.R. China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - Lin Shen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - Jin Xi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| | - He-Zuo Lü
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China.,Department of Immunology, Bengbu Medical College, Bengbu 233030, P.R. China
| | - Jian-Guo Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu 233004, P.R. China
| |
Collapse
|
38
|
Yang Y, Chen R, Wu X, Zhao Y, Fan Y, Xiao Z, Han J, Sun L, Wang X, Dai J. Rapid and Efficient Conversion of Human Fibroblasts into Functional Neurons by Small Molecules. Stem Cell Reports 2019; 13:862-876. [PMID: 31631018 PMCID: PMC6893066 DOI: 10.1016/j.stemcr.2019.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 01/15/2023] Open
Abstract
Recent studies have demonstrated that human astrocytes and fibroblasts can be directly converted into functional neurons by small molecules. However, fibroblasts, as a potentially better cell resource for transplantation, are not as easy to reprogram as astrocytes regarding their fate to neurons, and chemically induced neurons (iNs) with low efficiency from fibroblasts resulted in limited application for the treatment of neurological disorders, including depression. Here, we report that human fibroblasts can be efficiently and directly reprogrammed into glutamatergic neuron-like cells by serially exposing cells to a combination of small molecules. These iNs displayed neuronal transcriptional networks, and also exhibited mature firing patterns and formed functional synapses. Importantly, iNs could integrate into local circuits after transplantation into postnatal mouse brain. Our study provides a rapid and efficient transgene-free approach for chemically generating neuron-like cells from human fibroblasts. Furthermore, our approach offers strategies for disease modeling and drug discovery in central nervous system disorders. Small molecules efficiently reprogram human fibroblasts into glutamatergic neurons iNs show neuronal transcriptional networks resembling that of human primary neurons iNs can survive, mature, and integrate into local circuits after transplantation P7C3-A20 is the most important component of the cocktail in the reprogramming process
Collapse
Affiliation(s)
- Yaming Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruiguo Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jin Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Le Sun
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoqun Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
39
|
Jain S, LaFrancois JJ, Botterill JJ, Alcantara-Gonzalez D, Scharfman HE. Adult neurogenesis in the mouse dentate gyrus protects the hippocampus from neuronal injury following severe seizures. Hippocampus 2019; 29:683-709. [PMID: 30672046 PMCID: PMC6640126 DOI: 10.1002/hipo.23062] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/29/2018] [Accepted: 11/30/2018] [Indexed: 01/20/2023]
Abstract
Previous studies suggest that reducing the numbers of adult-born neurons in the dentate gyrus (DG) of the mouse increases susceptibility to severe continuous seizures (status epilepticus; SE) evoked by systemic injection of the convulsant kainic acid (KA). However, it was not clear if the results would be the same for other ways to induce seizures, or if SE-induced damage would be affected. Therefore, we used pilocarpine, which induces seizures by a different mechanism than KA. Also, we quantified hippocampal damage after SE. In addition, we used both loss-of-function and gain-of-function methods in adult mice. We hypothesized that after loss-of-function, mice would be more susceptible to pilocarpine-induced SE and SE-associated hippocampal damage, and after gain-of-function, mice would be more protected from SE and hippocampal damage after SE. For loss-of-function, adult neurogenesis was suppressed by pharmacogenetic deletion of dividing radial glial precursors. For gain-of-function, adult neurogenesis was increased by conditional deletion of pro-apoptotic gene Bax in Nestin-expressing progenitors. Fluoro-Jade C (FJ-C) was used to quantify neuronal injury and video-electroencephalography (video-EEG) was used to quantify SE. Pilocarpine-induced SE was longer in mice with reduced adult neurogenesis, SE had more power and neuronal damage was greater. Conversely, mice with increased adult-born neurons had shorter SE, SE had less power, and there was less neuronal damage. The results suggest that adult-born neurons exert protective effects against SE and SE-induced neuronal injury.
Collapse
Affiliation(s)
- Swati Jain
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - Justin J. Botterill
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan Kline Institute of Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY 10962, USA
- Departments of Child & Adolescent Psychiatry, Neuroscience & Physiology, and Psychiatry, New York Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
40
|
Lazutkin A, Podgorny O, Enikolopov G. Modes of division and differentiation of neural stem cells. Behav Brain Res 2019; 374:112118. [PMID: 31369774 DOI: 10.1016/j.bbr.2019.112118] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/25/2019] [Accepted: 07/28/2019] [Indexed: 01/09/2023]
Abstract
Hippocampal neurogenesis presents an unorthodox form of neuronal plasticity and may be relevant for the normal or abnormal functioning of the human and animal brain. As production of new neurons decreases after birth, purposefully activating stem cells to create additional new neurons may augment brain function or slow a disease's progression. Here, we describe current models of hippocampal stem cell maintenance and differentiation, and emphasize key features of neural stem cells' turnover that may define hippocampal neurogenesis enhancement attempts' long-term consequences. We argue that even the basic blueprint of how stem cells are maintained, divide, differentiate, and are eliminated is still contentious, with different models potentially leading to vastly different outcomes in regard to neuronal production and stem cell pool preservation. We propose that to manipulate neurogenesis for a long-term benefit, we must first understand the outline of the neural stem cells' lifecycle.
Collapse
Affiliation(s)
- Alexander Lazutkin
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States; Moscow Institute of Physics and Technology, Moscow, Russia; P.K. Anokhin Institute for Normal Physiology, Moscow, Russia
| | - Oleg Podgorny
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States; Koltzov Institute of Developmental Biology, Moscow, Russia
| | - Grigori Enikolopov
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States.
| |
Collapse
|
41
|
Abstract
The antiapoptotic, neuroprotective compound P7C3-A20 reduces neurological deficits when administered to murine in-vivo models of traumatic brain injury. P7C3-A20 is thought to exert its activity through small-molecule activation of the enzyme nicotinamide phosphoribosyltransferase. This enzyme converts nicotinamide to nicotinamide mononucleotide, the precursor to nicotinamide adenine dinucleotide synthesis. Alterations to this bioenergetic pathway have been shown to induce Wallerian degeneration (WD) of the distal neurite following injury. This study aimed to establish whether P7C3-A20, through induction of nicotinamide phosphoribosyltransferase activity, would affect the rate of WD. The model systems used were dissociated primary cortical neurons, dissociated superior cervical ganglion neurons and superior cervical ganglion explants. P7C3-A20 failed to show any protection against WD induced by neurite transection or vincristine administration. Furthermore, there was a concentration-dependent neurotoxicity. These findings are important in understanding the mechanism by which P7C3-A20 mediates its effects - a key step before moving to human clinical trials.
Collapse
|
42
|
Aggarwal T, Sushmita S, Verma AK. Recent advances in the synthesis of carbazoles from indoles. Org Biomol Chem 2019; 17:8330-8342. [DOI: 10.1039/c9ob01381d] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Synthesis of carbazoles using indoles as precursors through CH activation/annulation.
Collapse
Affiliation(s)
- Trapti Aggarwal
- Synthetic Organic Chemistry Laboratory
- Department of Chemistry
- University of Delhi
- Delhi-110007
- India
| | - Sushmita Sushmita
- Synthetic Organic Chemistry Laboratory
- Department of Chemistry
- University of Delhi
- Delhi-110007
- India
| | - Akhilesh K. Verma
- Synthetic Organic Chemistry Laboratory
- Department of Chemistry
- University of Delhi
- Delhi-110007
- India
| |
Collapse
|
43
|
Banerjee A, Guin A, Saha S, Mondal A, Maji MS. Formal [4 + 2] benzannulation of 2-alkenyl indoles with aldehydes: a route to structurally diverse carbazoles and bis-carbazoles. Org Biomol Chem 2019; 17:1822-1826. [DOI: 10.1039/c8ob02875c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Construction of structurally diverse carbazoles and bis-carbazoles by protecting-group-free formal [4 + 2]-benzannulation of 2-alkenyl indoles and aldehydes is demonstrated.
Collapse
Affiliation(s)
- Ankush Banerjee
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Avishek Guin
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Shuvendu Saha
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Anushree Mondal
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Modhu Sudan Maji
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| |
Collapse
|
44
|
Makhaeva GF, Boltneva NP, Kovaleva NV, Rudakova EV, Lushchekina SV, Aksinenko AY, Sokolov VB. Influence of the γ-carboline and carbazole pharmacophore moieties on anticholinesterase and antiradical activity of multifunctional agents for the treatment of neurodegenerative diseases. Russ Chem Bull 2018. [DOI: 10.1007/s11172-018-2282-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
45
|
Voorhees JR, Remy MT, Cintrón-Pérez CJ, El Rassi E, Kahn MZ, Dutca LM, Yin TC, McDaniel LM, Williams NS, Brat DJ, Pieper AA. (-)-P7C3-S243 Protects a Rat Model of Alzheimer's Disease From Neuropsychiatric Deficits and Neurodegeneration Without Altering Amyloid Deposition or Reactive Glia. Biol Psychiatry 2018; 84:488-498. [PMID: 29246437 PMCID: PMC6415524 DOI: 10.1016/j.biopsych.2017.10.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND In addition to cognitive deficits, Alzheimer's disease (AD) is associated with other neuropsychiatric symptoms, including severe depression. Indeed, depression often precedes cognitive deficits in patients with AD. Unfortunately, the field has seen only minimal therapeutic advances, underscoring the critical need for new treatments. P7C3 aminopropyl carbazoles promote neuronal survival by enhancing nicotinamide adenine dinucleotide flux in injured neurons. Neuroprotection with P7C3 compounds has been demonstrated in preclinical models of neurodegeneration by virtue of promoting neuronal survival independently of early disease-specific pathology, resulting in protection from cognitive deficits and depressive-like behavior. We hypothesize that P7C3 compounds might be uniquely applicable to patients with AD, given the comorbid presentation of depression and cognitive deficits. METHODS Aging male and female wild-type and TgF344-AD rats, a well-characterized preclinical AD model, were administered (-)-P7C3-S243 daily for 9 and 18 months, beginning at 6 months of age. Behavioral phenotypes related to cognition and depression were assessed at 15 and 24 months, and brain pathology and biochemistry were assessed at 24 months. RESULTS (-)-P7C3-S243 safely protected aging male and female wild-type and TgF344-AD rats from cognitive deficits and depressive-like behavior. Depressive-like behavior occurred earlier than cognitive deficits in TgF344-AD rats, consistent with AD in many patients. Treatment with (-)-P7C3-S243 blocked neurodegeneration in TgF344-AD rats, without altering amyloid deposition or indicators of neuroinflammation. CONCLUSIONS Neuronal cell death-specific treatment approaches, such as P7C3 compounds, may represent a new treatment approach for patients experiencing the combination of cognitive deficits and depression associated with AD.
Collapse
Affiliation(s)
- Jaymie R Voorhees
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA,Interdisciplinary Graduate Program in Human Toxicology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Matthew T Remy
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Coral J Cintrón-Pérez
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Eli El Rassi
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Michael Z Kahn
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Laura M Dutca
- The Iowa City Department of Veterans Affairs Center for the Prevention and Treatment of Visual Loss, Iowa City, Iowa, United States Departments of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, IA, USA
| | - Terry C Yin
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Latisha M McDaniel
- Department of Psychiatry, University of Iowa Carver College of Medicine Iowa City, IA, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dan J Brat
- Departments of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Andrew A Pieper
- Department of Psychiatry, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa; Department of Neurology, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa; Free Radical and Radiation Biology Program, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa; Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa; Pappajohn Biomedical Institute, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa; Interdisciplinary Graduate Program in Human Toxicology, Department of Occupational and Environmental Health, University of Iowa College of Public Health, Iowa City, Iowa; Iowa City VA Health Care System, Iowa City, Iowa; Weill Cornell Autism Research Program, Weill Cornell Medical College New York, New York.
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Gupta S, Chandna N, Singh AK, Jain N. Regioselective Synthesis of N 2-Alkylated-1,2,3 Triazoles and N 1-Alkylated Benzotriazoles: Cu 2S as a Recyclable Nanocatalyst for Oxidative Amination of N, N-Dimethylbenzylamines. J Org Chem 2018; 83:3226-3235. [PMID: 29463081 DOI: 10.1021/acs.joc.8b00107] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Copper chalcogenide nanoparticles (Cu2S) synthesized for the first time from a single-source precursor, CuSPh, act as highly efficient and reusable heterogeneous catalyst for regioselective amination of N, N-dimethylbenzylamines with various azoles. The reaction involves N-H/C-H cross-dehydrogenative coupling (CDC) and demonstrates wide functional group tolerance. It provides highly selective access to N1-alkylated benzotriazoles, N2-alkylated 1,2,3-triazoles and 4-phenyl-1,2,3-triazoles, and N-alkylated carbazoles in 70-89% yields under solvent-free conditions. The Cu2S nanocatalyst has been characterized by PXRD, XPS, SEM-EDX, and HR-TEM analysis. Mechanistic studies suggest that the reaction follows a radical pathway and involves an iminium ion intermediate.
Collapse
Affiliation(s)
- Sonu Gupta
- Department of Chemistry , Indian Institute of Technology , New Delhi , India 110016
| | - Nisha Chandna
- Department of Chemistry , Indian Institute of Technology , New Delhi , India 110016
| | - Ajai K Singh
- Department of Chemistry , Indian Institute of Technology , New Delhi , India 110016
| | - Nidhi Jain
- Department of Chemistry , Indian Institute of Technology , New Delhi , India 110016
| |
Collapse
|
49
|
Dogan HO, Alcigir ME. The Protective effect of P7C3 against DNA and neuron damage in rat pups with congenital hypothyroidism. Biomed Pharmacother 2018; 99:499-503. [DOI: 10.1016/j.biopha.2018.01.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 10/18/2022] Open
|
50
|
Wang Z, Hu J, Yang X, Feng X, Li X, Huang L, Chan ASC. Design, Synthesis, and Evaluation of Orally Bioavailable Quinoline-Indole Derivatives as Innovative Multitarget-Directed Ligands: Promotion of Cell Proliferation in the Adult Murine Hippocampus for the Treatment of Alzheimer's Disease. J Med Chem 2018; 61:1871-1894. [PMID: 29420891 DOI: 10.1021/acs.jmedchem.7b01417] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A novel series of quinoline-indole derivatives were synthesized and evaluated as multitarget-directed ligands for the treatment of Alzheimer's disease (AD). Biological evaluation revealed that the derivatives had multifunctional profiles including antioxidant effects, blood-brain barrier (BBB) penetration, biometal chelation, Aβ aggregation modulation, neurotrophic and neuroprotective properties. Moreover, several representative target derivatives demonstrated hippocampal cell proliferation in living adult mice by intracerebroventricular (icv) injection or oral administration. Further drug-like property analysis demonstrated that the optimized compound, 8d (WI-1758), had liver microsomal metabolic stability, was well tolerated (>2000 mg/kg), and had a rational pharmacokinetic profile, as well as an oral bioavailability of 14.1% and a positive log BB (-0.19) to cross the BBB in vivo. Pharmacodynamics studies demonstrated that chronic oral administration of 8d·HCl substantially ameliorated the cognitive and spatial memory deficits in APP/PS1 AD mice and noticeably reduced overall cerebral β-amyloid deposits.
Collapse
Affiliation(s)
- Zhiren Wang
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China.,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine , Hunan Normal University , Changsha 410013 , China
| | - Jinhui Hu
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine , Hunan Normal University , Changsha 410013 , China
| | - Xing Feng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine , Hunan Normal University , Changsha 410013 , China
| | - Xingshu Li
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China
| | - Ling Huang
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China
| | - Albert S C Chan
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China
| |
Collapse
|