1
|
Lopez Quezada L, Mba Medie F, Luu RJ, Gaibler RB, Gabriel EP, Rubio LD, Mulhern TJ, Marr EE, Borenstein JT, Fisher CR, Gard AL. Predicting Clinical Outcomes of SARS-CoV-2 Drug Efficacy with a High-Throughput Human Airway Microphysiological System. Adv Biol (Weinh) 2024; 8:e2300511. [PMID: 39123296 DOI: 10.1002/adbi.202300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/23/2023] [Indexed: 08/12/2024]
Abstract
The average cost to bring a new drug from its initial discovery to a patient's bedside is estimated to surpass $2 billion and requires over a decade of research and development. There is a need for new drug screening technologies that can parse drug candidates with increased likelihood of clinical utility early in development in order to increase the cost-effectiveness of this pipeline. For example, during the COVID-19 pandemic, resources were rapidly mobilized to identify effective therapeutic treatments but many lead antiviral compounds failed to demonstrate efficacy when progressed to human trials. To address the lack of predictive preclinical drug screening tools, PREDICT96-ALI, a high-throughput (n = 96) microphysiological system (MPS) that recapitulates primary human tracheobronchial tissue,is adapted for the evaluation of differential antiviral efficacy of native SARS-CoV-2 variants of concern. Here, PREDICT96-ALI resolves both the differential viral kinetics between variants and the efficacy of antiviral compounds over a range of drug doses. PREDICT96-ALI is able to distinguish clinically efficacious antiviral therapies like remdesivir and nirmatrelvir from promising lead compounds that do not show clinical efficacy. Importantly, results from this proof-of-concept study track with known clinical outcomes, demonstrate the feasibility of this technology as a prognostic drug discovery tool.
Collapse
Affiliation(s)
| | | | - Rebeccah J Luu
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | - Logan D Rubio
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | | | | | - Ashley L Gard
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
2
|
Ghorbanalipoor S, Matsumoto K, Gross N, Heimberg L, Krause M, Veldkamp W, Magens M, Zanken J, Neuschutz KJ, De Luca DA, Kridin K, Vidarsson G, Chakievska L, Visser R, Kunzel S, Recke A, Gupta Y, Boch K, Vorobyev A, Kalies K, Manz RA, Bieber K, Ludwig RJ. High throughput screening identifies repurposable drugs for modulation of innate and acquired immune responses. J Autoimmun 2024; 148:103302. [PMID: 39163739 DOI: 10.1016/j.jaut.2024.103302] [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: 12/06/2023] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/22/2024]
Abstract
A balanced immune system is essential to maintain adequate host defense and effective self-tolerance. While an immune system that fails to generate appropriate response will permit infections to develop, uncontrolled activation may lead to autoinflammatory or autoimmune diseases. To identify drug candidates capable of modulating immune cell functions, we screened 1200 small molecules from the Prestwick Chemical Library for their property to inhibit innate or adaptive immune responses. Our studies focused specifically on drug interactions with T cells, B cells, and polymorphonuclear leukocytes (PMNs). Candidate drugs that were validated in vitro were examined in preclinical models to determine their immunomodulatory impact in chronic inflammatory diseases, here investigated in chronic inflammatory skin diseases. Using this approach, we identified several candidate drugs that were highly effective in preclinical models of chronic inflammatory disease. For example, we found that administration of pyrvinium pamoate, an FDA-approved over-the-counter anthelmintic drug, suppressed B cell activation in vitro and halted the progression of B cell-dependent experimental pemphigoid by reducing numbers of autoantigen-specific B cell responses. In addition, in studies performed in gene-deleted mouse strains provided additional insight into the mechanisms underlying these effects, for example, the receptor-dependent actions of tamoxifen that inhibit immune-complex-mediated activation of PMNs. Collectively, our methods and findings provide a vast resource that can be used to identify drugs that may be repurposed and used to promote or inhibit cellular immune responses.
Collapse
Affiliation(s)
| | - Kazuko Matsumoto
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Natalie Gross
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Linda Heimberg
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Malin Krause
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Wendelien Veldkamp
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Moritz Magens
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Johannes Zanken
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Kerstin J Neuschutz
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - David A De Luca
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Khalaf Kridin
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Lenche Chakievska
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Remco Visser
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Sven Kunzel
- Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Andreas Recke
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Yask Gupta
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Katharina Boch
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Artem Vorobyev
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Kathrin Kalies
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
| | - Rudolf A Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
3
|
Phan UTT, Nguyen HD, Nguyen TKO, Tran TH, Le TH, Tran TTP. Anti-inflammatory effect of Piper longum L. fruit methanolic extract on lipopolysaccharide-treated RAW 264.7 murine macrophages. Heliyon 2024; 10:e26174. [PMID: 38404825 PMCID: PMC10884859 DOI: 10.1016/j.heliyon.2024.e26174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Context The Piper species was studied several potential properties such as anti-tumor, anti-inflammatory and antioxidant activity. However, the specific anti-inflammatory activity of the extract from the fruits of P. longum L. has not been investigated. Objectives Our study want to examine the anti-inflammatory effects of P. longum L. fruit methanolic extracts (PLE) on lipopolysachharide (LPS)-stimulated RAW 264.7 murine macrophages to understand the mechanism of this effect. Method This study examined the chemical profiling of PLE by LC-HRMS analysis and measured the presence of nitric oxide (NO), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) in the supernatant using the Griess reagent assay and enzyme-linked immunosorbent assay (ELISA), respectively. The mRNA expression of IL-6, TNF-α, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) were evaluated by using real-time quantitative polymerase chain reaction (RT-qPCR). Furthermore, the protein expression of COX-2, iNOS and the phosphorylation of MAPK family, c-Jun N-terminal kinase (JNK), p38 in protein level were observed by western blotting. Result PLE have detected 66 compounds which belong to different classes such as alkaloids, flavonoids, terpenoids, phenolics, lactones, and organic acids inhibited nitric oxide products with the IC50 = 28.5 ± 0.91 μg/mL. Moreover, PLE at 10-100 μg/mL up-regulate HO-1 protein expression from 3 to 10 folds at 3 h. It also downregulated the mRNA and protein expression of iNOS, COX-2, decreased IL-6 and TNF-α secretion by modulating the mitogen-activated protein kinase (MAPK) signaling pathway, specifically by decreasing the phosphorylation of p38 and JNK. Conclusion These results shown chemical profiling of PLE and demonstrated that PLE exhibits anti-inflammatory effects by regulating the MAPK family and could be a potential candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Uyen Thi Tu Phan
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Hai Dang Nguyen
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Thi Kieu Oanh Nguyen
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi, 12116, Viet Nam
| | - Thanh Huong Le
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| | - Thi Thu Phuong Tran
- University of Science and Technology of Hanoi (USTH), Vietnam Academic Science and Technology (VAST), 18 Hoang Quoc Viet, Nghia Do, Cau Giay, Hanoi, Viet Nam
| |
Collapse
|
4
|
Chepngetich J, Muriithi B, Gachie B, Thiong'o K, Jepkorir M, Gathirwa J, Kimani F, Mwitari P, Kiboi D. Amodiaquine drug pressure selects nonsynonymous mutations in pantothenate kinase 1, diacylglycerol kinase, and phosphatidylinositol-4 kinase in Plasmodium berghei ANKA. OPEN RESEARCH AFRICA 2023; 5:28. [PMID: 38915420 PMCID: PMC11195610 DOI: 10.12688/openresafrica.13436.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 06/26/2024]
Abstract
Background Lumefantrine (LM), piperaquine (PQ), and amodiaquine (AQ), the long-acting components of the artemisinin-based combination therapies (ACTs), are a cornerstone of malaria treatment in Africa. Studies have shown that PQ, AQ, and LM resistance may arise independently of predicted modes of action. Protein kinases have emerged as mediators of drug action and efficacy in malaria parasites; however, the link between top druggable Plasmodium kinases with LM, PQ, and AQ resistance remains unclear. Using LM, PQ, or AQ-resistant Plasmodium berghei parasites, we have evaluated the association of choline kinase (CK), pantothenate kinase 1 (PANK1), diacylglycerol kinase (DAGK), and phosphatidylinositol-4 kinase (PI4Kβ), and calcium-dependent protein kinase 1 (CDPK1) with LM, PQ, and AQ resistance in Plasmodium berghei ANKA. Methods We used in silico bioinformatics tools to identify ligand-binding motifs, active sites, and sequence conservation across the different parasites. We then used PCR and sequencing analysis to probe for single nucleotide polymorphisms (SNPs) within the predicted functional motifs in the CK, PANK1, DAGK, PI4Kβ, and CDPK1. Using qPCR analysis, we measured the mRNA amount of PANK1, DAGK, and PI4Kβ at trophozoites and schizonts stages. Results We reveal sequence conservation and unique ligand-binding motifs in the CK, PANK1, DAGK, PI4Kβ, and CDPK1 across malaria species. DAGK, PANK1, and PI4Kβ possessed nonsynonymous mutations; surprisingly, the mutations only occurred in the AQr parasites. PANK1 acquired Asn394His, while DAGK contained K270R and K292R mutations. PI4Kβ had Asp366Asn, Ser1367Arg, Tyr1394Asn and Asp1423Asn. We show downregulation of PANK1, DAGK, and PI4Kβ in the trophozoites but upregulation at the schizonts stages in the AQr parasites. Conclusions The selective acquisition of the mutations and the differential gene expression in AQ-resistant parasites may signify proteins under AQ pressure. The role of the mutations in the resistant parasites and their impact on drug responses require investigations using reverse genetics techniques in malaria parasites.
Collapse
Affiliation(s)
- Jean Chepngetich
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology and Innovation, Nairobi, 62000, 00200, Kenya
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Brenda Muriithi
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, 62000, 00200, Kenya
| | - Beatrice Gachie
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology and Innovation, Nairobi, 62000, 00200, Kenya
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Kevin Thiong'o
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Mercy Jepkorir
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Jeremiah Gathirwa
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Francis Kimani
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Peter Mwitari
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, Nairobi, 54840, 00200, Kenya
| | - Daniel Kiboi
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, 62000, 00200, Kenya
| |
Collapse
|
5
|
Wu YC, Lu MT, Chu PC, Chang CS. Novel 4-aminoquinoline analogs targeting the HIF-1α signaling pathway. Future Med Chem 2023; 15:1569-1582. [PMID: 37728024 DOI: 10.4155/fmc-2023-0169] [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] [Indexed: 09/21/2023] Open
Abstract
Background: The aminoquinoline core exhibits versatile pharmacological properties, particularly in the area of anticancer activity. This study was designed to investigate the potential of the 4-aminoquinoline scaffold in the development of anticancer agents by targeting the HIF-1α signaling pathway. Methodology: The authors synthesized multiple derivatives of 4-aminoquinoline containing heterocyclic rings by a microwave reactor and assessed the cytotoxicity and inhibitory effects of these derivatives on the HIF-1α signaling pathway. Conclusion: Compound 3s was identified as the most promising HIF-1α inhibitor due to its exceptional antiproliferative effects, with IC50 values of 0.6 and 53.3 nM observed in MiaPaCa-2 and MDA-MB-231 cells, respectively. Furthermore, compound 3s was found to inhibit HIF-1α expression by decreasing the level of HIF-1α mRNA.
Collapse
Affiliation(s)
- Yu-Chieh Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, 40604, Taiwan
| | - Meng-Tien Lu
- Department of Cosmeceutics & Graduate Institute of Cosmeceutics, China Medical University, Taichung, 40604, Taiwan
- Drug Development Center, China Medical University, Taichung, 40604, Taiwan
| | - Po-Chen Chu
- Department of Cosmeceutics & Graduate Institute of Cosmeceutics, China Medical University, Taichung, 40604, Taiwan
- Drug Development Center, China Medical University, Taichung, 40604, Taiwan
| | - Chih-Shiang Chang
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, 40604, Taiwan
- Drug Development Center, China Medical University, Taichung, 40604, Taiwan
| |
Collapse
|
6
|
Antinarelli LMR, Midlej V, da Silva EDS, Coelho EAF, da Silva AD, Coimbra ES. Exploring the repositioning of the amodiaquine as potential drug against visceral leishmaniasis: The in vitro effect against Leishmania infantum is associated with multiple mechanisms, involving mitochondria dysfunction, oxidative stress and loss of cell cycle control. Chem Biol Interact 2023; 371:110333. [PMID: 36592711 DOI: 10.1016/j.cbi.2022.110333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/14/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Visceral leishmaniasis (VL) is a progressive, debilitating, and potentially fatal disease if left untreated. As a neglected tropical disease (NTD), the available treatment is restricted to a few drugs, which typically must be administered over a long period but are associated with serious adverse effects and have variability in efficacy. In this sense, drug repositioning has been considered an excellent strategy in the search for alternative treatments, especially in reducing the time and cost of the research. In this work, the repositioning potential of amodiaquine (AQ), a well-known antimalarial drug, was investigated for the treatment of VL. AQ showed significant and selective activity against promastigotes (IC50 = 11.6 μg/mL) and intracellular amastigotes (IC50 = 2.4 μg/mL) of L. infantum, being 10 times more destructive to the intracellular parasites than the host cell. In addition, pre-treatment of macrophages with AQ caused a significant reduction in the infection index, indicating a prophylactic effect of this drug. SEM images showed that AQ induces strong shape alterations of the promastigotes with an increase in cell volume with rounding and ribbing (vertical ridges), as well as a shortened flagellum. In addition, AQ induced depolarization of the ΔΨm, an increase in ROS and neutral lipids levels, and changes in the cell cycle in promastigotes, without alterations to the permeability of the parasite plasma membrane. L. infantum-infected macrophages treated with AQ induced the activation of oxidative mechanisms by infected host cells, with an increase in ROS and NO levels. Finally, in vitro interactions between AQ and miltefosine were found to have an additive effect in both biological stages of the parasite, with the ∑FIC50 values ranging from 0.74 to 1.16 μg/mL and 0.54-1.11 μg/mL for promastigotes and intracellular amastigotes, respectively. Overall, these data highlight the utility of drug repurposing and indicate future preclinical testing for AQ itself or in combination as a potential VL treatment.
Collapse
Affiliation(s)
- Luciana M Ribeiro Antinarelli
- Department of Parasitology, Microbiology, and Immunology, Biological Sciences Institute, Federal University of Juiz de Fora, UFJF, Juiz de Fora, Minas Gerais, 36.036-900, Brazil; Postgraduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, UFMG, Belo Horizonte, Minas Gerais, 30130-100, Brazil
| | - Victor Midlej
- Laboratory of Cellular and Ultrastructure, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, 21040-900, Brazil
| | | | - Eduardo Antônio Ferraz Coelho
- Postgraduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, UFMG, Belo Horizonte, Minas Gerais, 30130-100, Brazil; Department of Clinical Pathology, COLTEC, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Adilson David da Silva
- Department of Chemistry, Institute of Exact Sciences, Federal University of Juiz de Fora, UFJF, Juiz de Fora, Minas Gerais, 36.036-900, Brazil
| | - Elaine Soares Coimbra
- Department of Parasitology, Microbiology, and Immunology, Biological Sciences Institute, Federal University of Juiz de Fora, UFJF, Juiz de Fora, Minas Gerais, 36.036-900, Brazil.
| |
Collapse
|
7
|
Shoaib S, Ansari MA, Kandasamy G, Vasudevan R, Hani U, Chauhan W, Alhumaidi MS, Altammar KA, Azmi S, Ahmad W, Wahab S, Islam N. An Attention towards the Prophylactic and Therapeutic Options of Phytochemicals for SARS-CoV-2: A Molecular Insight. Molecules 2023; 28:795. [PMID: 36677853 PMCID: PMC9864057 DOI: 10.3390/molecules28020795] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023] Open
Abstract
The novel pathogenic virus was discovered in Wuhan, China (December 2019), and quickly spread throughout the world. Further analysis revealed that the pathogenic strain of virus was corona but it was distinct from other coronavirus strains, and thus it was renamed 2019-nCoV or SARS-CoV-2. This coronavirus shares many characteristics with other coronaviruses, including SARS-CoV and MERS-CoV. The clinical manifestations raised in the form of a cytokine storm trigger a complicated spectrum of pathophysiological changes that include cardiovascular, kidney, and liver problems. The lack of an effective treatment strategy has imposed a health and socio-economic burden. Even though the mortality rate of patients with this disease is lower, since it is judged to be the most contagious, it is considered more lethal. Globally, the researchers are continuously engaged to develop and identify possible preventive and therapeutic regimens for the management of disease. Notably, to combat SARS-CoV-2, various vaccine types have been developed and are currently being tested in clinical trials; these have also been used as a health emergency during a pandemic. Despite this, many old antiviral and other drugs (such as chloroquine/hydroxychloroquine, corticosteroids, and so on) are still used in various countries as emergency medicine. Plant-based products have been reported to be safe as alternative options for several infectious and non-infectious diseases, as many of them showed chemopreventive and chemotherapeutic effects in the case of tuberculosis, cancer, malaria, diabetes, cardiac problems, and others. Therefore, plant-derived products may play crucial roles in improving health for a variety of ailments by providing a variety of effective cures. Due to current therapeutic repurposing efforts against this newly discovered virus, we attempted to outline many plant-based compounds in this review to aid in the fight against SARS-CoV-2.
Collapse
Affiliation(s)
- Shoaib Shoaib
- Department Biochemistry, Faculty of Medicine, J. N. Medical College, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Geetha Kandasamy
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Rajalakshimi Vasudevan
- Department of Pharmacology, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Waseem Chauhan
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India
| | - Maryam S. Alhumaidi
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Khadijah A. Altammar
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Sarfuddin Azmi
- Molecular Microbiology Biology Division, Scientific Research Centre (SRC), Prince Sultan Military Medical City (PSMMC), Riyadh 11159, Saudi Arabia
| | - Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Shadma Wahab
- Deparment of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Najmul Islam
- Department Biochemistry, Faculty of Medicine, J. N. Medical College, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
8
|
Emmanuel Chimeh E, Nicodemus Emeka N, Florence Nkechi N, Amaechi Linda O, Oka Samon A, Emmanuel Chigozie A, Parker Elijah J, Barine Innocent N, Ezike Tobechukwu C, Nwachukwu Philip A, Hope Chimbuezie N, Chidimma Peace E, Onyinye Mary-Jane O, Godspower Chima N, Theresa Chinyere E, Alotaibi Saqer S, Albogami Sarah M, Gaber El-Saber B. Bioactive Compounds, anti-inflammatory, anti-nociceptive and antioxidant potentials of ethanolic leaf fraction of Sida linifolia L. (Malvaceae). ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
9
|
Lawal B, Kuo YC, Rachmawati Sumitra M, Wu ATH, Huang HS. Identification of a novel immune-inflammatory signature of COVID-19 infections, and evaluation of pharmacokinetics and therapeutic potential of RXn-02, a novel small-molecule derivative of quinolone. Comput Biol Med 2022; 148:105814. [PMID: 35841781 PMCID: PMC9272679 DOI: 10.1016/j.compbiomed.2022.105814] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/07/2022] [Accepted: 07/03/2022] [Indexed: 01/18/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic and respiratory infection that has enormous damage to human lives and economies. It is caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), a non-pair-stranded positive-sense RNA virus. With increasing global threats and few therapeutic options, the discovery of new potential drug targets and the development of new therapy candidates against COVID-19 are urgently needed. Based on these premises, we conducted an analysis of transcriptomic datasets from SARS-CoV-2-infected patients and identified several SARS-CoV-2 infection signatures, among which TNFRSF5/PTPRC/IDO1/MKI67 appeared to be the most pertinent signature. Subsequent integrated bioinformatics analysis identified the signature as an important immunomodulatory and inflammatory signature of SARS-CoV-2 infection. It was suggested that this gene signature mediates the interplay of immune and immunosuppressive cells leading to infiltration-exclusion of effector memory T cells in the lungs, which is of translation relevance for developing novel SARS-CoV-2 drug and vaccine candidates. Consequently, we designed and synthesized a novel small-molecule quinoline derivative (RXn-02) and evaluated its pharmacokinetics in rats, revealing a peak plasma concentration (Cmax) and time to Cmax (Tmax) of 1.756 μg/mL and 0.6 h, respectively. Values of the area under the curve (AUC) (0–24 h) and AUC (0 h∼∞) were 18.90 and 71.20 μg h/mL, respectively. Drug absorption from the various regional segments revealed that the duodenum (49.84%), jejunum (47.885%), cecum (1.82%), and ileum (0.32%) were prime sites of RXn-02 absorption. No absorption was detected from the stomach, and the least was from the colon (0.19%). Interestingly, RXn-02 exhibited in vitro antiproliferative activities against hub gene hyper-expressing cell lines; A549 (IC50 = 48.1 μM), K-562 (IC50 = 100 μM), and MCF7 (IC50 = 0.047 μM) and against five cell lines originating from human lungs (IC50 range of 33.2–69.5 μM). In addition, RXn-02 exhibited high binding efficacies for targeting the TNFRSF5/PTPRC/IDO1/MK signature with binding affinities (ΔG) of −6.6, −6.0, −9.9, −6.9 kcal/mol respectively. In conclusion, our study identified a novel signature of SARS-CoV-2 pathogenesis. RXn-02 is a drug-like candidate with good in vivo pharmacokinetics and hence possesses great translational relevance worthy of further preclinical and clinical investigations for treating SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Bashir Lawal
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan; School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Maryam Rachmawati Sumitra
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Alexander T H Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; School of Pharmacy, National Defense Medical Center, Taipei, 11490, Taiwan; PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
10
|
Repurposing Histaminergic Drugs in Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23116347. [PMID: 35683024 PMCID: PMC9181091 DOI: 10.3390/ijms23116347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease with a strong neuroinflammatory component that contributes to severe demyelination, neurodegeneration and lesions formation in white and grey matter of the spinal cord and brain. Increasing attention is being paid to the signaling of the biogenic amine histamine in the context of several pathological conditions. In multiple sclerosis, histamine regulates the differentiation of oligodendrocyte precursors, reduces demyelination, and improves the remyelination process. However, the concomitant activation of histamine H1–H4 receptors can sustain either damaging or favorable effects, depending on the specifically activated receptor subtype/s, the timing of receptor engagement, and the central versus peripheral target district. Conventional drug development has failed so far to identify curative drugs for multiple sclerosis, thus causing a severe delay in therapeutic options available to patients. In this perspective, drug repurposing offers an exciting and complementary alternative for rapidly approving some medicines already approved for other indications. In the present work, we have adopted a new network-medicine-based algorithm for drug repurposing called SAveRUNNER, for quantifying the interplay between multiple sclerosis-associated genes and drug targets in the human interactome. We have identified new histamine drug-disease associations and predicted off-label novel use of the histaminergic drugs amodiaquine, rupatadine, and diphenhydramine among others, for multiple sclerosis. Our work suggests that selected histamine-related molecules might get to the root causes of multiple sclerosis and emerge as new potential therapeutic strategies for the disease.
Collapse
|
11
|
Ashmawy AI, El-Abhar HS, Abdallah DM, Ali MA. Chloroquine modulates the sulforaphane anti-obesity mechanisms in a high-fat diet model: Role of JAK-2/ STAT-3/ SOCS-3 pathway. Eur J Pharmacol 2022; 927:175066. [PMID: 35643302 DOI: 10.1016/j.ejphar.2022.175066] [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: 02/06/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/13/2022]
Abstract
The phytochemical sulforaphane (SFN) has been studied for its potential anti-obesity effect, but neither its molecular targets nor its interaction with the antimalarial drug chloroquine (CQ) has been fully delineated. Therefore, high-fat diet (HFD) obese rats were randomly allocated into one of five groups and were left untreated or gavaged orally with SFN (0.5 or 1 mg/kg), CQ (5 mg/kg), or their combination (0.5/5 mg/kg) for six successive weeks to assess their potential interaction and the enrolled mechanisms. SFN effectively reduced the HFD-induced weight gain, blood glucose, and serum leptin levels, and improved lipid profile. On the molecular level, SFN inhibited the lipogenesis-related enzymes, namely sterol regulatory element-binding protein (SREBP)-1c, fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC) in both liver and visceral white adipose tissue (vWAT) of HFD obese rats. SFN also turned off the inflammatory pathway conserved Janus kinase/signaling transducers and activators of transcription/suppressor of cytokine signaling (JAK-2/STAT-3/SOCS-3) in these tissues, as well as the inflammatory markers nuclear factor-kappa (NF-κ) B and interleukin (IL)-22 in serum. In contrast, SFN downregulated the gene expression of microRNA (miR-200a), while significantly increasing the autophagic parameters; viz., beclin-1, autophagy-related protein (ATG)-7, and microtubule-associated protein 2 light chain 3 (LC3-II) in both liver and vWAT. On most of the parameters mentioned above, treatment with CQ solely produced a satisfactory effect and intensified the low dose of SFN in the combination regimen. These findings demonstrated the beneficial effects of using CQ as an add-on anti-obesity medicine to SFN.
Collapse
Affiliation(s)
- Ahmed I Ashmawy
- Department of Pharmacology & Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology & Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mennatallah A Ali
- Department of Pharmacology & Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
12
|
Shiraki H, Tanaka S, Guo Y, Harada K, Hide I, Yasuda T, Sakai N. Potential role of inducible GPR3 expression under stimulated T cell conditions. J Pharmacol Sci 2022; 148:307-314. [PMID: 35177210 DOI: 10.1016/j.jphs.2022.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/01/2022] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptor 3 (GPR3) constitutively activates Gαs proteins without any ligands and is predominantly expressed in neurons. Since the expression and physiological role of GPR3 in immune cells is still unknown, we examined the possible role of GPR3 in T lymphocytes. The expression of GPR3 was upregulated 2 h after phorbol 12-myristate 13-acetate (PMA)/ionomycin stimulation and was sustained in Jurkat cells, a human T lymphocyte cell line. In addition, the expression of nuclear receptor 4 group A member 2 (NR4A2) was highly modulated by GPR3 expression. Additionally, GPR3 expression was linked with the transcriptional promoter activity of NR4A in Jurkat cells. In mouse CD4+ T cells, transient GPR3 expression was induced immediately after the antigen receptor stimulation. However, the expression of NR4A2 was not modulated in CD4+ T cells from GPR3-knockout mice after stimulation, and the population of Treg cells in thymocytes and splenocytes was not affected by GPR3 knockout. By contrast, spontaneous effector activation in both CD4+ T cells and CD8+ T cells was observed in GPR3-knockout mice. In summary, GPR3 is immediately induced by T cell stimulation and play an important role in the suppression of effector T cell activation.
Collapse
Affiliation(s)
- Hiroko Shiraki
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | - Yun Guo
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tomoharu Yasuda
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| |
Collapse
|
13
|
Silva Neto GJ, Silva LR, Omena RJMD, Aguiar ACC, Annunciato Y, Rosseto B, Gazarini ML, Heimfarth L, Quintans-Júnior LJ, Ferreira E, Meneghetti MR. Dual Quinoline-Hybrid Compounds with Antimalarial Activity Against Plasmodium falciparum Parasites. NEW J CHEM 2022. [DOI: 10.1039/d1nj05598d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Although we have at our disposal relatively low-cost drugs that can be prescribed for the treatment of malaria, the prevalence of resistant strains of the causative parasite has required the...
Collapse
|
14
|
Ding P, Liang C, Ouyang W, Li G, Xiao Q, Luo J. Inferring Synergistic Drug Combinations Based on Symmetric Meta-Path in a Novel Heterogeneous Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1562-1571. [PMID: 31714232 DOI: 10.1109/tcbb.2019.2951557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Combinatorial drug therapy is a promising way for treating cancers, which can reduce drug side effects and improve drug efficacy. However, due to the large-scale combinatorial space, it is difficult to quickly and effectively identify novel synergistic drug combinations for further implementing combinatorial drug therapy. The computational method of fusing multi-source knowledge is a time- and cost-efficient strategy to infer synergistic drug combinations for testing. However, for the existing computational methods of inferring synergistic drug combinations, it still remains a challenging to effectively combine multi-source information to achieve the desired results. Hence, in this study, we developed a novel Inference method of Synergistic Drug Combinations based on Symmetric Meta-Path (ISDCSMP), which can systematically and accurately prioritize synergistic drug combinations in a novel drug-target heterogeneous network integrating multi-source information. In the experiment, ISDCSMP outperformed the state-of-the-art methods in terms of AUC and precision on the benchmark dataset in five-fold cross validation. Moreover, we further illustrated performances of different ways for obtaining the combination coefficients, and analyzed the influences of the maximum meta-path length. The performances of various single meta-paths were described in five-fold cross validation. Finally, we confirmed the practical usefulness of ISDCSMP with the predicted novel synergistic drug combinations. The source code of ISDCSMP is available at https://github.com/KDDing/ISDCSMP.
Collapse
|
15
|
Harrington WE, Moore KA, Min AM, Gilder ME, Tun NW, Paw MK, Wiladphaingern J, Proux S, Chotivanich K, Rijken MJ, White NJ, Nosten F, McGready R. Falciparum but not vivax malaria increases the risk of hypertensive disorders of pregnancy in women followed prospectively from the first trimester. BMC Med 2021; 19:98. [PMID: 33902567 PMCID: PMC8077872 DOI: 10.1186/s12916-021-01960-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/16/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Malaria and hypertensive disorders of pregnancy (HDoP) affect millions of pregnancies worldwide, particularly those of young, first-time mothers. Small case-control studies suggest a positive association between falciparum malaria and risk of pre-eclampsia but large prospective analyses are lacking. METHODS We characterized the relationship between malaria in pregnancy and the development of HDoP in a large, prospectively followed cohort. Pregnant women living along the Thailand-Myanmar border, an area of low seasonal malaria transmission, were followed at antenatal clinics between 1986 and 2016. The relationships between falciparum and vivax malaria during pregnancy and the odds of gestational hypertension, pre-eclampsia, or eclampsia were examined using logistic regression amongst all women and then stratified by gravidity. RESULTS There were 23,262 singleton pregnancies in women who presented during the first trimester and were followed fortnightly. Falciparum malaria was associated with gestational hypertension amongst multigravidae (adjusted odds ratio (AOR) 2.59, 95%CI 1.59-4.23), whereas amongst primigravidae, it was associated with the combined outcome of pre-eclampsia/eclampsia (AOR 2.61, 95%CI 1.01-6.79). In contrast, there was no association between vivax malaria and HDoP. CONCLUSIONS Falciparum but not vivax malaria during pregnancy is associated with hypertensive disorders of pregnancy.
Collapse
Affiliation(s)
- Whitney E Harrington
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Kerryn A Moore
- London School of Hygiene and Tropical Medicine, London, UK
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Aung Myat Min
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Mary Ellen Gilder
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Nay Win Tun
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Moo Kho Paw
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | | | - Marcus J Rijken
- Utrecht University Medical Centre, Utrecht, the Netherlands
- Julius Centre Global Health, Utrecht, the Netherlands
| | - Nicholas J White
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, UK.
| |
Collapse
|
16
|
Rongyi C, Zongfei J, Jiang L, Sifan W, Lingying M, Ying S, Wenshu Y, Xiaomeng C, Xiufang K, Xiaomin D, Lili M, Yanshan L, Lindi J. Effect of hydroxychloroquine on angiographic progression in routine treatment of Takayasu arteritis. Mod Rheumatol 2021; 31:1135-1141. [PMID: 33497271 DOI: 10.1080/14397595.2021.1879347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVES Hydroxychloroquine (HCQ), an anti-malarial drug, is widely used in the treatment of rheumatic diseases. However, the benefits of HCQ in the treatment of Takayasu arteritis (TA) remain unclear, especially in terms of alleviation of vascular progression. METHODS This longitudinal observational retrospective study was based on the East China TA cohort. Patients received routine treatment with prednisone and immunosuppressants. Fifty TA patients who underwent magnetic resonance angiography two times within a 1.5-year follow-up period of monitoring vascular changes were divided into HCQ and non-HCQ groups according to whether HCQ was prescribed. Changes in angiographic features were compared. Multivariate Cox regression analysis was employed to further validate the results. RESULTS Of 50 TA patients, 21 were prescribed HCQ. The two groups shared a similar disease course, vascular types, prednisone with immunosuppressants intervention strategy, globin level, and disease remission rate at 6 months. The HCQ group showed greater reduction in the inflammatory indices erythrocyte sedimentation rate and C-reactive protein (CRP) level (p < .05), and a significantly lower incidence of angiographic progression than the non-HCQ group (19.0% vs. 51.7%, p = .035). After adjustment for age and usage of tocilizumab, angiographic progression was found to be independently associated with CRP (hazard ratio [95% confidence interval], HR [95% CI]: 1.102 [1.000-1.024], p = .046), and the usage of HCQ (HR [95% CI]: 0.266 [0.075-0.940], p = .040). CONCLUSION HCQ enhanced the anti-inflammatory effect of routine treatment strategies with prednisone and immunosuppressants, and alleviated angiographic progression in TA.
Collapse
Affiliation(s)
- Chen Rongyi
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Ji Zongfei
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Lin Jiang
- Department of Radiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Wu Sifan
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Ma Lingying
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Sun Ying
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Yu Wenshu
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Cui Xiaomeng
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Kong Xiufang
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Dai Xiaomin
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Ma Lili
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| | - Li Yanshan
- Department of Rheumatology and Immunology, Linyi People's Hospital, Shandong, China
| | - Jiang Lindi
- Department of Rheumatology, Zhongshan Hospital Fudan University, Shanghai, China.,Evidence-based Medicine Center, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Kamat S, Kumari M. Repurposing Chloroquine Against Multiple Diseases With Special Attention to SARS-CoV-2 and Associated Toxicity. Front Pharmacol 2021; 12:576093. [PMID: 33912030 PMCID: PMC8072386 DOI: 10.3389/fphar.2021.576093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
Chloroquine and its derivatives have been used since ages to treat malaria and have also been approved by the FDA to treat autoimmune diseases. The drug employs pH-dependent inhibition of functioning and signalling of the endosome, lysosome and trans-Golgi network, immunomodulatory actions, inhibition of autophagy and interference with receptor binding to treat cancer and many viral diseases. The ongoing pandemic of COVID-19 has brought the whole world on the knees, seeking an urgent hunt for an anti-SARS-CoV-2 drug. Chloroquine has shown to inhibit receptor binding of the viral particles, interferes with their replication and inhibits "cytokine storm". Though multiple modes of actions have been employed by chloroquine against multiple diseases, viral diseases can provide an added advantage to establish the anti-SARS-CoV-2 mechanism, the in vitro and in vivo trials against SARS-CoV-2 have yielded mixed results. The toxicological effects and dosage optimization of chloroquine have been studied for many diseases, though it needs a proper evaluation again as chloroquine is also associated with several toxicities. Moreover, the drug is inexpensive and is readily available in many countries. Though much of the hope has been created by chloroquine and its derivatives against multiple diseases, repurposing it against SARS-CoV-2 requires large scale, collaborative, randomized and unbiased clinical trials to avoid false promises. This review summarizes the use and the mechanism of chloroquine against multiple diseases, its side-effects, mechanisms and the different clinical trials ongoing against "COVID-19".
Collapse
Affiliation(s)
| | - Madhuree Kumari
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
18
|
Kambey PA, Chengcheng M, Xiaoxiao G, Abdulrahman AA, Kanwore K, Nadeem I, Jiao W, Gao D. The orphan nuclear receptor Nurr1 agonist amodiaquine mediates neuroprotective effects in 6-OHDA Parkinson's disease animal model by enhancing the phosphorylation of P38 mitogen-activated kinase but not PI3K/AKT signaling pathway. Metab Brain Dis 2021; 36:609-625. [PMID: 33507465 DOI: 10.1007/s11011-021-00670-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/07/2021] [Indexed: 01/23/2023]
Abstract
Recent studies implicate the defects or altered expression of the orphan nuclear receptor Nurr1 gene in the substantia nigra in Parkinson's disease pathogenesis. In an attempt to corroborate the treatment-modifying disease that would replicate the effect of Nurr1, it has been found that amodiaquine and Nurr1 had the same chemical scaffolding, indicating a crucial structure-activity relationship. Interestingly, amodiaquine stimulate the transcriptional function of Nurr1 by physical interaction with its ligand-binding domain (LBD). However, the signaling route by which Nurr1 is activated by amodiaquine to cause the protective effect remains to be elucidated. We first demonstrated that amodiaquine treatment ameliorated behavioural deficits in 6-OHDA Parkinson's disease mouse model, and it promoted dopaminergic neurons protection signified by Tyrosine hydroxylase (TH) and dopamine transporter (DAT) mRNA; Tyrosine hydroxylase (TH) protein expression level and the immunoreactivity in the substantia nigra compacta. Subsequently, we used inhibitors to ascertain the effect of amodiaquine on Akt and P38 Mapk as crucial signaling pathways for neuroprotection. Wortmannin (Akt Inhibitor) induced a significant reduction of Akt mRNA; however, there was no statistical difference between the amodiaquine-treated group and the control group suggesting that amodiaquine may not be the active stimulant of Akt. Western blot analysis confirmed that the phosphorylated Akt decreased significantly in the amodiaquine group compared to the control group. In the same vein, we found that amodiaquine substantially increased the level of phosphorylated P38 Mapk. When P38 Mapk inhibited by SB203580 (P38-Mapk Inhibitor), the total P38 Mapk but not the phosphorylated P38 Mapk decreased significantly, while tyrosine hydroxylase significantly increased. These results collectively suggest that amodiaquine can augment tyrosine hydroxylase expression via phosphorylated P38 Mapk while negatively regulating the phosphorylated Akt in protein expression.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ma Chengcheng
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Guo Xiaoxiao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ayanlaja Abiola Abdulrahman
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Iqra Nadeem
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wu Jiao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
19
|
Kayamba F, Malimabe T, Ademola IK, Pooe OJ, Kushwaha ND, Mahlalela M, van Zyl RL, Gordon M, Mudau PT, Zininga T, Shonhai A, Nyamori VO, Karpoormath R. Design and synthesis of quinoline-pyrimidine inspired hybrids as potential plasmodial inhibitors. Eur J Med Chem 2021; 217:113330. [PMID: 33744688 DOI: 10.1016/j.ejmech.2021.113330] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/16/2023]
Abstract
Presently, artemisinin-based combination therapy (ACT) is the first-line therapy of Plasmodium falciparum malaria. With the emergence of malaria parasites that are resistant to ACT, alternative antimalarial therapies are urgently needed. In line with this, we designed and synthesised a series of novel N-(7-chloroquinolin-4-yl)-N'-(4,6-diphenylpyrimidin-2-yl)alkanediamine hybrids (6a-7c) and evaluated their inhibitory activity against the NF54 chloroquine-susceptible strain as a promising class of antimalarial compounds. The antiplasmodial screening revealed that seven analogues showed promising to good activity with half-maximal inhibitory concentration (IC50) = 0.32 μM-4.30 μM. Compound 7a with 1,4-diamine butyl linker and 4-hydroxyl phenyl on fourth and sixth position of pyrimidine core showed the most prominent activity with an IC50 value of 0.32 ± 0.06 μM, with a favourable safety profile of 9.79 to human kidney epithelial (HEK293) cells. The remaining six analogues showed moderate activity with IC50 values ranging from 7.50 μM to 83.01 μM. We further investigated the binding affinities of the molecules to two essential cytosolic P. falciparum heat shock protein 70 homologues; PfHsp70-1 and PfHsp70-z. Compound 7a exhibited the highest binding affinity for both PfHsp70s with KD in a lower nanomolar range (4.4-11.4 nM). Furthermore, molecular docking revealed that compounds 6, 6k, 7b and 7a exhibited better fitness in PfHsp70-1 with compound 7a showing the highest and lowest binding scores of -9.8 kcal/mol. Therefore, we speculate that PfHsp70-1 is one of the targets of these inhibitors. The bioisoteric replacement of the groups at phenyl ring at the fourth and sixth position of the pyrimidine core had a constructive association with antiplasmodial activity. The promising antiplasmodial activity of the synthesised analogues illustrates how crucial molecular hybridisation is as a strategy in the development of quinoline-pyrimidine hybrids as prospective antiprotozoal agents.
Collapse
Affiliation(s)
- Francis Kayamba
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Teboho Malimabe
- Pharmacology Division, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa; WITS Research Institute for Malaria (WRIM), Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Idowu Kehinde Ademola
- School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Ofentse Jacob Pooe
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Mavela Mahlalela
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Robyn L van Zyl
- Pharmacology Division, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa; WITS Research Institute for Malaria (WRIM), Faculty of Health Sciences, University of Witwatersrand, Johannesburg, 2193, South Africa
| | - Michelle Gordon
- School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Pertunia T Mudau
- Department of Biochemistry University of Venda, School of Mathematical and Natural Sciences, Thohoyandou, 0950, South Africa
| | - Tawanda Zininga
- Department of Biochemistry University of Venda, School of Mathematical and Natural Sciences, Thohoyandou, 0950, South Africa; Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Addmore Shonhai
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Vincent O Nyamori
- School of Chemistry and Physics, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa.
| |
Collapse
|
20
|
Hajimolaali M, Mohammadian H, Torabi A, Shirini A, Khalife Shal M, Barazandeh Nezhad H, Iranpour S, Baradaran Eftekhari R, Dorkoosh F. Application of chloroquine as an endosomal escape enhancing agent: new frontiers for an old drug. Expert Opin Drug Deliv 2021; 18:877-889. [PMID: 33455479 DOI: 10.1080/17425247.2021.1873272] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Adequate transfection efficiency is indispensable to safe and effective delivery of therapeutically active agents, particularly in cancer. Endosomal escape is regarded as a critical and determining step devoted a significant number of studies of the drug/gene delivery field. AREAS COVERED This paper critically reviews the fundamental properties of chloroquine (CQ), its pharmacokinetics, pharmacodynamics, and clinical applications and the present knowledge of CQ application as an endosomal escape enhancing agent. Different approaches to enhance the endosomal escape process of nanoparticles have been introduced including use of endosomal escape enhancing agents. Application of CQ as either a pre-treatment modality in which cells or animals are exposed to CQ prior to the main treatment or a component of co-delivery systems where CQ and other anti-cancer agents are simultaneously entered the cancer cells, is discussed with recent studies. EXPERT OPINION CQ is founded to intervene with the natural process of endosomal maturation. Moreover, CQ seems to increase the effectiveness of gene delivery by its electrostatic interaction with negatively charged components of the transferred genetic molecules. Endosomal escape might be regarded as the bottleneck of efficient gene delivery and CQ as an effective and available endosomal escape enhancing agent deserves more sophisticated studies.
Collapse
Affiliation(s)
- Mohammad Hajimolaali
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Pátrai, Greece
| | - Hosein Mohammadian
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Torabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Shirini
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Khalife Shal
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sheida Iranpour
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Baradaran Eftekhari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread worldwide since its first incidence in Wuhan, China, in December 2019. Although the case fatality rate of COVID-19 appears to be lower than that of SARS and Middle East respiratory syndrome (MERS), the higher transmissibility of SARS-CoV-2 has caused the total fatality to surpass other viral diseases, reaching more than 1 million globally as of October 6, 2020. The rate at which the disease is spreading calls for a therapy that is useful for treating a large population. Multiple intersecting viral and host factor targets involved in the life cycle of the virus are being explored. Because of the frequent mutations, many coronaviruses gain zoonotic potential, which is dependent on the presence of cell receptors and proteases, and therefore the targeting of the viral proteins has some drawbacks, as strain-specific drug resistance can occur. Moreover, the limited number of proteins in a virus makes the number of available targets small. Although SARS-CoV and SARS-CoV-2 share common mechanisms of entry and replication, there are substantial differences in viral proteins such as the spike (S) protein. In contrast, targeting cellular factors may result in a broader range of therapies, reducing the chances of developing drug resistance. In this Review, we discuss the role of primary host factors such as the cell receptor angiotensin-converting enzyme 2 (ACE2), cellular proteases of S protein priming, post-translational modifiers, kinases, inflammatory cells, and their pharmacological intervention in the infection of SARS-CoV-2 and related viruses.
Collapse
Affiliation(s)
- Anil Mathew Tharappel
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Subodh Kumar Samrat
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Zhong Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY 12208, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY 12201, USA
| |
Collapse
|
22
|
Melatonin potentials against viral infections including COVID-19: Current evidence and new findings. Virus Res 2020; 287:198108. [PMID: 32768490 PMCID: PMC7405774 DOI: 10.1016/j.virusres.2020.198108] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022]
Abstract
Viral infections are dangerous diseases for human health worldwide, which lead to significant morbidity and mortality each year. Because of their importance and the lack of effective therapeutic approaches, further attempts should be made to discover appropriate alternative or complementary treatments. Melatonin, a multifunctional neurohormone mainly synthesized and secreted by the pineal gland, plays some roles in the treatment of viral infections. Regarding a deadly outbreak of COVID-19 across the world, we decided to discuss melatonin functions against various viral infections including COVID-19. Therefore, in this review, we summarize current evidence on melatonin therapy for viral infections with focus on possible underlying mechanisms of melatonin actions.
Collapse
|
23
|
Kalra RS, Tomar D, Meena AS, Kandimalla R. SARS-CoV-2, ACE2, and Hydroxychloroquine: Cardiovascular Complications, Therapeutics, and Clinical Readouts in the Current Settings. Pathogens 2020; 9:E546. [PMID: 32645974 PMCID: PMC7400328 DOI: 10.3390/pathogens9070546] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/30/2020] [Accepted: 07/05/2020] [Indexed: 01/08/2023] Open
Abstract
The rapidly evolving coronavirus disease 2019 (COVID-19, caused by severe acute respiratory syndrome coronavirus 2- SARS-CoV-2), has greatly burdened the global healthcare system and led it into crisis in several countries. Lack of targeted therapeutics led to the idea of repurposing broad-spectrum drugs for viral intervention. In vitro analyses of hydroxychloroquine (HCQ)'s anecdotal benefits prompted its widespread clinical repurposing globally. Reports of emerging cardiovascular complications due to its clinical prescription are revealing the crucial role of angiotensin-converting enzyme 2 (ACE2), which serves as a target receptor for SARS-CoV-2. In the present settings, a clear understanding of these targets, their functional aspects and physiological impact on cardiovascular function are critical. In an up-to-date format, we shed light on HCQ's anecdotal function in stalling SARS-CoV-2 replication and immunomodulatory activities. While starting with the crucial role of ACE2, we here discuss the impact of HCQ on systemic cardiovascular function, its associated risks, and the scope of HCQ-based regimes in current clinical settings. Citing the extent of HCQ efficacy, the key considerations and recommendations for the use of HCQ in clinics are further discussed. Taken together, this review provides crucial insights into the role of ACE2 in SARS-CoV-2-led cardiovascular activity, and concurrently assesses the efficacy of HCQ in contemporary clinical settings.
Collapse
Affiliation(s)
- Rajkumar Singh Kalra
- AIST-INDIA DAILAB, DBT-AIST International Center for Translational & Environmental Research (DAICENTER), National Institute of Advanced Industrial Science & Technology (AIST), Higashi 1-1-1, Tsukuba 305 8565, Japan
| | - Dhanendra Tomar
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Avtar Singh Meena
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Uppal Road, Hyderabad 500 007, Telangana State, India;
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India;
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, Telangana State, India
| |
Collapse
|
24
|
Yang C, Wu HL, Li ZH, Chen XC, Su HY, Guo XY, An N, Jing KP, Pan QJ, Liu HF. Autophagy Inhibition Sensitizes Renal Tubular Epithelial Cell to G1 Arrest Induced by Transforming Growth Factor beta (TGF-β). Med Sci Monit 2020; 26:e922673. [PMID: 32555132 PMCID: PMC7297028 DOI: 10.12659/msm.922673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Cell cycle arrest and autophagy have been demonstrated to be involved in various transforming growth factor (TGF)-β-mediated phenotype alterations of tubular epithelial cells (TECs) and tubulointerstitial fibrosis. But the relationship between cell cycle arrest and the autophagy induced by TGF-β has not been explored well. Material/Methods The effects of autophagy inhibition on TGF-β-induced cell cycle arrest in TECs were explored in vitro. Human kidney-2 (HK-2) cells were stimulated by TGF-β with or without a combined treatment of autophagy inhibitor chloroquine (CQ) or bafilomycin A1 (Baf). Results Autophagy inhibition by CQ or Baf promotes the suppression of growth in TGF-β-treated HK-2 cells, as detected by the Cell Counting Kit-8 (CCK-8) method. In addition, CQ or Baf stimulation enhances G1 arrest in TGF-β treated HK-2 cells, as investigated using propidium iodide (PI) staining and flow cytometry, which was further confirmed by a decrease in the expression of phosphorylated retinoblastoma protein (p-RB) and cyclin-dependent kinase 4 (CDK4). The upregulation of p21 induced by CQ or Baf may mediate an enhanced G1 arrest in TGF-β treated HK-2 cells. Western blot analysis showed that TGF-β-induced expression of extracellular matrix fibronectin was notably upregulated in the presence of autophagy inhibitors. Conclusions Inhibition of autophagy sensitizes the TECs to G1 arrest and proliferation suppression induced by TGF-β that contributes to the induction of tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Chen Yang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Hong-Luan Wu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Zhi-Hang Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Xiao-Cui Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Hong-Yong Su
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Xiao-Yan Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Ning An
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Kai-Peng Jing
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Qing-Jun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China (mainland)
| |
Collapse
|
25
|
Uddin M, Mustafa F, Rizvi TA, Loney T, Al Suwaidi H, Al-Marzouqi AHH, Kamal Eldin A, Alsabeeha N, Adrian TE, Stefanini C, Nowotny N, Alsheikh-Ali A, Senok AC. SARS-CoV-2/COVID-19: Viral Genomics, Epidemiology, Vaccines, and Therapeutic Interventions. Viruses 2020; 12:E526. [PMID: 32397688 PMCID: PMC7290442 DOI: 10.3390/v12050526] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic is due to infection caused by the novel SARS-CoV-2 virus that impacts the lower respiratory tract. The spectrum of symptoms ranges from asymptomatic infections to mild respiratory symptoms to the lethal form of COVID-19 which is associated with severe pneumonia, acute respiratory distress, and fatality. To address this global crisis, up-to-date information on viral genomics and transcriptomics is crucial for understanding the origins and global dispersion of the virus, providing insights into viral pathogenicity, transmission, and epidemiology, and enabling strategies for therapeutic interventions, drug discovery, and vaccine development. Therefore, this review provides a comprehensive overview of COVID-19 epidemiology, genomic etiology, findings from recent transcriptomic map analysis, viral-human protein interactions, molecular diagnostics, and the current status of vaccine and novel therapeutic intervention development. Moreover, we provide an extensive list of resources that will help the scientific community access numerous types of databases related to SARS-CoV-2 OMICs and approaches to therapeutics related to COVID-19 treatment.
Collapse
Affiliation(s)
- Mohammed Uddin
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Farah Mustafa
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE; (F.M.); (A.H.H.A.-M.)
| | - Tahir A. Rizvi
- Department of Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE;
| | - Tom Loney
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
| | - Hanan Al Suwaidi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
| | - Ahmed H. Hassan Al-Marzouqi
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE; (F.M.); (A.H.H.A.-M.)
| | - Afaf Kamal Eldin
- Department of Food, Nutrition and Health, United Arab Emirates University, Al Ain, UAE;
| | | | - Thomas E. Adrian
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
| | - Cesare Stefanini
- Department of Biomedical Engineering, Healthcare Engineering Innovation Center (HEIC), Khalifa University, Abu Dhabi, UAE;
| | - Norbert Nowotny
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
- Viral Zoonoses, Emerging and Vector-Borne Infections Group, Institute of Virology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Alawi Alsheikh-Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
| | - Abiola C. Senok
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE; (M.U.); (T.L.); (H.A.S.); (T.E.A.); (N.N.)
| |
Collapse
|
26
|
Meyerowitz EA, Vannier AGL, Friesen MGN, Schoenfeld S, Gelfand JA, Callahan MV, Kim AY, Reeves PM, Poznansky MC. Rethinking the role of hydroxychloroquine in the treatment of COVID-19. FASEB J 2020; 34:6027-6037. [PMID: 32350928 PMCID: PMC7267640 DOI: 10.1096/fj.202000919] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023]
Abstract
There are currently no proven or approved treatments for coronavirus disease 2019 (COVID-19). Early anecdotal reports and limited in vitro data led to the significant uptake of hydroxychloroquine (HCQ), and to lesser extent chloroquine (CQ), for many patients with this disease. As an increasing number of patients with COVID-19 are treated with these agents and more evidence accumulates, there continues to be no high-quality clinical data showing a clear benefit of these agents for this disease. Moreover, these agents have the potential to cause harm, including a broad range of adverse events including serious cardiac side effects when combined with other agents. In addition, the known and potent immunomodulatory effects of these agents which support their use in the treatment of auto-immune conditions, and provided a component in the original rationale for their use in patients with COVID-19, may, in fact, undermine their utility in the context of the treatment of this respiratory viral infection. Specifically, the impact of HCQ on cytokine production and suppression of antigen presentation may have immunologic consequences that hamper innate and adaptive antiviral immune responses for patients with COVID-19. Similarly, the reported in vitro inhibition of viral proliferation is largely derived from the blockade of viral fusion that initiates infection rather than the direct inhibition of viral replication as seen with nucleoside/tide analogs in other viral infections. Given these facts and the growing uncertainty about these agents for the treatment of COVID-19, it is clear that at the very least thoughtful planning and data collection from randomized clinical trials are needed to understand what if any role these agents may have in this disease. In this article, we review the datasets that support or detract from the use of these agents for the treatment of COVID-19 and render a data informed opinion that they should only be used with caution and in the context of carefully thought out clinical trials, or on a case-by-case basis after rigorous consideration of the risks and benefits of this therapeutic approach.
Collapse
Affiliation(s)
- Eric A. Meyerowitz
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
| | - Augustin G. L. Vannier
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Morgan G. N. Friesen
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Sara Schoenfeld
- Division of Allergy, Immunology and RheumatologyMGH and HMSBostonMAUSA
| | - Jeffrey A. Gelfand
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Michael V. Callahan
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
- Special Advisor to the Assistant Secretary of Public Health Preparedness and Response U.S Dept of Health and Human ServicesWashingtonDCUSA
| | - Arthur Y. Kim
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
| | - Patrick M. Reeves
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Mark C. Poznansky
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| |
Collapse
|
27
|
Maruta H, Kittaka A. Chemical evolution for taming the 'pathogenic kinase' PAK1. Drug Discov Today 2020; 25:959-964. [PMID: 32348877 PMCID: PMC7194552 DOI: 10.1016/j.drudis.2020.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 01/14/2023]
Abstract
PAK1 is the major ‘pathogenic’ kinase. Several potent PAK1 blockers developed are introduced for treatment of a wide variety of PAK1-dependent diseases including cancers and pandemic COVID-19 infection.
To celebrate the 25th anniversary of the cloning of the first mammalian p21-activated kinases (PAKs) (RAC/CDC42-activated kinases) by Ed Manser, the first international PAK symposium was held in NYC in October 2019. Among six distinct PAKs in mammals, PAK1 is the major ‘pathogenic kinase’, the abnormal activation of which is responsible for a wide variety of diseases and disorders including cancers, ageing processes and infectious and inflammatory diseases such as pandemic coronaviral infection. Recently, for a clinical application, a few potent (highly cell-permeable and water-soluble) PAK1 blockers have been developed from natural or synthetic PAK1 blockers (triptolide, vitamin D3 and ketorolac) via a series of ‘chemical evolutions’ that boost pharmacological activities >500 times.
Collapse
|
28
|
Maruta H, He H. PAK1-blockers: Potential Therapeutics against COVID-19. MEDICINE IN DRUG DISCOVERY 2020; 6:100039. [PMID: 32313880 PMCID: PMC7166201 DOI: 10.1016/j.medidd.2020.100039] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
PAK1 (RAC/CDC42-activated kinase 1) is the major “pathogenic” kinase whose abnormal activation causes a wide variety of diseases/disorders including cancers, inflammation, malaria and pandemic viral infection including influenza, HIV and COVID-19. Since Louis Pasteur who developed a vaccine against rabies in 1885, in general a series of “specific” vaccines have been used for treatment of viral infection, mainly because the majority of pre-existing antibiotics are either anti-bacterial or anti-fungal, thereby being ineffective against viruses in general. However, it takes 12–18 months till the effective vaccine becomes available. Until then ventilator (O2 supplier) would be the most common tool for saving the life of COVID-19 patients. Thus, as alternative potentially more direct “broad-spectrum” signalling mechanism–based COVID-19 therapeutics, several natural and synthetic PAK1-blockers such as propolis, melatonin, ciclesonide, hydroxy chloroquine (HQ), ivermection, and ketorolac, which are readily available in the market, are introduced here. PAK1 is the major “pathogenic “kinase essential for infection of many viruses including COVID-19. Several PAK1-blockers such as propolis, melatonin, anti-malaria drugs, ivermectin, cicloresonide and ketorolac are readily available in the market. PAK1-blockers, interfering with the pathogenic process as well as promoting immune system, could serve as potential therapeutic agents against COVID-19.
Collapse
Affiliation(s)
| | - Hong He
- University of Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
29
|
Barker BS, Spampanato J, McCarren HS, Smolik M, Jackson CE, Hornung EN, Yeung DT, Dudek FE, McDonough JH. Screening for Efficacious Anticonvulsants and Neuroprotectants in Delayed Treatment Models of Organophosphate-induced Status Epilepticus. Neuroscience 2020; 425:280-300. [PMID: 31783100 PMCID: PMC6935402 DOI: 10.1016/j.neuroscience.2019.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 01/04/2023]
Abstract
Organophosphorus (OP) compounds are deadly chemicals that exert their intoxicating effects through the irreversible inhibition of acetylcholinesterase (AChE). In addition to an excess of peripheral ailments, OP intoxication induces status epilepticus (SE) which if left untreated may lead to permanent brain damage or death. Benzodiazepines are typically the primary therapies for OP-induced SE, but these drugs lose efficacy as treatment time is delayed. The CounterACT Neurotherapeutic Screening (CNS) Program was therefore established by the National Institutes of Health (NIH) to discover novel treatments that may be administered adjunctively with the currently approved medical countermeasures for OP-induced SE in a delayed treatment scenario. The CNS program utilizes in vivo EEG recordings and Fluoro-JadeB (FJB) histopathology in two established rat models of OP-induced SE, soman (GD) and diisopropylfluorophosphate (DFP), to evaluate the anticonvulsant and neuroprotectant efficacy of novel adjunct therapies when administered at 20 or 60 min after the induction of OP-induced SE. Here we report the results of multiple compounds that have previously shown anticonvulsant or neuroprotectant efficacy in other models of epilepsy or trauma. Drugs tested were ganaxolone, diazoxide, bumetanide, propylparaben, citicoline, MDL-28170, and chloroquine. EEG analysis revealed that ganaxolone demonstrated the most robust anticonvulsant activity, whereas all other drugs failed to attenuate ictal activity in both models of OP-induced SE. FJB staining demonstrated that none of the tested drugs had widespread neuroprotective abilities. Overall these data suggest that neurosteroids may represent the most promising anticonvulsant option for OP-induced SE out of the seven unique mechanisms tested here. Additionally, these results suggest that drugs that provide significant neuroprotection from OP-induced SE without some degree of anticonvulsant activity are elusive, which further highlights the necessity to continue screening novel adjunct treatments through the CNS program.
Collapse
Affiliation(s)
- Bryan S Barker
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA.
| | - Jay Spampanato
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Hilary S McCarren
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Melissa Smolik
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Cecelia E Jackson
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - Eden N Hornung
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| | - David T Yeung
- Chemical Countermeasures Research Program, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA
| | - F Edward Dudek
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - John H McDonough
- Medical Toxicology Research Division, Neuroscience Department, U.S. Army Medical Research Institute of Chemical Defense, 8350 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, USA
| |
Collapse
|
30
|
T-lymphocytes response persists following Plasmodium berghei strain Anka infection resolution and may contribute to later experimental cerebral malaria outcomes. J Neuroimmunol 2019; 330:5-11. [PMID: 30763800 DOI: 10.1016/j.jneuroim.2019.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 01/20/2023]
Abstract
Several studies have proposed cerebral malaria (CM) as a CD4+ and CD8+ T lymphocyte-mediated disease. However, there are no data regarding the recruitment and/or persistence of these cells in the CNS following the phase of infection resolution. Glutamate-mediate excitotoxicity has also been implicated in CM. Blockade of glutamate NMDA receptors by its noncompetitive antagonist MK801 modulates cytokine and neurotrophic factors expression preventing cognitive and depressive-like behavior in experimental CM. Herein, we aim to investigate the role of T lymphocytes in later outcomes in CM, and whether the protective role of MK801 is associated with T lymphocytes response.
Collapse
|
31
|
Kanvinde S, Chhonker YS, Ahmad R, Yu F, Sleightholm R, Tang W, Jaramillo L, Chen Y, Sheinin Y, Li J, Murry DJ, Singh AB, Oupický D. Pharmacokinetics and efficacy of orally administered polymeric chloroquine as macromolecular drug in the treatment of inflammatory bowel disease. Acta Biomater 2018; 82:158-170. [PMID: 30342282 DOI: 10.1016/j.actbio.2018.10.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 12/27/2022]
Abstract
Inflammatory bowel disease is a chronic inflammation of the gastrointestinal tract with poor understanding of its pathogenesis and no effective cure. The goal of this study was to evaluate the feasibility of orally administered non-degradable polymeric chloroquine (pCQ) to locally reduce colon inflammation. The pCQ was synthesized by radical copolymerization of N-(2-hydroxypropyl)methacrylamide with methacryloylated hydroxychloroquine (HCQ). The anti-inflammatory activity of orally administered pCQ versus HCQ was tested in a mouse model of colitis induced by Citrobacter rodentium (C. rodentium). Single-dose pharmacokinetic and biodistribution studies performed in the colitis model indicated negligible systemic absorption (p ≤ 0.001) and localization of pCQ in the gastrointestinal tract. A multi-dose therapeutic study demonstrated that the localized pCQ treatment resulted in significant reduction in the colon inflammation (p ≤ 0.05). Enhanced suppression of pro-inflammatory cytokines IL-6 (p ≤ 0.01) and IL1-β and opposing upregulation of IL-2 (p ≤ 0.05) recently reported to be involved in downstream anti-inflammatory events suggested that the anti-inflammatory effects of the pCQ are mediated by altering mucosal immune homeostasis. Overall, the reported findings demonstrate a potential of pCQ as a novel polymer therapeutic option in inflammatory bowel disease with the potential of local effects and minimized systemic toxicity.
Collapse
Affiliation(s)
- Shrey Kanvinde
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | | | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, United States
| | - Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Richard Sleightholm
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Lee Jaramillo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Yi Chen
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Yuri Sheinin
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Daryl J Murry
- Department of Pharmacy Practice, University of Nebraska Medical Center, United States
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
32
|
Jung HY, Kim B, Ryu HG, Ji Y, Park S, Choi SH, Lee D, Lee IK, Kim M, Lee YJ, Song W, Lee YH, Choi HJ, Hyun CK, Holzapfel WH, Kim KT. Amodiaquine improves insulin resistance and lipid metabolism in diabetic model mice. Diabetes Obes Metab 2018. [PMID: 29516607 DOI: 10.1111/dom.13284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS Although peroxisome proliferator-activated receptors (PPARs)α/γ dual agonists can be beneficial for treatment of dyslipidemia in patients with type 2 diabetes, their use is limited owing to various side effects, including body weight gain, edema, and heart failure. We aimed to demonstrate that amodiaquine, an antimalarial agent, has potential as a PPARα/γ dual agonist with low risk of adverse effects. METHODS We screened a Prestwick library (Prestwick Chemical; Illkirch, France) to identify novel PPARα/γ dual agonists and selected amodiaquine (4-[(7-chloroquinolin-4-yl)amino]-2-[(diethylamino)methyl]phenol), which activated both PPAR-α & -γ, for further investigation. We performed both in vitro, including glucose uptake assay and fatty acid oxidation assay, and in vivo studies to elucidate the anti-diabetic and anti-obesity effects of amodiaquine. RESULTS Amodiaquine selectively activated the transcriptional activities of PPARα/γ and enhanced both fatty acid oxidation and glucose uptake without altering insulin secretion in vitro. In high-fat diet-induced obese and genetically modified obese/diabetic mice, amodiaquine not only remarkably ameliorated insulin resistance, hyperlipidemia, and fatty liver but also decreased body weight gain. CONCLUSION Our findings suggest that amodiaquine exerts beneficial effects on glucose and lipid metabolism by concurrent activation of PPARα/γ. Furthermore, amodiaquine acts as an alternative insulin-sensitizing agent with a positive influence on lipid metabolism and has potential to prevent and treat type 2 diabetes while reducing the risk of lipid abnormalities.
Collapse
Affiliation(s)
- Hoe-Yune Jung
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- R&D Center, NovMetaPharma Co., Ltd., Pohang, Republic of Korea
| | - Bobae Kim
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Hye Guk Ryu
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yosep Ji
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
| | - Soyoung Park
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
| | - Seung Hee Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Dohyun Lee
- R&D Center, NovMetaPharma Co., Ltd., Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Munki Kim
- Bio Convergence Team, Advanced Bio Convergence Center, Pohang, Republic of Korea
| | - You Jeong Lee
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS), Pohang, Republic of Korea
| | - Woojin Song
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Division of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Hee Lee
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Division of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyung Jin Choi
- Functional Neuroanatomy of Metabolism Regulation Laboratory, Department of Anatomy, Division of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang-Kee Hyun
- School of Life Science, Handong Global University, Pohang, Republic of Korea
| | - Wilhelm H Holzapfel
- Department of Advanced Green Energy and Environment, Handong Global University, Pohang, Republic of Korea
| | - Kyong-Tai Kim
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| |
Collapse
|
33
|
Won HY, Shin JH, Oh S, Jeong H, Hwang ES. Enhanced CD25 +Foxp3 + regulatory T cell development by amodiaquine through activation of nuclear receptor 4A. Sci Rep 2017; 7:16946. [PMID: 29208963 PMCID: PMC5717225 DOI: 10.1038/s41598-017-17073-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/21/2017] [Indexed: 01/10/2023] Open
Abstract
CD4+ T cells play key roles in the regulation of immune responses against pathogenic infectious antigens via development into effector T helper and induced regulatory T (iTreg) cells. Particularly, CD4+CD25+Foxp3+ iTreg cells are crucial for maintaining immune homeostasis and controlling inflammatory diseases. Anti-inflammatory drugs that enhance iTreg cell generation would be effective at preventing and treating inflammatory and autoimmune diseases. In this study, we examined whether anti-malarial and anti-arthritic amodiaquine (AQ) could affect iTreg cell development. Despite the anti-proliferative activity of AQ, AQ only moderately decreased iTreg cell proliferation but substantially increased IL-2 production by iTreg cells. Furthermore, AQ dose-dependently increased iTreg cell development and significantly upregulated iTreg cell markers including CD25. Interestingly, CD25 expression was decreased at later stages of iTreg cell development but was sustained in the presence of AQ, which was independent of IL-2 signaling pathway. AQ directly increased CD25 gene transcription by enhancing the DNA-binding and transcriptional activity of nuclear receptor 4 A. Most importantly, in vivo administration of AQ attenuated inflammatory colitis, resulted in the increased iTreg cells and decreased inflammatory cytokines. The ability of anti-malarial AQ to potentiate iTreg cell development makes it a promising drug for preventing and treating inflammatory and autoimmune diseases.
Collapse
MESH Headings
- Amodiaquine/pharmacology
- Animals
- Cell Proliferation/drug effects
- Colitis/drug therapy
- Colitis/etiology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Forkhead Transcription Factors/metabolism
- Interleukin-2/metabolism
- Interleukin-2 Receptor alpha Subunit/genetics
- Interleukin-2 Receptor alpha Subunit/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Receptors, Interleukin-2/metabolism
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/physiology
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Hee Yeon Won
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Ji Hyun Shin
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Sera Oh
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Hana Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea.
| |
Collapse
|
34
|
Novel anti-adipogenic activity of anti-malarial amodiaquine through suppression of PPARγ activity. Arch Pharm Res 2017; 40:1336-1343. [PMID: 29071567 DOI: 10.1007/s12272-017-0965-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/26/2017] [Indexed: 01/17/2023]
Abstract
Amodiaquine (AQ) was developed as a selective drug against Plasmodium falciparum malaria infection and has received increasing attention as a therapeutic agent for the treatment of rheumatoid arthritis, Parkinson's disease, and cancer due to its anti-inflammatory, anti-proliferative, and autophagic-lysosomal blockade properties. As autophagy activation is involved in promoting adipogenic differentiation, we examined whether anti-autophagic AQ affected adipocyte differentiation of 3T3-L1 pre-adipocytes. AQ dose-dependently and significantly suppressed adipocyte differentiation in conjunction with decreases in lipid droplet formation and expression of adipogenic markers including adiponectin, adipocyte fatty acid-binding protein 2 (aP2), resistin, and leptin. Although peroxisome proliferator-activated receptor γ (PPARγ) decreases by inhibition of autophagy, AQ treatment did not induce PPARγ degradation despite the suppression of autophagolysosomal degradation. Instead, AQ suppressed the PPARγ activity to transcriptionally activate the aP2 gene transcription through the selective prevention of nuclear localization of PPARγ. These results demonstrated the novel anti-adipogenic activity of AQ and identified its underlying mechanism that AQ suppressed adipogenic gene expression and lipid formation by inhibiting nuclear localization of PPARγ in an autophagy-independent manner. AQ is recommended as a safe and effective anti-obesity drug for controlling overweight and obesity.
Collapse
|
35
|
Bae SJ, Min YK, Hwang ES. Potent osteogenic activity of a novel imidazobenzimidazole derivative, IBIP. Biochem Biophys Res Commun 2017; 487:409-414. [DOI: 10.1016/j.bbrc.2017.04.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/14/2017] [Indexed: 12/27/2022]
|
36
|
Won HY, Hwang ES. Transcriptional modulation of regulatory T cell development by novel regulators NR4As. Arch Pharm Res 2016; 39:1530-1536. [PMID: 27778276 DOI: 10.1007/s12272-016-0803-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023]
Abstract
Regulatory T (Treg) cells with high expression of both CD25 and Foxp3 are developed in the thymus and also peripheral tissues. Treg cells suppress the activation and functions of effector T cells raised against specific antigens and are crucial for maintaining immune homeostasis. Treg cell development is associated with the induction of and epigenetic alterations of forkhead transcription factor Foxp3. Foxp3 expression is increased by the activation of several transcription factors including nuclear factor-kappa B (NF-κB), nuclear factor of activated T cells (NFAT), and Smad3 in response to various signals such as TGFβ, retinoic acid, and rapamycin. Recently, the orphan nuclear receptor 4A proteins (NR4As) including NR4A1 (Nur77), NR4A2 (Nurr1), and NR4A3 (Nor1) are reported to regulate Treg cell development through activation of Foxp3 and have therapeutic potentials in treating immune disorders. This review summarizes the function and regulatory mechanisms of Treg cells and also implicates current advances in immunomodulatory functions of NR4As and their therapeutic potentials in inflammation and cancer.
Collapse
Affiliation(s)
- Hee Yeon Won
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea.
| |
Collapse
|