1
|
Khalid H, Mohamed H, Eltoukhy A, Saeed MT, Song Y. Harnessing marine resources for Alzheimer's therapy: A review integrating bioactivity and molecular docking. Eur J Pharmacol 2025; 997:177611. [PMID: 40216183 DOI: 10.1016/j.ejphar.2025.177611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition resulting in cognitive impairment and the formation of neurofibrillary tangles and plaques in the brain. The drivers of AD's molecular progression and pathology include the accumulation of amyloid β protein (Aβ); thus, Aβ is an intervention target. However, the limitations in clinical trials of Aβ-targeted medicine and the failure to intervene in disease progression have raised concerns about the use of this drug and its veracious route. In particular, we comprehensively reviewed the potential effect of marine compounds and the mechanism of isolation and extraction from marine organisms resulting in the optimization of AD treatment. Furthermore, the hub compounds were docked with Beta-secretase receptors to strengthen the extrapolation of mechanistic interactions thus inhibiting the activity of an enzyme. An extensive review revealed that marine aquaculture and its byproducts are a promising source and isolated with green methods or less investment, ensuring their sustainability. MNPs harbor specific pharmacological features that enable them to exert neuroprotective effects by minimizing events such as Aβ peptide formation and reactive oxygen species (ROS) generation.
Collapse
Affiliation(s)
- Hina Khalid
- Colin Ratledge Center of Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China.
| | - Hassan Mohamed
- Colin Ratledge Center of Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China; Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt.
| | - Adel Eltoukhy
- Colin Ratledge Center of Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China; Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt.
| | - Muhammad Tariq Saeed
- Colin Ratledge Center of Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China; Department of Diet and Nutritional Sciences, Ibadat International University, Islamabad, 45750, Pakistan.
| | - Yuanda Song
- Colin Ratledge Center of Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China; School of Basic Medicine, Qilu Medical University, Renmin West Road No. 1678, University Town, Zibo 255300, Shandong, China.
| |
Collapse
|
2
|
Cho JM, Park SK, Mookherjee S, Peters EC, Pires PW, Symons JD. Bryostatin-1 improves function in arteries with suppressed endothelial cell autophagy. GeroScience 2025:10.1007/s11357-025-01650-5. [PMID: 40220153 DOI: 10.1007/s11357-025-01650-5] [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: 01/29/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
We have previously reported that when autophagy is suppressed in endothelial cells (ECs), a glycolytic defect limits shear-stress -induced ATP production to an extent that purinergic 2Y1 receptor (P2Y1R)-mediated activation of EC nitric oxide (NO) synthase (eNOS) is compromised. Subsequently we demonstrated the functional relevance of this finding in arteries from mice with genetic, pharmacological, and age-associated EC autophagy impairment. Using gain and loss of function approaches in vitro, we further revealed that p-PKCδT505 serves as a signaling link between P2Y1R activation and NO generation. Here we sought to discern the functional relevance of this observation. First, shear-stress- induced activating phosphorylation of eNOS (p-eNOSS1177) that is otherwise prevented by knockdown of autophagy-related gene 3 (Atg3) in ECs was restored by the PKC agonist bryostatin-1. Next, in murine models of genetic and age-associated EC autophagy compromise, depressed vasodilation displayed by femoral and cerebral arteries was reversed by bryostatin-1 in a manner that could be prevented by concurrent NO synthase inhibition. Finally, the bryostatin-1-mediated normalization of intraluminal flow-induced vasodilation observed in femoral arteries from both models of EC autophagy disruption was mitigated by inhibiting downstream targets of p-PKCδT505 i.e., p-PKDS744/S748 and p-PKDS916. These findings provide evidence that stimulating PKC/PKD has strategic potential to restore compromised endothelial function in pathologies associated with suppressed EC autophagy e.g., aging.
Collapse
Affiliation(s)
- Jae Min Cho
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA.
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA.
| | - Seul-Ki Park
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - Sohom Mookherjee
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - Paulo W Pires
- Department of Physiology, University of Arizona, Tucson, AZ, USA
- Sarver Heart Center, University of Arizona Health Science Center, Tucson, AZ, USA
| | - J David Symons
- Department of Nutrition and Integrative Physiology, Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
3
|
Zhang T, Ouyang Z, Zhang Y, Sun H, Kong L, Xu Q, Qu J, Sun Y. Marine Natural Products in Inflammation-Related Diseases: Opportunities and Challenges. Med Res Rev 2025. [PMID: 40202793 DOI: 10.1002/med.22109] [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: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
In recent decades, the potentiality of marine natural products (MNPs) in the medical field has been increasingly recognized. Natural compounds derived from marine microorganisms, algae, and invertebrates have shown significant promise for treating inflammation-related diseases. In this review, we cover the three primary sources of MNPs and their diverse and unique chemical structures and bioactivities. This review aims to summarize the progress of MNPs in combating inflammation-related diseases. Moreover, we cover the functions and mechanisms of MNPs in diseases, highlighting their functions in regulating inflammatory signaling pathways, cellular stress responses, and gut microbiota, among others. Meanwhile, we focus on key technologies and scientific methods to address the current limitations and challenges in MNPs.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Yueran Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
Li L, Zhao M, van Meurs M, Brouwers-Haspels I, den Dekker RJH, Wilmsen MEP, Grashof DGB, van de Werken HJG, Rao S, Rokx C, Mueller YM, Katsikis PD. Bryostatin-1 enhances the proliferation and functionality of exhausted CD8+ T cells by upregulating MAP Kinase 11. Front Immunol 2025; 15:1509874. [PMID: 39877358 PMCID: PMC11772198 DOI: 10.3389/fimmu.2024.1509874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Bryostatin-1, a potent agonist of the protein kinase C, has been studied for HIV and cancer therapies. In HIV research, it has shown anti-HIV effects during acute infection and reactivation of latent HIV in chronic infection. As effective CD8+ T cell responses are essential for eliminating reactivated virus and achieving a cure, it is important to investigate how bryostatin-1 affects HIV-specific CD8+ T cells. HIV-specific CD8+ T cells often become exhausted, showing reduced proliferative potential and impaired cytokine production, a dysfunction also observed in cancer. Therefore, we further investigated how bryostatin-1 directly impacts exhausted CD8+ T cells. Methods PBMCs from people with HIV (PWH) were treated with bryostatin-1 and tracked with proliferation dye for cell expansion. One day 6, HIV-specific CD8+ T cells were detected by tetramers staining and examined by flow cytometry. By utilizing an established in vitro murine T cell exhaustion system, changes in inhibitory receptors, transcription factors, cytokine production and killing capacity of bryostatin-1 treated exhausted CD8+ T cells were determined by flow cytometry. RNA-seq analysis was performed to study transcriptional changes in these cells. Results We found that bryostatin-1 improved the expansion and decreased PD-1 expression of HIV-specific CD8+ T cells. Bryostatin-1 enhanced the functionality and proliferation while decreasing inhibitory receptor expression of in vitro generated exhausted CD8+ T cells. Bryostatin-1 upregulated TCF-1 and decreased TOX expression. These changes were confirmed through RNA-seq analysis. RNA-seq revealed that mitogen-activated protein kinases (MAPK) 11 was significantly downregulated in exhausted CD8+ T cells, however, it greatly upregulated after bryostatin-1 treatment. Inhibition of MAPK11 in bryostatin-1-treated cells blocked the increased proliferation and IFN-γ production induced by bryostatin-1, but did not affect other bryostatin-1 induced effects, such as the reduction of inhibitory receptors. Discussion Our data demonstrate that bryostatin-1 induces a MAPK 11-dependent improvement in the proliferative and functional capacity of exhausted T cells. This study provides a rationale for bryostatin-1's potential to help eradicate the HIV reservoir during treatment, and it may also contribute to cancer immunotherapy by functionally improving exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Ling Li
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Manzhi Zhao
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marjan van Meurs
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | | | - Merel E. P. Wilmsen
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dwin G. B. Grashof
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Shringar Rao
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Casper Rokx
- Department of Internal Medicine, and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yvonne M. Mueller
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Peter D. Katsikis
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
5
|
Shanmukha S, Godfrey WH, Gharibani P, Lee JJ, Guo Y, Deng X, Wender PA, Kornberg MD, Kim PM. TPPB modulates PKC activity to attenuate neuroinflammation and ameliorate experimental multiple sclerosis. Front Cell Neurosci 2024; 18:1373557. [PMID: 38841204 PMCID: PMC11150779 DOI: 10.3389/fncel.2024.1373557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
Protein kinase C (PKC) plays a key role in modulating the activities of the innate immune cells of the central nervous system (CNS). A delicate balance between pro-inflammatory and regenerative activities by microglia and CNS-associated macrophages is necessary for the proper functioning of the CNS. Thus, a maladaptive activation of these CNS innate immune cells results in neurodegeneration and demyelination associated with various neurologic disorders, such as multiple sclerosis (MS) and Alzheimer's disease. Prior studies have demonstrated that modulation of PKC activity by bryostatin-1 (bryo-1) and its analogs (bryologs) attenuates the pro-inflammatory processes by microglia/CNS macrophages and alleviates the neurologic symptoms in experimental autoimmune encephalomyelitis (EAE), an MS animal model. Here, we demonstrate that (2S,5S)-(E,E)-8-(5-(4-(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam (TPPB), a structurally distinct PKC modulator, has a similar effect to bryo-1 on CNS innate immune cells both in vitro and in vivo, attenuating neuroinflammation and resulting in CNS regeneration and repair. This study identifies a new structural class of PKC modulators, which can therapeutically target CNS innate immunity as a strategy to treat neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wesley H. Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Judy J. Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul A. Wender
- Departments of Chemistry and Chemical and Systems Biology, Stanford University, Stanford, CA, United States
| | - Michael D. Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul M. Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Shanmukha S, Godfrey WH, Gharibani P, Lee JJ, Guo Y, Deng X, Wender PA, Kornberg MD, Kim PM. TPPB modulates PKC activity to attenuate neuroinflammation and ameliorate experimental multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578637. [PMID: 38370818 PMCID: PMC10871289 DOI: 10.1101/2024.02.02.578637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Protein kinase C (PKC) plays a key role in modulating the activities of the innate immune cells of the central nervous system (CNS). A delicate balance between pro-inflammatory and regenerative activities by microglia and CNS-associated macrophages is necessary for the proper functioning of the CNS. Thus, a maladaptive activation of these CNS innate immune cells results in neurodegeneration and demyelination associated with various neurologic disorders, such as multiple sclerosis (MS) and Alzheimer's disease. Prior studies have demonstrated that modulation of PKC activity by bryostatin-1 (bryo-1) and its analogs (bryologs) attenuates the pro-inflammatory processes by microglia/CNS macrophages and alleviates the neurologic symptoms in experimental autoimmune encephalomyelitis (EAE), an MS animal model. Here, we demonstrate that (2S,5S)-(E,E)-8-(5-(4(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam (TPPB), a structurally distinct PKC modulator, has a similar effect to bryo-1 on CNS innate immune cells both in vitro and in vivo, attenuating neuroinflammation and resulting in CNS regeneration and repair. This study identifies a new structural class of PKC modulators, which can therapeutically target CNS innate immunity as a strategy to treat neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Wesley H. Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Judy J. Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine; Baltimore, Maryland, 21287, USA
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Paul A. Wender
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, California, 94305, USA
| | - Michael D. Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Paul M. Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| |
Collapse
|
7
|
Arora R, Babbar R, Dabra A, Chopra B, Deswal G, Grewal AS. Marine-derived Compounds: A Powerful Platform for the Treatment of Alzheimer's Disease. Cent Nerv Syst Agents Med Chem 2024; 24:166-181. [PMID: 38305396 DOI: 10.2174/0118715249269050231129103002] [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: 08/08/2023] [Revised: 09/18/2023] [Accepted: 10/31/2023] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is a debilitating form of dementia that primarily affects cholinergic neurons in the brain, significantly reducing an individual's capacity for learning and creative skills and ultimately resulting in an inability to carry out even basic daily tasks. As the elderly population is exponentially increasing, the disease has become a significant concern for society. Therefore, neuroprotective substances have garnered considerable interest in addressing this universal issue. Studies have shown that oxidative damage to neurons contributes to the pathophysiological processes underlying AD progression. In AD, tau phosphorylation and glutamate excitotoxicity may play essential roles, but no permanent cure for AD is available. The existing therapies only manage the early symptoms of AD and often come with numerous side effects and toxicities. To address these challenges, researchers have turned to nature and explored various sources such as plants, animals, and marine organisms. Many historic holy books from different cultures emphasize that adding marine compounds to the regular diet enhances brain function and mitigates its decline. Consequently, researchers have devoted significant time to identifying potentially active neuroprotective substances from marine sources. Marine-derived compounds are gaining recognition due to their abundant supply of diverse chemical compounds with biological and pharmacological potential and unique mechanisms of action. Several studies have reported that plants exhibit multitarget potential in treating AD. In light of this, the current study focuses on marine-derived components with excellent potential for treating this neurodegenerative disease.
Collapse
Affiliation(s)
- Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Ritchu Babbar
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Abhishek Dabra
- Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Bhawna Chopra
- Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Geeta Deswal
- Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | | |
Collapse
|
8
|
Wouters F, Bogie J, Wullaert A, van der Hilst J. Recent Insights in Pyrin Inflammasome Activation: Identifying Potential Novel Therapeutic Approaches in Pyrin-Associated Autoinflammatory Syndromes. J Clin Immunol 2023; 44:8. [PMID: 38129719 DOI: 10.1007/s10875-023-01621-5] [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: 08/28/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
Pyrin is a cytosolic protein encoded by the MEFV gene, predominantly expressed in innate immune cells. Upon activation, it forms an inflammasome, a multimolecular complex that enables the activation and secretion of IL-1β and IL-18. In addition, the Pyrin inflammasome activates Gasdermin D leading to pyroptosis, a highly pro-inflammatory cell death. Four autoinflammatory syndromes are associated with Pyrin inflammasome dysregulation: familial Mediterranean fever, hyper IgD syndrome/mevalonate kinase deficiency, pyrin-associated autoinflammation with neutrophilic dermatosis, and pyogenic arthritis, pyoderma gangrenosum, and acne syndrome. In this review, we discuss recent advances in understanding the molecular mechanisms regulating the two-step model of Pyrin inflammasome activation. Based on these insights, we discuss current pharmacological options and identify a series of existing molecules with therapeutic potential for the treatment of pyrin-associated autoinflammatory syndromes.
Collapse
Affiliation(s)
- Flore Wouters
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium.
| | - Jeroen Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
- University MS Center Hasselt, 3900, Pelt, Belgium
| | - Andy Wullaert
- Department of Internal Medicine and Paediatrics, Ghent University, 9052, Ghent, Belgium
- VIB-UGent Center for Inflammation Research VIB, 9052, Ghent, Belgium
- Laboratory of Protein Science, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jeroen van der Hilst
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium.
- Department of Infectious Diseases and Immune Pathology, Jessa General Hospital and Limburg Clinical Research Center, Hasselt, Belgium.
| |
Collapse
|
9
|
Silnitsky S, Rubin SJS, Zerihun M, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part I: Protein Kinase C Activation and Its Role in Cancer and Cardiovascular Diseases. Int J Mol Sci 2023; 24:17600. [PMID: 38139428 PMCID: PMC10743896 DOI: 10.3390/ijms242417600] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Protein kinases are one of the most significant drug targets in the human proteome, historically harnessed for the treatment of cancer, cardiovascular disease, and a growing number of other conditions, including autoimmune and inflammatory processes. Since the approval of the first kinase inhibitors in the late 1990s and early 2000s, the field has grown exponentially, comprising 98 approved therapeutics to date, 37 of which were approved between 2016 and 2021. While many of these small-molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP binding pocket have been massively successful for oncological indications, their poor selectively for protein kinase isozymes have limited them due to toxicities in their application to other disease spaces. Thus, recent attention has turned to the use of alternative allosteric binding mechanisms and improved drug platforms such as modified peptides to design protein kinase modulators with enhanced selectivity and other pharmacological properties. Herein we review the role of different protein kinase C (PKC) isoforms in cancer and cardiovascular disease, with particular attention to PKC-family inhibitors. We discuss translational examples and carefully consider the advantages and limitations of each compound (Part I). We also discuss the recent advances in the field of protein kinase modulators, leverage molecular docking to model inhibitor-kinase interactions, and propose mechanisms of action that will aid in the design of next-generation protein kinase modulators (Part II).
Collapse
Affiliation(s)
- Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| |
Collapse
|
10
|
Bjornsdottir G, Chalmer MA, Stefansdottir L, Skuladottir AT, Einarsson G, Andresdottir M, Beyter D, Ferkingstad E, Gretarsdottir S, Halldorsson BV, Halldorsson GH, Helgadottir A, Helgason H, Hjorleifsson Eldjarn G, Jonasdottir A, Jonasdottir A, Jonsdottir I, Knowlton KU, Nadauld LD, Lund SH, Magnusson OT, Melsted P, Moore KHS, Oddsson A, Olason PI, Sigurdsson A, Stefansson OA, Saemundsdottir J, Sveinbjornsson G, Tragante V, Unnsteinsdottir U, Walters GB, Zink F, Rødevand L, Andreassen OA, Igland J, Lie RT, Haavik J, Banasik K, Brunak S, Didriksen M, T Bruun M, Erikstrup C, Kogelman LJA, Nielsen KR, Sørensen E, Pedersen OB, Ullum H, Masson G, Thorsteinsdottir U, Olesen J, Ludvigsson P, Thorarensen O, Bjornsdottir A, Sigurdardottir GR, Sveinsson OA, Ostrowski SR, Holm H, Gudbjartsson DF, Thorleifsson G, Sulem P, Stefansson H, Thorgeirsson TE, Hansen TF, Stefansson K. Rare variants with large effects provide functional insights into the pathology of migraine subtypes, with and without aura. Nat Genet 2023; 55:1843-1853. [PMID: 37884687 PMCID: PMC10632135 DOI: 10.1038/s41588-023-01538-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 09/18/2023] [Indexed: 10/28/2023]
Abstract
Migraine is a complex neurovascular disease with a range of severity and symptoms, yet mostly studied as one phenotype in genome-wide association studies (GWAS). Here we combine large GWAS datasets from six European populations to study the main migraine subtypes, migraine with aura (MA) and migraine without aura (MO). We identified four new MA-associated variants (in PRRT2, PALMD, ABO and LRRK2) and classified 13 MO-associated variants. Rare variants with large effects highlight three genes. A rare frameshift variant in brain-expressed PRRT2 confers large risk of MA and epilepsy, but not MO. A burden test of rare loss-of-function variants in SCN11A, encoding a neuron-expressed sodium channel with a key role in pain sensation, shows strong protection against migraine. Finally, a rare variant with cis-regulatory effects on KCNK5 confers large protection against migraine and brain aneurysms. Our findings offer new insights with therapeutic potential into the complex biology of migraine and its subtypes.
Collapse
Affiliation(s)
| | - Mona A Chalmer
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | | | | | | | | | | | | | | | - Bjarni V Halldorsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Reykjavik University, School of Technology, Reykjavik, Iceland
| | - Gisli H Halldorsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Hannes Helgason
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Ingileif Jonsdottir
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Sigrun H Lund
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Physical Sciences, School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Pall Melsted
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | | | | | | | | | | | | | - Linn Rødevand
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jannicke Igland
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Department of Health and Social Science, Centre for Evidence-Based Practice, Western Norway University of Applied Science, Bergen, Norway
| | - Rolv T Lie
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Didriksen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Mie T Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine Health, Aarhus University, Aarhus, Denmark
| | - Lisette J A Kogelman
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | - Kaspar R Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ole B Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Jes Olesen
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Copenhagen, Denmark
| | - Petur Ludvigsson
- Department of Pediatrics, Landspitali University Hostpital, Reykjavik, Iceland
| | - Olafur Thorarensen
- Department of Pediatrics, Landspitali University Hostpital, Reykjavik, Iceland
| | | | | | - Olafur A Sveinsson
- Laeknasetrid Clinic, Reykjavik, Iceland
- Department of Neurology, Landspitali University Hospital, Reykjavik, Iceland
| | - Sisse R Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hilma Holm
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | - Thomas F Hansen
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
11
|
Singh RK, Kumar S, Kumar S, Shukla A, Kumar N, Patel AK, Yadav LK, Kaushalendra, Antiwal M, Acharya A. Potential implications of protein kinase Cα in pathophysiological conditions and therapeutic interventions. Life Sci 2023; 330:121999. [PMID: 37536614 DOI: 10.1016/j.lfs.2023.121999] [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: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
PKCα is a molecule with many functions that play an important role in cell survival and death to maintain cellular homeostasis. Alteration in the normal functioning of PKCα is responsible for the complicated etiology of many pathologies, including cancer, cardiovascular diseases, kidney complications, neurodegenerative diseases, diabetics, and many others. Several studies have been carried out over the years on this kinase's function, and regulation in normal physiology and pathological conditions. A lot of data with antithetical results have therefore accumulated over time to create a complex framework of physiological implications connected to the PKCα function that needs comprehensive elucidation. In light of this information, we critically analyze the multiple roles played by PKCα in basic cellular processes and their molecular mechanism during various pathological conditions. This review further discusses the current approaches to manipulating PKCα signaling amplitude in the patient's favour and proposed PKCα as a therapeutic target to reverse pathological states.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Lab of Hematopoiesis and Leukemia, KSBS, Indian Institute of Technology, Delhi, New Delhi 110016, India; Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sanjay Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sandeep Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Naveen Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Anand Kumar Patel
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Lokesh Kumar Yadav
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Kaushalendra
- Department of Zoology, Pachhunga University College Campus, Mizoram University, Aizawl 796001, India
| | - Meera Antiwal
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Arbind Acharya
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
12
|
Pérez-Vargas J, Shapira T, Olmstead AD, Villanueva I, Thompson CAH, Ennis S, Gao G, De Guzman J, Williams DE, Wang M, Chin A, Bautista-Sánchez D, Agafitei O, Levett P, Xie X, Nuzzo G, Freire VF, Quintana-Bulla JI, Bernardi DI, Gubiani JR, Suthiphasilp V, Raksat A, Meesakul P, Polbuppha I, Cheenpracha S, Jaidee W, Kanokmedhakul K, Yenjai C, Chaiyosang B, Teles HL, Manzo E, Fontana A, Leduc R, Boudreault PL, Berlinck RGS, Laphookhieo S, Kanokmedhakul S, Tietjen I, Cherkasov A, Krajden M, Nabi IR, Niikura M, Shi PY, Andersen RJ, Jean F. Discovery of lead natural products for developing pan-SARS-CoV-2 therapeutics. Antiviral Res 2023; 209:105484. [PMID: 36503013 PMCID: PMC9729583 DOI: 10.1016/j.antiviral.2022.105484] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/26/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a global public health crisis. The reduced efficacy of therapeutic monoclonal antibodies against emerging SARS-CoV-2 variants of concern (VOCs), such as omicron BA.5 subvariants, has underlined the need to explore a novel spectrum of antivirals that are effective against existing and evolving SARS-CoV-2 VOCs. To address the need for novel therapeutic options, we applied cell-based high-content screening to a library of natural products (NPs) obtained from plants, fungi, bacteria, and marine sponges, which represent a considerable diversity of chemical scaffolds. The antiviral effect of 373 NPs was evaluated using the mNeonGreen (mNG) reporter SARS-CoV-2 virus in a lung epithelial cell line (Calu-3). The screening identified 26 NPs with half-maximal effective concentrations (EC50) below 50 μM against mNG-SARS-CoV-2; 16 of these had EC50 values below 10 μM and three NPs (holyrine A, alotaketal C, and bafilomycin D) had EC50 values in the nanomolar range. We demonstrated the pan-SARS-CoV-2 activity of these three lead antivirals against SARS-CoV-2 highly transmissible Omicron subvariants (BA.5, BA.2 and BA.1) and highly pathogenic Delta VOCs in human Calu-3 lung cells. Notably, holyrine A, alotaketal C, and bafilomycin D, are potent nanomolar inhibitors of SARS-CoV-2 Omicron subvariants BA.5 and BA.2. The pan-SARS-CoV-2 activity of alotaketal C [protein kinase C (PKC) activator] and bafilomycin D (V-ATPase inhibitor) suggest that these two NPs are acting as host-directed antivirals (HDAs). Future research should explore whether PKC regulation impacts human susceptibility to and the severity of SARS-CoV-2 infection, and it should confirm the important role of human V-ATPase in the VOC lifecycle. Interestingly, we observed a synergistic action of bafilomycin D and N-0385 (a highly potent inhibitor of human TMPRSS2 protease) against Omicron subvariant BA.2 in human Calu-3 lung cells, which suggests that these two highly potent HDAs are targeting two different mechanisms of SARS-CoV-2 entry. Overall, our study provides insight into the potential of NPs with highly diverse chemical structures as valuable inspirational starting points for developing pan-SARS-CoV-2 therapeutics and for unravelling potential host factors and pathways regulating SARS-CoV-2 VOC infection including emerging omicron BA.5 subvariants.
Collapse
Affiliation(s)
- Jimena Pérez-Vargas
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Tirosh Shapira
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Andrea D Olmstead
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Ivan Villanueva
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Connor A H Thompson
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Siobhan Ennis
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Guang Gao
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Joshua De Guzman
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - David E Williams
- Departments of Chemistry and Earth, Ocean & Atmospheric Science, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Meng Wang
- Departments of Chemistry and Earth, Ocean & Atmospheric Science, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Aaleigha Chin
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Diana Bautista-Sánchez
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Olga Agafitei
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Paul Levett
- British Columbia Centre for Disease Control Public Health Laboratory, Vancouver, BC, V5Z 4R4, Canada
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Genoveffa Nuzzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, 80078, Pozzuoli, Italy
| | - Vitor F Freire
- Instituto de Química de São Carlos, Universidade de São Paulo, CP780, CEP13560-970, São Carlos, SP, Brazil
| | - Jairo I Quintana-Bulla
- Instituto de Química de São Carlos, Universidade de São Paulo, CP780, CEP13560-970, São Carlos, SP, Brazil
| | - Darlon I Bernardi
- Instituto de Química de São Carlos, Universidade de São Paulo, CP780, CEP13560-970, São Carlos, SP, Brazil
| | - Juliana R Gubiani
- Instituto de Química de São Carlos, Universidade de São Paulo, CP780, CEP13560-970, São Carlos, SP, Brazil
| | - Virayu Suthiphasilp
- Center of Chemical Innovation for Sustainability (CIS), School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Achara Raksat
- Center of Chemical Innovation for Sustainability (CIS), School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Pornphimol Meesakul
- Center of Chemical Innovation for Sustainability (CIS), School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Isaraporn Polbuppha
- Center of Chemical Innovation for Sustainability (CIS), School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | | | - Wuttichai Jaidee
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Kwanjai Kanokmedhakul
- Natural Products Research Unit, Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Chavi Yenjai
- Natural Products Research Unit, Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Boonyanoot Chaiyosang
- Natural Products Research Unit, Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Helder Lopes Teles
- Instituto de Ciências Exatas e Naturais, Universidade Federal de Rondonópolis, CEP 78736-900, Rondonópolis, MT, Brazil
| | - Emiliano Manzo
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, 80078, Pozzuoli, Italy
| | - Angelo Fontana
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry, National Research Council, Via Campi Flegrei 34, 80078, Pozzuoli, Italy; Department of Biology, Università di Napoli "Federico II", Via Cupa Nuova Cinthia 21, 80126, Napoli, Italy
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Roberto G S Berlinck
- Instituto de Química de São Carlos, Universidade de São Paulo, CP780, CEP13560-970, São Carlos, SP, Brazil
| | - Surat Laphookhieo
- Center of Chemical Innovation for Sustainability (CIS), School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Somdej Kanokmedhakul
- Natural Products Research Unit, Department of Chemistry and Center for Innovation in Chemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Ian Tietjen
- Departments of Chemistry and Earth, Ocean & Atmospheric Science, University of British Columbia, Vancouver, BC V6T 1Z1, Canada; The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Mel Krajden
- British Columbia Centre for Disease Control Public Health Laboratory, Vancouver, BC, V5Z 4R4, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Ivan Robert Nabi
- Department of Cellular and Physiological Sciences, School of Biomedical Engineering, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Raymond J Andersen
- Departments of Chemistry and Earth, Ocean & Atmospheric Science, University of British Columbia, Vancouver, BC V6T 1Z1, Canada.
| | - François Jean
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
13
|
Das R, Rauf A, Mitra S, Emran TB, Hossain MJ, Khan Z, Naz S, Ahmad B, Meyyazhagan A, Pushparaj K, Wan CC, Balasubramanian B, Rengasamy KR, Simal-Gandara J. Therapeutic potential of marine macrolides: An overview from 1990 to 2022. Chem Biol Interact 2022; 365:110072. [PMID: 35952775 DOI: 10.1016/j.cbi.2022.110072] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 01/05/2023]
Abstract
The sea is a vast ecosystem that has remained primarily unexploited and untapped, resulting in numerous organisms. Consequently, marine organisms have piqued the interest of scientists as an abundant source of natural resources with unique structural features and fascinating biological activities. Marine macrolide is a top-class natural product with a heavily oxygenated polyene backbone containing macrocyclic lactone. In the last few decades, significant efforts have been made to isolate and characterize macrolides' chemical and biological properties. Numerous macrolides are extracted from different marine organisms such as marine microorganisms, sponges, zooplankton, molluscs, cnidarians, red algae, tunicates, and bryozoans. Notably, the prominent macrolide sources are fungi, dinoflagellates, and sponges. Marine macrolides have several bioactive characteristics such as antimicrobial (antibacterial, antifungal, antimalarial, antiviral), anti-inflammatory, antidiabetic, cytotoxic, and neuroprotective activities. In brief, marine organisms are plentiful in naturally occurring macrolides, which can become the source of efficient and effective therapeutics for many diseases. This current review summarizes these exciting and promising novel marine macrolides in biological activities and possible therapeutic applications.
Collapse
Affiliation(s)
- Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, 94640, Pakistan.
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
| | - Md Jamal Hossain
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka, 1205, Bangladesh.
| | - Zidan Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh.
| | - Saima Naz
- Department of Biotechnology, Bacha Khan University, Charsadda, KPK, Pakistan.
| | - Bashir Ahmad
- Department of Biotechnology, Bacha Khan University, Charsadda, KPK, Pakistan.
| | - Arun Meyyazhagan
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru, Karnataka, 560076, India.
| | - Karthika Pushparaj
- Department of Zoology, School of Biosciences, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, 641 043, Tamil Nadu, India.
| | - Chunpeng Craig Wan
- Jiangxi Key Laboratory for Postharvest Technology and Nondestructive Testing of Fruit &Vegetables, Collaborative Innovation Center of Postharvest Key Technology and Quality Safety of Fruit & Vegetables, College of Agronomy, Jiangxi Agricultural University Nanchang, 330045, Jiangxi, China.
| | | | - Kannan Rr Rengasamy
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, 600077, India.
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E-32004 Ourense, Spain.
| |
Collapse
|
14
|
Barman B, Kushwaha A, Thakur MK. Muscarinic Acetylcholine Receptors-Mediated Activation of PKC Restores the Hippocampal Immediate Early Gene Expression and CREB Phosphorylation in Scopolamine-Induced Amnesic Mice. Mol Neurobiol 2022; 59:5722-5733. [DOI: 10.1007/s12035-022-02940-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/23/2022] [Indexed: 10/17/2022]
|
15
|
Hemmerling F, Piel J. Strategies to access biosynthetic novelty in bacterial genomes for drug discovery. Nat Rev Drug Discov 2022; 21:359-378. [PMID: 35296832 DOI: 10.1038/s41573-022-00414-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 12/17/2022]
Abstract
Bacteria provide a rich source of natural products with potential therapeutic applications, such as novel antibiotic classes or anticancer drugs. Bioactivity-guided screening of bacterial extracts and characterization of biosynthetic pathways for drug discovery is now complemented by the availability of large (meta)genomic collections, placing researchers into the postgenomic, big-data era. The progress in next-generation sequencing and the rise of powerful computational tools provide unprecedented insights into unexplored taxa, ecological niches and 'biosynthetic dark matter', revealing diverse and chemically distinct natural products in previously unstudied bacteria. In this Review, we discuss such sources of new chemical entities and the implications for drug discovery with a particular focus on the strategies that have emerged in recent years to identify and access novelty.
Collapse
Affiliation(s)
- Franziska Hemmerling
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland
| | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland.
| |
Collapse
|
16
|
Sanapala P, Pola S, Nageswara Rao Reddy N, Pallaval VB. Expanding Role of Marine Natural Compounds in Immunomodulation: Challenges and Future Perspectives. MARINE BIOMATERIALS 2022:307-349. [DOI: 10.1007/978-981-16-5374-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Targeting PKC in microglia to promote remyelination and repair in the CNS. Curr Opin Pharmacol 2021; 62:103-108. [PMID: 34965482 DOI: 10.1016/j.coph.2021.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 01/28/2023]
Abstract
Microglia and CNS-infiltrating macrophages play significant roles in the pathogenesis of neuroinflammatory and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Prolonged and dysregulated inflammatory responses by these innate immune cells can have deleterious effects on the surrounding CNS microenvironment, which can worsen neurodegeneration and demyelination. However, although chronic activation of pro-inflammatory microglia is maladaptive, other functional microglial subtypes play beneficial roles during CNS repair and regeneration. Therefore, there is a tremendous interest in understanding the underlying mechanism of the activation of these reparative/regenerative microglia. In this review, we focus on the potential role of PKC, a downstream signaling molecule of TREM2 and PLCγ2, and PKC modulators in promoting the activation of reparative/regenerative microglial subtypes as a novel therapy for neuroinflammatory and neurodegenerative diseases.
Collapse
|
18
|
Singh RK, Kumar S, Tomar MS, Verma PK, Kumar A, Kumar S, Kumar N, Singh JP, Acharya A. Putative role of natural products as Protein Kinase C modulator in different disease conditions. ACTA ACUST UNITED AC 2021; 29:397-414. [PMID: 34216003 DOI: 10.1007/s40199-021-00401-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Protein kinase C (PKC) is a promising drug target for various therapeutic areas. Natural products derived from plants, animals, microorganisms, and marine organisms have been used by humans as medicine from prehistoric times. Recently, several compounds derived from plants have been found to modulate PKC activities through competitive binding with ATP binding site, and other allosteric regions of PKC. As a result fresh race has been started in academia and pharmaceutical companies to develop an effective naturally derived small-molecule inhibitor to target PKC activities. Herein, in this review, we have discussed several natural products and their derivatives, which are reported to have an impact on PKC signaling cascade. METHODS All information presented in this review article regarding the regulation of PKC by natural products has been acquired by a systematic search of various electronic databases, including ScienceDirect, Scopus, Google Scholar, Web of science, ResearchGate, and PubMed. The keywords PKC, natural products, curcumin, rottlerin, quercetin, ellagic acid, epigallocatechin-3 gallate, ingenol 3 angelate, resveratrol, protocatechuic acid, tannic acid, PKC modulators from marine organism, bryostatin, staurosporine, midostaurin, sangivamycin, and other relevant key words were explored. RESULTS The natural products and their derivatives including curcumin, rottlerin, quercetin, ellagic acid, epigallocatechin-3 gallate, ingenol 3 angelate, resveratrol, bryostatin, staurosporine, and midostaurin play a major role in the management of PKC activity during various disease progression. CONCLUSION Based on the comprehensive literature survey, it could be concluded that various natural products can regulate PKC activity during disease progression. However, extensive research is needed to circumvent the challenge of isoform specific regulation of PKC by natural products.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | | | - Munendra Singh Tomar
- Department of Pharmaceutical Science, School of Pharmacy, University of Colorado, Denver, USA
| | | | - Amit Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Sandeep Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Naveen Kumar
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India
| | - Jai Prakash Singh
- Department of Panchkarma, Institute of Medical Science, BHU, Varanasi, India, 221005
| | - Arbind Acharya
- Department of Zoology, Institute of Science, BHU, Varanasi, 221005, India.
| |
Collapse
|
19
|
Silva M, Seijas P, Otero P. Exploitation of Marine Molecules to Manage Alzheimer's Disease. Mar Drugs 2021; 19:md19070373. [PMID: 34203244 PMCID: PMC8307759 DOI: 10.3390/md19070373] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are sociosanitary challenges of today, as a result of increased average life expectancy, with Alzheimer’s disease being one of the most prevalent. This pathology is characterized by brain impairment linked to a neurodegenerative process culminating in cognitive decline and behavioral disorders. Though the etiology of this pathology is still unknown, it is usually associated with the appearance of senile plaques and neurofibrillary tangles. The most used prophylaxis relies on anticholinesterase drugs and NMDA receptor antagonists, whose main action is to relieve symptoms and not to treat or prevent the disease. Currently, the scientific community is gathering efforts to disclose new natural compounds effective against Alzheimer’s disease and other neurodegenerative pathologies. Marine natural products have been shown to be promising candidates, and some have been proven to exert a high neuroprotection effect, constituting a large reservoir of potential drugs and nutraceutical agents. The present article attempts to describe the processes of extraction and isolation of bioactive compounds derived from sponges, algae, marine bacteria, invertebrates, crustaceans, and tunicates as drug candidates against AD, with a focus on the success of pharmacological activity in the process of finding new and effective drug compounds.
Collapse
Affiliation(s)
- Marisa Silva
- MARE—Marine and Environmental Sciences Centre, Faculty of Sciences, University of Lisbon, Campo Grande, 1749-016 Lisbon, Portugal;
- Department of Plant Biology, Faculty of Science, University of Lisbon, Campo Grande, 1749-016 Lisbon, Portugal
| | - Paula Seijas
- Department of Pharmacology, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Paz Otero
- Department of Pharmacology, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research (CIAL), Campus of International Excellence UAM+CSIC, 28049 Madrid, Spain
- Nutrition and Bromatology Group, CITACA, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, 32004 Ourense, Spain
- Correspondence: or
| |
Collapse
|
20
|
Lenz KD, Klosterman KE, Mukundan H, Kubicek-Sutherland JZ. Macrolides: From Toxins to Therapeutics. Toxins (Basel) 2021; 13:347. [PMID: 34065929 PMCID: PMC8150546 DOI: 10.3390/toxins13050347] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/17/2022] Open
Abstract
Macrolides are a diverse class of hydrophobic compounds characterized by a macrocyclic lactone ring and distinguished by variable side chains/groups. Some of the most well characterized macrolides are toxins produced by marine bacteria, sea sponges, and other species. Many marine macrolide toxins act as biomimetic molecules to natural actin-binding proteins, affecting actin polymerization, while other toxins act on different cytoskeletal components. The disruption of natural cytoskeletal processes affects cell motility and cytokinesis, and can result in cellular death. While many macrolides are toxic in nature, others have been shown to display therapeutic properties. Indeed, some of the most well known antibiotic compounds, including erythromycin, are macrolides. In addition to antibiotic properties, macrolides have been shown to display antiviral, antiparasitic, antifungal, and immunosuppressive actions. Here, we review each functional class of macrolides for their common structures, mechanisms of action, pharmacology, and human cellular targets.
Collapse
Affiliation(s)
| | | | | | - Jessica Z. Kubicek-Sutherland
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA; (K.D.L.); (K.E.K.); (H.M.)
| |
Collapse
|
21
|
Abramson E, Hardman C, Shimizu AJ, Hwang S, Hester LD, Snyder SH, Wender PA, Kim PM, Kornberg MD. Designed PKC-targeting bryostatin analogs modulate innate immunity and neuroinflammation. Cell Chem Biol 2021; 28:537-545.e4. [PMID: 33472023 PMCID: PMC8052272 DOI: 10.1016/j.chembiol.2020.12.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/02/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammation characterizes multiple neurologic diseases, including primary inflammatory conditions such as multiple sclerosis and classical neurodegenerative diseases. Aberrant activation of the innate immune system contributes to disease progression, but drugs modulating innate immunity, particularly within the central nervous system (CNS), are lacking. The CNS-penetrant natural product bryostatin-1 attenuates neuroinflammation by targeting innate myeloid cells. Supplies of natural bryostatin-1 are limited, but a recent scalable good manufacturing practice (GMP) synthesis has enabled access to it and its analogs (bryologs), the latter providing a path to more efficacious, better tolerated, and more accessible agents. Here, we show that multiple synthetically accessible bryologs replicate the anti-inflammatory effects of bryostatin-1 on innate immune cells in vitro, and a lead bryolog attenuates neuroinflammation in vivo, actions mechanistically dependent on protein kinase C (PKC) binding. Our findings identify bryologs as promising drug candidates for targeting innate immunity in neuroinflammation and create a platform for evaluation of synthetic PKC modulators in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Efrat Abramson
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Clayton Hardman
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Akira J Shimizu
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Soonmyung Hwang
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Lynda D Hester
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Solomon H Snyder
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD 21287, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Paul A Wender
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Paul M Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD 21287, USA.
| | - Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA.
| |
Collapse
|
22
|
Marsillo A, David L, Gerges B, Kerr D, Sadek R, Lasiychuk V, Salame D, Soliman Y, Menkes S, Chatterjee A, Mancuso A, Banerjee P. PKC epsilon as a neonatal target to correct FXS-linked AMPA receptor translocation in the hippocampus, boost PVN oxytocin expression, and normalize adult behavior in Fmr1 knockout mice. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166048. [PMID: 33359697 DOI: 10.1016/j.bbadis.2020.166048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Fragile X Syndrome (FXS) is an inherited developmental disorder caused by the non-expression of the Fmr1 gene. FXS is associated with abnormal social and anxiety behavior that is more prominent among males. Given that oxytocin (OXT) regulates both social and anxiety behavior, we studied the effect of FXS in the hypothalamic paraventricular nucleus (PVN), the major central source of OXT. We observed a significant suppression of protein kinase C epsilon (PKCε) (34%) in the ventral hippocampal CA1 region of postnatal day-18 (P18) male Fmr1 knockout (KO) mice, which displayed social behavior deficits and hyper-anxiety in adulthood. These mice also displayed a 39% increase in cell surface α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor (AMPAR) at P18 (measured by the surface level of the AMPAR subunit GluR2), thereby indicating excitation of the CA1 neurons. It is known that neuronal activation at CA1 is linked to an inhibition of the PVN neurons. As expected, these mice also displayed a 25% suppression of oxytocin+ (OXT+) cells in the PVN at P20. Stimulating PKCε during postnatal days 6-,14 (P6-14) mice using a selective activator, dicyclopropyl-linoleic acid (DCP-LA), corrected AMPAR externalization in CA1 and suppression of OXT+ cell number in PVN in a PKCε dependent manner. Most notably, neonatal DCP-LA treatment rescued social behavior deficits and hyper-anxiety, displayed by adult (≥P60) male but not female KO mice. Thus, neonatal stimulation of PKCε could be a strategy to correct endophenotypic anomalies during brain development and aberrant adult behavior of the FXS males to the wild-type levels.
Collapse
Affiliation(s)
- Alexandra Marsillo
- CUNY Doctoral Programs in Biology, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Lovena David
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Bishoy Gerges
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Daniel Kerr
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Rodina Sadek
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Vitaliy Lasiychuk
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - David Salame
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Youstina Soliman
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Silvia Menkes
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Aheli Chatterjee
- Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Andrew Mancuso
- CUNY Doctoral Programs in Biochemistry, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America
| | - Probal Banerjee
- CUNY Doctoral Programs in Biology, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America; Department of Chemistry, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America; Center for Developmental Neuroscience, The College of Staten Island (CUNY), Staten Island, NY 10314-6609, United States of America.
| |
Collapse
|
23
|
Avila C, Angulo-Preckler C. Bioactive Compounds from Marine Heterobranchs. Mar Drugs 2020; 18:657. [PMID: 33371188 PMCID: PMC7767343 DOI: 10.3390/md18120657] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/22/2022] Open
Abstract
The natural products of heterobranch molluscs display a huge variability both in structure and in their bioactivity. Despite the considerable lack of information, it can be observed from the recent literature that this group of animals possesses an astonishing arsenal of molecules from different origins that provide the molluscs with potent chemicals that are ecologically and pharmacologically relevant. In this review, we analyze the bioactivity of more than 450 compounds from ca. 400 species of heterobranch molluscs that are useful for the snails to protect themselves in different ways and/or that may be useful to us because of their pharmacological activities. Their ecological activities include predator avoidance, toxicity, antimicrobials, antifouling, trail-following and alarm pheromones, sunscreens and UV protection, tissue regeneration, and others. The most studied ecological activity is predation avoidance, followed by toxicity. Their pharmacological activities consist of cytotoxicity and antitumoral activity; antibiotic, antiparasitic, antiviral, and anti-inflammatory activity; and activity against neurodegenerative diseases and others. The most studied pharmacological activities are cytotoxicity and anticancer activities, followed by antibiotic activity. Overall, it can be observed that heterobranch molluscs are extremely interesting in regard to the study of marine natural products in terms of both chemical ecology and biotechnology studies, providing many leads for further detailed research in these fields in the near future.
Collapse
Affiliation(s)
- Conxita Avila
- Department of Evolutionary Biology, Ecology, and Environmental Sciences, Biodiversity Research Institute (IrBIO), Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Catalonia, Spain;
| | - Carlos Angulo-Preckler
- Department of Evolutionary Biology, Ecology, and Environmental Sciences, Biodiversity Research Institute (IrBIO), Faculty of Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Catalonia, Spain;
- Norwegian College of Fishery Science, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9019 Tromsø, Norway
| |
Collapse
|
24
|
Shikov AN, Flisyuk EV, Obluchinskaya ED, Pozharitskaya ON. Pharmacokinetics of Marine-Derived Drugs. Mar Drugs 2020; 18:E557. [PMID: 33182407 PMCID: PMC7698100 DOI: 10.3390/md18110557] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
Marine organisms represent an excellent source of innovative compounds that have the potential for the development of new drugs. The pharmacokinetics of marine drugs has attracted increasing interest in recent decades due to its effective and potential contribution to the selection of rational dosage recommendations and the optimal use of the therapeutic arsenal. In general, pharmacokinetics studies how drugs change after administration via the processes of absorption, distribution, metabolism, and excretion (ADME). This review provides a summary of the pharmacokinetics studies of marine-derived active compounds, with a particular focus on their ADME. The pharmacokinetics of compounds derived from algae, crustaceans, sea cucumber, fungus, sea urchins, sponges, mollusks, tunicate, and bryozoan is discussed, and the pharmacokinetics data in human experiments are analyzed. In-depth characterization using pharmacokinetics is useful for obtaining information for understanding the molecular basis of pharmacological activity, for correct doses and treatment schemes selection, and for more effective drug application. Thus, an increase in pharmacokinetic research on marine-derived compounds is expected in the near future.
Collapse
Affiliation(s)
- Alexander N. Shikov
- Department of Technology of Pharmacutical Formulations, St. Petersburg State Chemical Pharmaceutical University, Prof. Popov, 14a, Saint-Petersburg 197376, Russia;
- Murmansk Marine Biological Institute of the Russian Academy of Sciences (MMBI RAS), Vladimirskaya, 17, Murmansk 183010, Russia; (E.D.O.); (O.N.P.)
| | - Elena V. Flisyuk
- Department of Technology of Pharmacutical Formulations, St. Petersburg State Chemical Pharmaceutical University, Prof. Popov, 14a, Saint-Petersburg 197376, Russia;
| | - Ekaterina D. Obluchinskaya
- Murmansk Marine Biological Institute of the Russian Academy of Sciences (MMBI RAS), Vladimirskaya, 17, Murmansk 183010, Russia; (E.D.O.); (O.N.P.)
| | - Olga N. Pozharitskaya
- Murmansk Marine Biological Institute of the Russian Academy of Sciences (MMBI RAS), Vladimirskaya, 17, Murmansk 183010, Russia; (E.D.O.); (O.N.P.)
| |
Collapse
|
25
|
Meenambal R, Srinivas Bharath MM. Nanocarriers for effective nutraceutical delivery to the brain. Neurochem Int 2020; 140:104851. [PMID: 32976906 DOI: 10.1016/j.neuint.2020.104851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders are common among aging populations around the globe. Most are characterized by loss of neurons, protein aggregates, oxidative stress, mitochondrial damage, neuroinflammation among others. Although symptomatic treatment using conventional pharmacotherapy has been widely employed, their therapeutic success is limited due to varied reasons. In the need to identify an alternative approach, researchers successfully demonstrated the therapeutic utility of plant-derived nutraceuticals in cell and animal models of neurodegenerative conditions. However, most nutraceuticals failed during clinical trials in humans owing to their poor bioavailability in vivo and limited permeability across the blood brain barrier (BBB). The current emphasis is therefore on the improved delivery of nutraceuticals to the brain. In this regard, development of nanoparticle conjugated nutraceuticals to enhance bioavailability and therapeutic efficacy in the brain has gained attention. Here, we review the research advances in nanoparticles conjugated nutraceuticals applied in neurodegenerative disorders and discuss their advantages and limitations, clinical trials and toxicity concerns.
Collapse
Affiliation(s)
- Rugmani Meenambal
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India.
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India; Neurotoxicology Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India.
| |
Collapse
|
26
|
Martins M, Silva R, M. M. Pinto M, Sousa E. Marine Natural Products, Multitarget Therapy and Repurposed Agents in Alzheimer's Disease. Pharmaceuticals (Basel) 2020; 13:E242. [PMID: 32933034 PMCID: PMC7558913 DOI: 10.3390/ph13090242] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial disease characterized by the presence of amyloid plaques, neurofibrillary tangles, and nerve cell death that affects, mainly, older people. After decades of investigation, the search for an efficacious treatment for AD remains and several strategies can be and are being employed in this journey. In this review, four of the most promising strategies, alongside with its most promising agents under investigation or development are highlighted. Marine natural products (MNP) are a source of unique chemical structures with useful biological activities for AD treatment. One of the most promising compounds, a marine-derived acidic oligosaccharide (GV-971) just passed phase III clinical trials with a unique mechanism of action. Combination therapy and multitargeted-directed ligand therapy (MTDL) are also two important strategies, with several examples in clinical trials, based on the belief that the best approach for AD is a therapy capable of modulating multiple target pathways. Drug repurposing, a strategy that requires a smaller investment and is less time consuming, is emerging as a strong contender with a variety of pharmacological agents resurfacing in an attempt to identify a therapeutic candidate capable of modifying the course of this disease.
Collapse
Affiliation(s)
- Márcia Martins
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Madalena M. M. Pinto
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
27
|
Wei J, Gou Z, Wen Y, Luo Q, Huang Z. Marine compounds targeting the PI3K/Akt signaling pathway in cancer therapy. Biomed Pharmacother 2020; 129:110484. [PMID: 32768966 DOI: 10.1016/j.biopha.2020.110484] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/18/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer is a disease characterized by overproliferation, including that due to transformation, apoptosis disorders, proliferation, invasion, angiogenesis and metastasis, and is one of the deadliest diseases. Currently, conservative chemotherapy is used for cancer treatment due to a lack of effective drugs. The PI3K/Akt signaling pathway plays a very essential role in the pathogenesis of many cancers, and abnormal activation of this pathway leads to abnormal expression of a series of downstream proteins, which ultimately results in the excessive proliferation of cancer cells. Therefore, the PI3K/Akt signaling pathway is a critical target in cancer treatment. Marine drugs have attracted much attention in recent years, and studies have found that many extracts from oceanic animals, plants and microorganisms or their metabolites exert antitumor effects, including antiproliferative effects or the induction of cell cycle arrest, apoptosis or autophagy. However, most anticancer targets and the mechanisms of marine compounds remain unclear. The great potential of the development of marine drugs provides a new direction for cancer treatment. This review focuses on marine compounds that target the PI3K/Akt signaling pathway for the prevention and treatment of cancer and provides comprehensive information for those interested in research on marine drugs.
Collapse
Affiliation(s)
- Jiaen Wei
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Zhanping Gou
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Ying Wen
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Qiaohong Luo
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Zunnan Huang
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Guangdong Medical University, Dongguan, Guangdong 523808, China; Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
28
|
Ciavatta ML, Lefranc F, Vieira LM, Kiss R, Carbone M, van Otterlo WAL, Lopanik NB, Waeschenbach A. The Phylum Bryozoa: From Biology to Biomedical Potential. Mar Drugs 2020; 18:E200. [PMID: 32283669 PMCID: PMC7230173 DOI: 10.3390/md18040200] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/31/2020] [Accepted: 04/06/2020] [Indexed: 01/06/2023] Open
Abstract
Less than one percent of marine natural products characterized since 1963 have been obtained from the phylum Bryozoa which, therefore, still represents a huge reservoir for the discovery of bioactive metabolites with its ~6000 described species. The current review is designed to highlight how bryozoans use sophisticated chemical defenses against their numerous predators and competitors, and which can be harbored for medicinal uses. This review collates all currently available chemoecological data about bryozoans and lists potential applications/benefits for human health. The core of the current review relates to the potential of bryozoan metabolites in human diseases with particular attention to viral, brain, and parasitic diseases. It additionally weighs the pros and cons of total syntheses of some bryozoan metabolites versus the synthesis of non-natural analogues, and explores the hopes put into the development of biotechnological approaches to provide sustainable amounts of bryozoan metabolites without harming the natural environment.
Collapse
Affiliation(s)
- Maria Letizia Ciavatta
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Chimica Biomolecolare (ICB), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (M.L.C.); (M.C.)
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Leandro M. Vieira
- Departamento de Zoologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil;
| | - Robert Kiss
- Retired – formerly at the Fonds National de la Recherche Scientifique (FRS-FNRS), 1000 Brussels, Belgium;
| | - Marianna Carbone
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Chimica Biomolecolare (ICB), Via Campi Flegrei 34, 80078 Pozzuoli, Italy; (M.L.C.); (M.C.)
| | - Willem A. L. van Otterlo
- Department of Chemistry and Polymer Science, University of Stellenbosch, Private Bag X1, Matieland 7602, South Africa;
| | - Nicole B. Lopanik
- School of Earth and Atmospheric Sciences, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW In addition to preventive protocols and antiretroviral therapy, HIV-1 eradication has been considered as an additional strategy to help fight the AIDS epidemic. With the support of multiple funding agencies, research groups worldwide have been developing protocols to achieve either a sterilizing or a functional cure for HIV-infection. RECENT FINDINGS Most of the studies focus on the elimination or suppression of circulating CD4+ T cells, the best characterized HIV-1 latent reservoir. The role of the central nervous system (CNS) as a latent reservoir is still controversial. Although brain macrophages and astrocytes are susceptible to HIV-1 infection, it has not been ascertained whether the CNS carries latent HIV-1 during cART and, if so, whether the virus can be reactivated and spread to other compartments after ART interruption. Here, we examine the implications of HIV-1 eradication strategies on the CNS, regardless of whether it is a true latent reservoir and, if so, whether it is present in all patients.
Collapse
|
30
|
Opposed Actions of PKA Isozymes (RI and RII) and PKC Isoforms (cPKCβI and nPKCε) in Neuromuscular Developmental Synapse Elimination. Cells 2019; 8:cells8111304. [PMID: 31652775 PMCID: PMC6912401 DOI: 10.3390/cells8111304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background: During neuromuscular junction (NMJ) development, synapses are produced in excess. By sensing the activity-dependent release of ACh, adenosine, and neurotrophins, presynaptic receptors prompt axonal competition and loss of the unnecessary axons. The receptor action is mediated by synergistic and antagonistic relations when they couple to downstream kinases (mainly protein kinases A and C (PKA and PKC)), which phosphorylate targets involved in axonal disconnection. Here, we directly investigated the involvement of PKA subunits and PKC isoforms in synapse elimination. Methods: Selective PKA and PKC peptide modulators were applied daily to the Levator auris longus (LAL) muscle surface of P5–P8 transgenic B6.Cg-Tg (Thy1-YFP) 16 Jrs/J (and also C57BL/6J) mice, and the number of axons and the postsynaptic receptor cluster morphology were evaluated in P9 NMJ. Results: PKA (PKA-I and PKA-II isozymes) acts at the pre- and postsynaptic sites to delay both axonal elimination and nAChR cluster differentiation, PKC activity promotes both axonal loss (a cPKCβI and nPKCε isoform action), and postsynaptic nAChR cluster maturation (a possible role for PKCθ). Moreover, PKC-induced changes in axon number indirectly influence postsynaptic maturation. Conclusions: PKC and PKA have opposed actions, which suggests that changes in the balance of these kinases may play a major role in the mechanism of developmental synapse elimination.
Collapse
|
31
|
Blanco FA, Czikora A, Kedei N, You Y, Mitchell GA, Pany S, Ghosh A, Blumberg PM, Das J. Munc13 Is a Molecular Target of Bryostatin 1. Biochemistry 2019; 58:3016-3030. [PMID: 31243993 PMCID: PMC6620733 DOI: 10.1021/acs.biochem.9b00427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Bryostatin
1 is a natural macrolide shown to improve neuronal connections and
enhance memory in mice. Its mechanism of action is largely attributed
to the modulation of novel and conventional protein kinase Cs (PKCs)
by binding to their regulatory C1 domains. Munc13-1 is a C1 domain-containing
protein that shares common endogenous and exogenous activators with
novel and conventional PKC subtypes. Given the essential role of Munc13-1
in the priming of synaptic vesicles and neuronal transmission overall,
we explored the potential interaction between bryostatin 1 and Munc13-1.
Our results indicate that in vitro bryostatin 1 binds
to both the isolated C1 domain of Munc13-1 (Ki = 8.07 ± 0.90 nM) and the full-length Munc13-1 protein
(Ki = 0.45 ± 0.04 nM). Furthermore,
confocal microscopy and immunoblot analysis demonstrated that in intact
HT22 cells bryostatin 1 mimics the actions of phorbol esters, a previously
established class of Munc13-1 activators, and induces plasma membrane
translocation of Munc13-1, a hallmark of its activation. Consistently,
bryostatin 1 had no effect on the Munc13-1H567K construct
that is insensitive to phorbol esters. Effects of bryostatin 1 on
the other Munc13 family members, ubMunc13-2 and bMunc13-2, resembled
those of Munc13-1 for translocation. Lastly, we observed an increased
level of expression of Munc13-1 following a 24 h incubation with bryostatin
1 in both HT22 and primary mouse hippocampal cells. This study characterizes
Munc13-1 as a molecular target of bryostatin 1. Considering the crucial
role of Munc13-1 in neuronal function, these findings provide strong
support for the potential role of Munc13s in the actions of bryostatin
1.
Collapse
Affiliation(s)
- Francisco A Blanco
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy , University of Houston , Houston , Texas 77204 , United States
| | - Agnes Czikora
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Noemi Kedei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Youngki You
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy , University of Houston , Houston , Texas 77204 , United States
| | - Gary A Mitchell
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Satyabrata Pany
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy , University of Houston , Houston , Texas 77204 , United States
| | - Anamitra Ghosh
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy , University of Houston , Houston , Texas 77204 , United States
| | - Peter M Blumberg
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Joydip Das
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy , University of Houston , Houston , Texas 77204 , United States
| |
Collapse
|
32
|
Yang XA, Zweifach A. Temperature-Dependent Expression of a CFP-YFP FRET Diacylglycerol Sensor Enables Multiple-Read Screening for Compounds That Affect C1 Domains. SLAS DISCOVERY 2019; 24:682-692. [PMID: 30802416 DOI: 10.1177/2472555219830086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intramolecular CFP-YFP fluorescence resonance energy transfer (FRET) sensors expressed in cells are powerful research tools but have seen relatively little use in screening. We exploited the discovery that the expression of a CFP-YFP FRET diacylglycerol sensor (DAGR) increases over time when cells are incubated at room temperature to assess requirements for robust measurements using a Molecular Devices Spectramax i3x fluorescence plate reader. Expression levels resulting in YFP fluorescence >10-fold higher than untransfected cells and phorbol ester-stimulated FRET ratio changes of 60% or more were required to consistently give robust Z' > 0.5. As a means of confirming that these conditions are suitable for screening, we developed a novel multiple-read protocol to assay the NCI's Mechanistic Set III for agonists and antagonists of C1 domain activation. Sixteen compounds prevented C1 domain translocation. However, none blocked phorbol ester-stimulated protein kinase C (PKC) activity assessed using a phospho-specific antibody-six actually stimulated PKC activity. Cytometry, which produces higher Z' for a given FRET ratio change, might have been a better approach for discovering antagonists, as it would have allowed lower phorbol ester concentrations to be used. We conclude that CFP-YFP FRET measured in a Spectramax i3x plate reader can be used for screening under the conditions we defined. Our strategy of varying expression level and FRET ratio could be useful to others for determining conditions needed for robust cell-based intramolecular CFP-YFP FRET measurements on their instrumentation.
Collapse
Affiliation(s)
- Xiuyi Alexander Yang
- 1 Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT, USA
| | - Adam Zweifach
- 1 Department of Molecular and Cell Biology, University of Connecticut at Storrs, Storrs, CT, USA
| |
Collapse
|
33
|
Plisson F, Piggott AM. Predicting Blood⁻Brain Barrier Permeability of Marine-Derived Kinase Inhibitors Using Ensemble Classifiers Reveals Potential Hits for Neurodegenerative Disorders. Mar Drugs 2019; 17:E81. [PMID: 30699889 PMCID: PMC6410078 DOI: 10.3390/md17020081] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/19/2022] Open
Abstract
The recent success of small-molecule kinase inhibitors as anticancer drugs has generated significant interest in their application to other clinical areas, such as disorders of the central nervous system (CNS). However, most kinase inhibitor drug candidates investigated to date have been ineffective at treating CNS disorders, mainly due to poor blood⁻brain barrier (BBB) permeability. It is, therefore, imperative to evaluate new chemical entities for both kinase inhibition and BBB permeability. Over the last 35 years, marine biodiscovery has yielded 471 natural products reported as kinase inhibitors, yet very few have been evaluated for BBB permeability. In this study, we revisited these marine natural products and predicted their ability to cross the BBB by applying freely available open-source chemoinformatics and machine learning algorithms to a training set of 332 previously reported CNS-penetrant small molecules. We evaluated several regression and classification models, and found that our optimised classifiers (random forest, gradient boosting, and logistic regression) outperformed other models, with overall cross-validated model accuracies of 80%⁻82% and 78%⁻80% on external testing. All 3 binary classifiers predicted 13 marine-derived kinase inhibitors with appropriate physicochemical characteristics for BBB permeability.
Collapse
Affiliation(s)
- Fabien Plisson
- CONACYT, Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Centro de Investigación y de Estudios Avanzados del IPN, Irapuato, Guanajuato 36824, Mexico.
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Andrew M Piggott
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
34
|
Lanni C, Fagiani F, Racchi M, Preda S, Pascale A, Grilli M, Allegri N, Govoni S. Beta-amyloid short- and long-term synaptic entanglement. Pharmacol Res 2019; 139:243-260. [DOI: 10.1016/j.phrs.2018.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
|
35
|
Safaeinejad F, Bahrami S, Redl H, Niknejad H. Inhibition of Inflammation, Suppression of Matrix Metalloproteinases, Induction of Neurogenesis, and Antioxidant Property Make Bryostatin-1 a Therapeutic Choice for Multiple Sclerosis. Front Pharmacol 2018; 9:625. [PMID: 29971003 PMCID: PMC6018466 DOI: 10.3389/fphar.2018.00625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/24/2018] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease characterized by inflammation and myelin damage. Pro-inflammatory cytokines, oxidative stress, high level of matrix metalloproteinases (MMPs) activity and blood-brain barrier (BBB) damage, immune-mediated destruction of myelin and neuron loss are involved in the pathogenesis of MS. The currently approved treatments for MS include injectable drugs (interferon-β and glatiramer acetate), oral drugs (fingolimod), and monoclonal antibodies (natalizumab). The mentioned therapeutic choices are mostly focused on the inhibition of inflammation. Therefore, the search for a multi-target therapeutic choice remains unchallenged. It seems that a drug with anti-inflammatory, oxidative stress inhibitory, reduction of MMPs activity, and neurogenesis stimulatory properties may be effective for treatment of MS. In this regard, Bryostatin-1 as a macrolide and marine natural product has been selected as a therapeutic choice. Studies indicate that Bryostatin-1 has anti-inflammatory and antioxidant properties and decreases MMPs level and BBB damage. Furthermore, Bryostatin-1 has a neuroprotective effect and promotes neurogenesis and differentiation of oligodendrocyte progenitor stem cells as a critical step for remyelination/myelogenesis. Based on these properties, we hypothesized here that Bryostatin-1 is an effective treatment in MS.
Collapse
Affiliation(s)
- Fahimeh Safaeinejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Centre, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Marsden MD, Wu X, Navab SM, Loy BA, Schrier AJ, DeChristopher BA, Shimizu AJ, Hardman CT, Ho S, Ramirez CM, Wender PA, Zack JA. Characterization of designed, synthetically accessible bryostatin analog HIV latency reversing agents. Virology 2018; 520:83-93. [PMID: 29800728 PMCID: PMC6018613 DOI: 10.1016/j.virol.2018.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 11/15/2022]
Abstract
HIV latency in resting CD4+ T cell represents a key barrier preventing cure of the infection with antiretroviral drugs alone. Latency reversing agents (LRAs) can activate HIV expression in latently infected cells, potentially leading to their elimination through virus-mediated cytopathic effects, host immune responses, and/or therapeutic strategies targeting cells actively expressing virus. We have recently described several structurally simplified analogs of the PKC modulator LRA bryostatin (termed bryologs) designed to improve synthetic accessibility, tolerability in vivo, and efficacy in inducing HIV latency reversal. Here we report the comparative performance of lead bryologs, including their effects in reducing cell surface expression of HIV entry receptors, inducing proinflammatory cytokines, inhibiting short-term HIV replication, and synergizing with histone deacetylase inhibitors to reverse HIV latency. These data provide unique insights into structure-function relationships between A- and B-ring bryolog modifications and activities in primary cells, and suggest that bryologs represent promising leads for preclinical advancement.
Collapse
Affiliation(s)
- Matthew D Marsden
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA 90095, United States.
| | - Xiaomeng Wu
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Sara M Navab
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Brian A Loy
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Adam J Schrier
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Brian A DeChristopher
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Akira J Shimizu
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Clayton T Hardman
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Stephen Ho
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States
| | - Christina M Ramirez
- Department of Biostatistics, School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Paul A Wender
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, United States.
| | - Jerome A Zack
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA 90095, United States; Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
37
|
Mai HN, Sharma N, Shin EJ, Nguyen BT, Nguyen PT, Jeong JH, Jang CG, Cho EH, Nah SY, Kim NH, Nabeshima T, Kim HC. Exposure to far-infrared rays attenuates methamphetamine-induced recognition memory impairment via modulation of the muscarinic M1 receptor, Nrf2, and PKC. Neurochem Int 2018; 116:63-76. [PMID: 29572053 DOI: 10.1016/j.neuint.2018.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 01/15/2023]
Abstract
We demonstrated that activation of protein kinase Cδ (PKCδ) and inactivation of the glutathione peroxidase-1 (GPx-1)-dependent systems are critical for methamphetamine (MA)-induced recognition memory impairment. We also demonstrated that exposure to far-infrared rays (FIR) causes induction of the glutathione (GSH)-dependent system, including induction of the GPx-1 gene. Here, we investigated whether exposure to FIR rays affects MA-induced recognition memory impairment and whether it modulates PKC, cholinergic receptors, and the GSH-dependent system. Because the PKC activator bryostatin-1 mainly induces PKCα, PKCε, and PKCδ, we assessed expression of these proteins after MA treatment. MA treatment selectively increased PKCδ expression and its phosphorylation. Exposure to FIR rays significantly attenuated MA-induced increases in PKCδ phosphorylation. Importantly, bryostatin-1 potentiated MA-induced phosphorylation of PKCδ. MA treatment significantly decreased M1, M3, and M4 muscarinic acetylcholine receptors (mAChRs) and β2 nicotinic acetylcholine receptor expression. Of these, the decrease was most pronounced in M1 mAChR. Exposure to FIR significantly attenuated MA-induced decreases in the M1 mAChR and phospho-ERK1/2, while it facilitated Nrf2-dependent GSH induction. Dicyclomine, an M1 mAChR antagonist, and l-buthionine-(S, R)-sulfoximine (BSO), an inhibitor of GSH synthesis, counteracted against the protective potentials mediated by FIR. More importantly, the memory-enhancing potential of FIR rays was significantly counteracted by bryostatin-1, dicyclomine, and BSO. Our results suggest that exposure to FIR rays attenuates MA-induced impairment in recognition memory via up-regulation of M1 mAChR, Nrf2-dependent GSH induction, and ERK1/2 phosphorylation by inhibiting PKCδ phosphorylation by bryostatin-1.
Collapse
Affiliation(s)
- Huynh Nhu Mai
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Phuong Tram Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Eun-Hee Cho
- Department of Internal Medicine, Medical School, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, KonKuk University, Seoul 05029, Republic of Korea
| | - Nam Hun Kim
- College of Forest and Environmental Sciences, Kangwon National University, Chunchon 24341, Republic of Korea.
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, Aichi 470-1192, Japan; Aino University, Ibaragi, 567-0012, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| |
Collapse
|
38
|
Abstract
Multiple sclerosis (MS) is an inflammatory disorder targeting the central nervous system (CNS). The relapsing-remitting phase of MS is largely driven by peripheral activation of autoreactive T-helper (Th) 1 and Th17 lymphocytes. In contrast, compartmentalized inflammation within the CNS, including diffuse activation of innate myeloid cells, characterizes the progressive phase of MS, the most debilitating phase that currently lacks satisfactory treatments. Recently, bryostatin-1 (bryo-1), a naturally occurring, CNS-permeable compound with a favorable safety profile in humans, has been shown to act on antigen-presenting cells to promote differentiation of lymphocytes into Th2 cells, an action that might benefit Th1-driven inflammatory conditions such as MS. In the present study, we show that bryo-1 provides marked benefit in mice with experimental autoimmune encephalomyelitis (EAE), an experimental MS animal model. Preventive treatment with bryo-1 abolishes the onset of neurologic deficits in EAE. More strikingly, bryo-1 reverses neurologic deficits after EAE onset, even when treatment is initiated at a late stage of disease when peak adaptive immunity has subsided. Treatment with bryo-1 in vitro promotes an anti-inflammatory phenotype in antigen-presenting dendritic cells, macrophages, and to a lesser extent, lymphocytes. These findings suggest the potential for bryo-1 as a therapeutic agent in MS, particularly given its established clinical safety. Furthermore, the benefit of bryo-1, even in late treatment of EAE, combined with its targeting of innate myeloid cells suggests therapeutic potential in progressive forms of MS.
Collapse
|
39
|
Tomàs JM, Garcia N, Lanuza MA, Nadal L, Tomàs M, Hurtado E, Simó A, Cilleros V. Membrane Receptor-Induced Changes of the Protein Kinases A and C Activity May Play a Leading Role in Promoting Developmental Synapse Elimination at the Neuromuscular Junction. Front Mol Neurosci 2017; 10:255. [PMID: 28848391 PMCID: PMC5552667 DOI: 10.3389/fnmol.2017.00255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023] Open
Abstract
Synapses that are overproduced during histogenesis in the nervous system are eventually lost and connectivity is refined. Membrane receptor signaling leads to activity-dependent mutual influence and competition between axons directly or with the involvement of the postsynaptic cell and the associated glial cell/s. Presynaptic muscarinic acetylcholine (ACh) receptors (subtypes mAChR; M1, M2 and M4), adenosine receptors (AR; A1 and A2A) and the tropomyosin-related kinase B receptor (TrkB), among others, all cooperate in synapse elimination. Between these receptors there are several synergistic, antagonic and modulatory relations that clearly affect synapse elimination. Metabotropic receptors converge in a limited repertoire of intracellular effector kinases, particularly serine protein kinases A and C (PKA and PKC), to phosphorylate protein targets and bring about structural and functional changes leading to axon loss. In most cells A1, M1 and TrkB operate mainly by stimulating PKC whereas A2A, M2 and M4 inhibit PKA. We hypothesize that a membrane receptor-induced shifting in the protein kinases A and C activity (inhibition of PKA and/or stimulation of PKC) in some nerve endings may play an important role in promoting developmental synapse elimination at the neuromuscular junction (NMJ). This hypothesis is supported by: (i) the tonic effect (shown by using selective inhibitors) of several membrane receptors that accelerates axon loss between postnatal days P5-P9; (ii) the synergistic, antagonic and modulatory effects (shown by paired inhibition) of the receptors on axonal loss; (iii) the fact that the coupling of these receptors activates/inhibits the intracellular serine kinases; and (iv) the increase of the PKA activity, the reduction of the PKC activity or, in most cases, both situations simultaneously that presumably occurs in all the situations of singly and paired inhibition of the mAChR, AR and TrkB receptors. The use of transgenic animals and various combinations of selective and specific PKA and PKC inhibitors could help to elucidate the role of these kinases in synapse maturation.
Collapse
Affiliation(s)
- Josep M Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Anna Simó
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| | - Víctor Cilleros
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i VirgiliReus, Spain
| |
Collapse
|
40
|
Bharath LP, Cho JM, Park SK, Ruan T, Li Y, Mueller R, Bean T, Reese V, Richardson RS, Cai J, Sargsyan A, Pires K, Anandh Babu PV, Boudina S, Graham TE, Symons JD. Endothelial Cell Autophagy Maintains Shear Stress-Induced Nitric Oxide Generation via Glycolysis-Dependent Purinergic Signaling to Endothelial Nitric Oxide Synthase. Arterioscler Thromb Vasc Biol 2017; 37:1646-1656. [PMID: 28684613 DOI: 10.1161/atvbaha.117.309510] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/19/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Impaired endothelial cell (EC) autophagy compromises shear stress-induced nitric oxide (NO) generation. We determined the responsible mechanism. APPROACH AND RESULTS On autophagy compromise in bovine aortic ECs exposed to shear stress, a decrease in glucose uptake and EC glycolysis attenuated ATP production. We hypothesized that decreased glycolysis-dependent purinergic signaling via P2Y1 (P2Y purinoceptor 1) receptors, secondary to impaired autophagy in ECs, prevents shear-induced phosphorylation of eNOS (endothelial nitric oxide synthase) at its positive regulatory site S1117 (p-eNOSS1177) and NO generation. Maneuvers that restore glucose transport and glycolysis (eg, overexpression of GLUT1 [glucose transporter 1]) or purinergic signaling (eg, addition of exogenous ADP) rescue shear-induced p-eNOSS1177 and NO production in ECs with impaired autophagy. Conversely, inhibiting glucose transport via GLUT1 small interfering RNA, blocking purinergic signaling via ectonucleotidase-mediated ATP/ADP degradation (eg, apyrase), or inhibiting P2Y1 receptors using pharmacological (eg, MRS2179 [2'-deoxy-N6-methyladenosine 3',5'-bisphosphate tetrasodium salt]) or genetic (eg, P2Y1-receptor small interfering RNA) procedures inhibit shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Supporting a central role for PKCδT505 (protein kinase C delta T505) in relaying the autophagy-dependent purinergic-mediated signal to eNOS, we find that (1) shear stress-induced activating phosphorylation of PKCδT505 is negated by inhibiting autophagy, (2) shear-induced p-eNOSS1177 and NO generation are restored in autophagy-impaired ECs via pharmacological (eg, bryostatin) or genetic (eg, constitutively active PKCδ) activation of PKCδT505, and (3) pharmacological (eg, rottlerin) and genetic (eg, PKCδ small interfering RNA) PKCδ inhibition prevents shear-induced p-eNOSS1177 and NO generation in ECs with intact autophagy. Key nodes of dysregulation in this pathway on autophagy compromise were revealed in human arterial ECs. CONCLUSIONS Targeted reactivation of purinergic signaling and PKCδ has strategic potential to restore compromised NO generation in pathologies associated with suppressed EC autophagy.
Collapse
Affiliation(s)
- Leena P Bharath
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jae Min Cho
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Seul-Ki Park
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ting Ruan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Youyou Li
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Robert Mueller
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Tyler Bean
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Van Reese
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Russel S Richardson
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Jinjin Cai
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Ashot Sargsyan
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Karla Pires
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Pon Velayutham Anandh Babu
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Sihem Boudina
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - Timothy E Graham
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.)
| | - J David Symons
- From the Department of Nutrition and Integrative Physiology, College of Health (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., R.S.R., K.P., V.A.B., S.B., T.E.G., J.D.S.) and Molecular Medicine Program (J.C., A.S., S.B., T.E.G., J.D.S.), University of Utah, Salt Lake City; Division of Endocrinology, Metabolism, and Diabetes, University of Utah School of Medicine, Salt Lake City (L.P.B., J.M.C., S.-K.P., T.R., Y.L., R.M., T.B., J.C., A.S., K.P., S.B., T.E.G., J.D.S.); and University of Utah Geriatric Research, Education, and Clinical Center, George E. Whalen VA Medical Center, Salt Lake City (V.R., R.S.R.).
| |
Collapse
|
41
|
Park H, Ha J, Koo JY, Park J, Park SB. Label-free target identification using in-gel fluorescence difference via thermal stability shift. Chem Sci 2016; 8:1127-1133. [PMID: 28451252 PMCID: PMC5369521 DOI: 10.1039/c6sc03238a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/20/2016] [Indexed: 12/24/2022] Open
Abstract
A label-free method for proteome-wide target identification was developed using in-gel fluorescence difference caused by thermal stability shift.
Target engagement is a prerequisite for the therapeutic effects of bioactive small molecules, and unbiased identification of their target proteins can facilitate drug discovery and chemical biology research. Structural modifications of bioactive natural products for target identification exhibit potential limitations such as synthetic difficulties, limited supplies from natural sources, and loss of original efficacy. Herein, we developed a label-free method for proteome-wide target identification using in-gel fluorescence difference caused by thermal stability shift, namely TS-FITGE. Quantitative intra-gel image analysis of each protein spot revealed target proteins with shifted thermal stability upon drug engagement, and plotting of melting curves by inter-gel analysis confirmed the positive targets. We demonstrated the robustness and applicability of the TS-FITGE method by identifying target proteins, including membrane-anchored proteins, of complex bioactive compounds. Furthermore, we identified and functionally validated nucleophosmin as a novel target protein of hordenine, a natural product upregulator of in vitro translation.
Collapse
Affiliation(s)
- Hankum Park
- Department of Biophysics and Chemical Biology , Seoul National University , Seoul - 08826 , Korea .
| | - Jaeyoung Ha
- Department of Biophysics and Chemical Biology , Seoul National University , Seoul - 08826 , Korea .
| | - Ja Young Koo
- CRI Center for Chemical Proteomics , Department of Chemistry , Seoul National University , Seoul - 08826 , Korea
| | - Jongmin Park
- CRI Center for Chemical Proteomics , Department of Chemistry , Seoul National University , Seoul - 08826 , Korea
| | - Seung Bum Park
- Department of Biophysics and Chemical Biology , Seoul National University , Seoul - 08826 , Korea . .,CRI Center for Chemical Proteomics , Department of Chemistry , Seoul National University , Seoul - 08826 , Korea
| |
Collapse
|
42
|
Nijampatnam B, Dutta S, Velu SE. Recent advances in isolation, synthesis, and evaluation of bioactivities of bispyrroloquinone alkaloids of marine origin. Chin J Nat Med 2016; 13:561-77. [PMID: 26253489 DOI: 10.1016/s1875-5364(15)30052-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Indexed: 12/29/2022]
Abstract
The ocean continues to provide a plethora of unique scaffolds capable of remarkable biological applications. A large number of pyrroloiminoquinone alkaloids, including discorhabdins, epinardins, batzellines, makaluvamines, and veiutamine, have been isolated from various marine organisms. A class of pyrroloiminoquinone-related alkaloids, known as bispyrroloquinones, is the focus of this review article. This family of marine alkaloids, which contain an aryl substituted bispyrroloquinone ring system, includes three subclasses of alkaloids namely, wakayin, tsitsikammamines A-B, and zyzzyanones A-D. Both wakayin and the tsitsikammamines contain a tetracyclic fused bispyrroloiminoquinone ring system, while zyzzyanones contain a fused tricyclic bispyrroloquinone ring system. The unique chemical structures of these marine natural products and their diverse biological properties, including antifungal and antimicrobial activity, as well as the potent, albeit generally nonspecific and universal cytotoxicities, have attracted great interest of synthetic chemists over the past three decades. Tsitsikammamines, wakayin, and several of their analogs show inhibition of topoisomerases. One additional possible mechanism of anticancer activity of tsitsikammamines analogs that has been discovered recently is through the inhibition of indoleamine 2, 3-dioxygenase, an enzyme involved in tumoral immune resistance. This review discusses the isolation, synthesis, and evaluation of bioactivities of bispyrroloquinone alkaloids and their analogs.
Collapse
Affiliation(s)
| | - Shilpa Dutta
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sadanandan E Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
43
|
Plummer S, Manning T, Baker T, McGreggor T, Patel M, Wylie G, Phillips D. Isolation, analytical measurements, and cell line studies of the iron-bryostatin-1 complex. Bioorg Med Chem Lett 2016; 26:2489-2497. [PMID: 27068183 DOI: 10.1016/j.bmcl.2016.03.099] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/27/2016] [Accepted: 03/28/2016] [Indexed: 10/22/2022]
Abstract
Bryostatin-1 is a marine natural product that has demonstrated medicinal activity in pre-clinical and clinical trials for the treatment of cancer, Alzheimer's disease, effects of stroke, and HIV. In this study, iron-bryostatin-1 was obtained using a pharmaceutical aquaculture technique developed by our lab that cultivates marine bacteria for marine natural product extraction. Analytical measurements (1)H and (13)C NMR, mass spectrometry, and flame atomic absorption were utilized to confirm the presence of an iron-bryostatin-1 complex. The iron-bryostatin-1 complex produced was then tested against the National Cancer Institute's 60 cell line panel. Adding iron to bryostatin-1 lowered the anti-cancer efficacy of the compound.
Collapse
Affiliation(s)
- Sydney Plummer
- Department of Chemistry, Valdosta State University, Valdosta, GA 31698, United States
| | - Thomas Manning
- Department of Chemistry, Valdosta State University, Valdosta, GA 31698, United States.
| | - Tess Baker
- Department of Chemistry, Valdosta State University, Valdosta, GA 31698, United States
| | - Tysheon McGreggor
- Department of Chemistry, Valdosta State University, Valdosta, GA 31698, United States
| | - Mehulkumar Patel
- Department of Chemistry, Valdosta State University, Valdosta, GA 31698, United States
| | - Greg Wylie
- NMR Facility, Department of Chemistry, Texas A&M, College Station, TX 77843, United States
| | - Dennis Phillips
- PAMS Facility, Department of Chemistry, University of Georgia, Athens, GA 30602, United States
| |
Collapse
|
44
|
Kelsey JS, Cataisson C, Chen J, Herrmann MA, Petersen ME, Baumann DO, McGowan KM, Yuspa SH, Keck GE, Blumberg PM. Biological activity of the bryostatin analog Merle 23 on mouse epidermal cells and mouse skin. Mol Carcinog 2016; 55:2183-2195. [PMID: 26859836 DOI: 10.1002/mc.22460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/06/2016] [Accepted: 01/06/2016] [Indexed: 12/19/2022]
Abstract
Bryostatin 1, a complex macrocyclic lactone, is the subject of multiple clinical trials for cancer chemotherapy. Although bryostatin 1 biochemically functions like the classic mouse skin tumor promoter phorbol 12-myristate 13-acetate (PMA) to bind to and activate protein kinase C, paradoxically, it fails to induce many of the typical phorbol ester responses, including tumor promotion. Intense synthetic efforts are currently underway to develop simplified bryostatin analogs that preserve the critical functional features of bryostatin 1, including its lack of tumor promoting activity. The degree to which bryostatin analogs maintain the unique pattern of biological behavior of bryostatin 1 depends on the specific cellular system and the specific response. Merle 23 is a significantly simplified bryostatin analog that retains bryostatin like activity only to a limited extent. Here, we show that in mouse epidermal cells the activity of Merle 23 was either similar to bryostatin 1 or intermediate between bryostatin 1 and PMA, depending on the specific parameter examined. We then examined the hyperplastic and tumor promoting activity of Merle 23 on mouse skin. Merle 23 showed substantially reduced hyperplasia and was not tumor promoting at a dose comparable to that for PMA. These results suggest that there may be substantial flexibility in the design of bryostatin analogs that retain its lack of tumor promoting activity. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jessica S Kelsey
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Christophe Cataisson
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jinqiu Chen
- Collaborative Protein Technology Resource, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michelle A Herrmann
- Collaborative Protein Technology Resource, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mark E Petersen
- Department of Chemistry, University of Utah, Salt Lake City, Utah
| | - David O Baumann
- Department of Chemistry, University of Utah, Salt Lake City, Utah
| | - Kevin M McGowan
- Department of Chemistry, University of Utah, Salt Lake City, Utah
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Gary E Keck
- Department of Chemistry, University of Utah, Salt Lake City, Utah
| | - Peter M Blumberg
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
45
|
Cyanobacterial Metabolite Calothrixins: Recent Advances in Synthesis and Biological Evaluation. Mar Drugs 2016; 14:17. [PMID: 26771620 PMCID: PMC4728514 DOI: 10.3390/md14010017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/22/2015] [Accepted: 01/04/2016] [Indexed: 12/30/2022] Open
Abstract
The marine environment is host to unparalleled biological and chemical diversity, making it an attractive resource for the discovery of new therapeutics for a plethora of diseases. Compounds that are extracted from cyanobacteria are of special interest due to their unique structural scaffolds and capacity to produce potent pharmaceutical and biotechnological traits. Calothrixins A and B are two cyanobacterial metabolites with a structural assembly of quinoline, quinone, and indole pharmacophores. This review surveys recent advances in the synthesis and evaluation of the biological activities of calothrixins. Due to the low isolation yields from the marine source and the promise this scaffold holds for anticancer and antimicrobial drugs, organic and medicinal chemists around the world have embarked on developing efficient synthetic routes to produce calothrixins. Since the first review appeared in 2009, 11 novel syntheses of calothrixins have been published in the efforts to develop methods that contain fewer steps and higher-yielding reactions. Calothrixins have shown their potential as topoisomerase I poisons for their cytotoxicity in cancer. They have also been observed to target various aspects of RNA synthesis in bacteria. Further investigation into the exact mechanism for their bioactivity is still required for many of its analogs.
Collapse
|
46
|
Lucke-Wold BP, Logsdon AF, Smith KE, Turner RC, Alkon DL, Tan Z, Naser ZJ, Knotts CM, Huber JD, Rosen CL. Bryostatin-1 Restores Blood Brain Barrier Integrity following Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2015; 52:1119-1134. [PMID: 25301233 PMCID: PMC5000781 DOI: 10.1007/s12035-014-8902-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023]
Abstract
Recent wars in Iraq and Afghanistan have accounted for an estimated 270,000 blast exposures among military personnel. Blast traumatic brain injury (TBI) is the 'signature injury' of modern warfare. Blood brain barrier (BBB) disruption following blast TBI can lead to long-term and diffuse neuroinflammation. In this study, we investigate for the first time the role of bryostatin-1, a specific protein kinase C (PKC) modulator, in ameliorating BBB breakdown. Thirty seven Sprague-Dawley rats were used for this study. We utilized a clinically relevant and validated blast model to expose animals to moderate blast exposure. Groups included: control, single blast exposure, and single blast exposure + bryostatin-1. Bryostatin-1 was administered i.p. 2.5 mg/kg after blast exposure. Evan's blue, immunohistochemistry, and western blot analysis were performed to assess injury. Evan's blue binds to albumin and is a marker for BBB disruption. The single blast exposure caused an increase in permeability compared to control (t = 4.808, p < 0.05), and a reduction back toward control levels when bryostatin-1 was administered (t = 5.113, p < 0.01). Three important PKC isozymes, PKCα, PKCδ, and PKCε, were co-localized primarily with endothelial cells but not astrocytes. Bryostatin-1 administration reduced toxic PKCα levels back toward control levels (t = 4.559, p < 0.01) and increased the neuroprotective isozyme PKCε (t = 6.102, p < 0.01). Bryostatin-1 caused a significant increase in the tight junction proteins VE-cadherin, ZO-1, and occludin through modulation of PKC activity. Bryostatin-1 ultimately decreased BBB breakdown potentially due to modulation of PKC isozymes. Future work will examine the role of bryostatin-1 in preventing chronic neurodegeneration following repetitive neurotrauma.
Collapse
Affiliation(s)
- Brandon P Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Aric F Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Kelly E Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Ryan C Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Daniel L Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, 26506, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Office of Professional Studies in Health Sciences, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Chelsea M Knotts
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
| | - Jason D Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Charles L Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26506, USA.
- Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV, 26506-9183, USA.
| |
Collapse
|
47
|
Larsen EM, Wilson MR, Taylor RE. Conformation-activity relationships of polyketide natural products. Nat Prod Rep 2015; 32:1183-206. [PMID: 25974024 PMCID: PMC4443481 DOI: 10.1039/c5np00014a] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Polyketides represent an important class of secondary metabolites that interact with biological targets connected to a variety of disease-associated pathways. Remarkably, nature's assembly lines, polyketide synthases, manufacture these privileged structures through a combinatorial mixture of just a few structural units. This review highlights the role of these structural elements in shaping a polyketide's conformational preferences, the use of computer-based molecular modeling and solution NMR studies in the identification of low-energy conformers, and the importance of conformational analogues in probing the bound conformation. In particular, this review covers several examples wherein conformational analysis complements classic structure-activity relationships in the design of biologically active natural product analogues.
Collapse
Affiliation(s)
- Erik M Larsen
- University of Notre Dame, Department of Chemistry & Biochemistry, 250 Nieuwland Science Hall, Notre Dame, Indiana, USA.
| | | | | |
Collapse
|
48
|
Lencz T, Malhotra AK. Targeting the schizophrenia genome: a fast track strategy from GWAS to clinic. Mol Psychiatry 2015; 20:820-6. [PMID: 25869805 PMCID: PMC4486648 DOI: 10.1038/mp.2015.28] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/10/2015] [Accepted: 01/23/2015] [Indexed: 12/23/2022]
Abstract
The Psychiatric Genomics Consortium-Schizophrenia Workgroup (PGC-SCZ) has recently published a genomewide association study (GWAS) identifying >100 genetic loci, encompassing a total of 341 protein-coding genes, attaining genomewide significance for susceptibility to schizophrenia. Given the extremely long time (12-15 years) and expense (>$1 billion) associated with the development of novel drug targets, repurposing of drugs with known and validated targets may be the most expeditious path toward deriving clinical utility from these GWAS findings. In the present study, we examined all genes within loci implicated by the PGC-SCZ GWAS against databases of targets of both approved and registered pharmaceutical compounds. We identified 20 potential schizophrenia susceptibility genes that encode proteins that are the targets of approved drugs. Of these, we prioritized genes/targets that are of clear neuropsychiatric interest and that are also sole members of the linkage disequilibrium block surrounding a PGC-SCZ GWAS hit. In addition to DRD2, 5 genes meet these criteria: CACNA1C, CACNB2, CACNA1I, GRIN2A and HCN1. An additional 20 genes coding for proteins that are the targets of drugs in registered clinical trials, but without approved indications, were also identified. Although considerable work is still required to fully explicate the biological implications of the PGC-SCZ GWAS results, pathways related to these known, druggable targets may represent a promising starting point.
Collapse
Affiliation(s)
- T Lencz
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA,Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA,Departments of Psychiatry and Molecular Medicine, Hofstra University School of Medicine, Hempstead, NY, USA,Division of Psychiatry Research, Zucker Hillside Hospital, 75-59 263rd Street, Glen Oaks, NY 11004, USA. E-mail: or
| | - A K Malhotra
- Division of Psychiatry Research, Zucker Hillside Hospital, Glen Oaks, NY, USA,Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA,Departments of Psychiatry and Molecular Medicine, Hofstra University School of Medicine, Hempstead, NY, USA,Division of Psychiatry Research, Zucker Hillside Hospital, 75-59 263rd Street, Glen Oaks, NY 11004, USA. E-mail: or
| |
Collapse
|
49
|
Abstract
The protein kinases C (PKCs) are a family of serine/threonine kinases involved in regulating multiple essential cellular processes such as survival, proliferation, and differentiation. Of particular interest is the novel, calcium-independent PKCθ which plays a central role in immune responses. PKCθ shares structural similarities with other PKC family members, mainly consisting of an N-terminal regulatory domain and a C-terminal catalytic domain tethered by a hinge region. This isozyme, however, is unique in that it translocates to the immunological synapse between a T cell and an antigen-presenting cell (APC) upon T cell receptor-peptide MHC recognition. Thereafter, PKCθ interacts physically and functionally with downstream effectors to mediate T cell activation and differentiation, subsequently leading to inflammation. PKCθ-specific perturbations have been identified in several diseases, most notably autoimmune disorders, and hence the modulation of its activity presents an attractive therapeutic intervention. To that end, many inhibitors of PKCs and PKCθ have been developed and tested in preclinical and clinical studies. And although selectivity remains a challenge, results are promising for the future development of effective PKCθ inhibitors that would greatly advance the treatment of several T-cell mediated diseases.
Collapse
|
50
|
Neumann JT, Thompson JW, Raval AP, Cohan CH, Koronowski KB, Perez-Pinzon MA. Increased BDNF protein expression after ischemic or PKC epsilon preconditioning promotes electrophysiologic changes that lead to neuroprotection. J Cereb Blood Flow Metab 2015; 35:121-30. [PMID: 25370861 PMCID: PMC4294405 DOI: 10.1038/jcbfm.2014.185] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 01/06/2023]
Abstract
Ischemic preconditioning (IPC) via protein kinase C epsilon (PKCɛ) activation induces neuroprotection against lethal ischemia. Brain-derived neurotrophic factor (BDNF) is a pro-survival signaling molecule that modulates synaptic plasticity and neurogenesis. Interestingly, BDNF mRNA expression increases after IPC. In this study, we investigated whether IPC or pharmacological preconditioning (PKCɛ activation) promoted BDNF-induced neuroprotection, if neuroprotection by IPC or PKCɛ activation altered neuronal excitability, and whether these changes were BDNF-mediated. We used both in vitro (hippocampal organotypic cultures and cortical neuronal-glial cocultures) and in vivo (acute hippocampal slices 48 hours after preconditioning) models of IPC or PKCɛ activation. BDNF protein expression increased 24 to 48 hours after preconditioning, where inhibition of the BDNF Trk receptors abolished neuroprotection against oxygen and glucose deprivation (OGD) in vitro. In addition, there was a significant decrease in neuronal firing frequency and increase in threshold potential 48 hours after preconditioning in vivo, where this threshold modulation was dependent on BDNF activation of Trk receptors in excitatory cortical neurons. In addition, 48 hours after PKCɛ activation in vivo, the onset of anoxic depolarization during OGD was significantly delayed in hippocampal slices. Overall, these results suggest that after IPC or PKCɛ activation, there are BDNF-dependent electrophysiologic modifications that lead to neuroprotection.
Collapse
Affiliation(s)
- Jake T Neumann
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - John W Thompson
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Ami P Raval
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Charles H Cohan
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [4] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Kevin B Koronowski
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [4] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|