1
|
Zhou Q, Zheng Z, Yin S, Duan D, Liao X, Xiao Y, He J, Zhong J, Zeng Z, Su L, Luo L, Dong C, Chen J, Li J. Nicotinamide mitigates visceral leishmaniasis by regulating inflammatory response and enhancing lipid metabolism. Parasit Vectors 2024; 17:288. [PMID: 38971783 PMCID: PMC11227177 DOI: 10.1186/s13071-024-06370-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/21/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Currently, treatment regimens for visceral leishmaniasis (VL) are limited because of the presence of numerous adverse effects. Nicotinamide, a readily available and cost-effective vitamin, has been widely acknowledged for its safety profile. Several studies have demonstrated the anti-leishmanial effects of nicotinamide in vitro. However, the potential role of nicotinamide in Leishmania infection in vivo remains elusive. METHODS In this study, we assessed the efficacy of nicotinamide as a therapeutic intervention for VL caused by Leishmania infantum in an experimental mouse model and investigated its underlying molecular mechanisms. The potential molecular mechanism was explored through cytokine analysis, examination of spleen lymphocyte subsets, liver RNA-seq analysis, and pathway validation. RESULTS Compared to the infection group, the group treated with nicotinamide demonstrated significant amelioration of hepatosplenomegaly and recovery from liver pathological damage. The NAM group exhibited parasite reduction rates of 79.7% in the liver and 86.7% in the spleen, respectively. Nicotinamide treatment significantly reduced the activation of excessive immune response in infected mice, thereby mitigating hepatosplenomegaly and injury. Furthermore, nicotinamide treatment enhanced fatty acid β-oxidation by upregulating key enzymes to maintain lipid homeostasis. CONCLUSIONS Our findings provide initial evidence supporting the safety and therapeutic efficacy of nicotinamide in the treatment of Leishmania infection in BALB/c mice, suggesting its potential as a viable drug for VL.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhiwan Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
| | - Shuangshuang Yin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Dengbinpei Duan
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xuechun Liao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yuying Xiao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jinlei He
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
| | - Junchao Zhong
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Zeng
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
- Chong Qing Animal Disease Prevention and Control Center, Chongqing, China
| | - Liang Su
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
- Chong Qing Animal Disease Prevention and Control Center, Chongqing, China
| | - Lu Luo
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
- Chong Qing Animal Disease Prevention and Control Center, Chongqing, China
| | - Chunxia Dong
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China
- Chong Qing Animal Disease Prevention and Control Center, Chongqing, China
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China.
| | - Jiao Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.
- Sichuan-Chongqing jointly-established Research Platform of Zoonosis, Chengdu, China.
| |
Collapse
|
2
|
Yan Y, Liao Z, Shen J, Zhu Z, Cao Y. Nicotinamide potentiates amphotericin B activity against Candida albicans. Virulence 2022; 13:1533-1542. [PMID: 36068709 PMCID: PMC9467617 DOI: 10.1080/21505594.2022.2119656] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Amphotericin B (AmB) is a widely used antifungal agent especially for the therapy of systemic fungal infections. However, the severe side effects of AmB often leads to the premature termination of the treatment. So it is imperative to find the drugs that can both reduce the dosage and enhance the antifungal efficacy of AmB. Here we demonstrated that Nicotinamide (NAM), a cheap and safe vitamin, could enhance the antifungal activities of AmB. We demonstrated the synergistic interaction of NAM and AmB against Candida albicans as well as other Candida spp. and Cryptococcus neoformans. Moreover, NAM could enhance of the activity of AmB against biofilm. This enhancement was also observed in disseminated candidiasis in vivo. Our further study revealed that AmB could induce oxidative damage through the modification of histone acetylation. AmB could inhibit the expression of HST3, an H3K56 deacetylase in C. albicans. The immunoblotting test revealed excessive H3K56ac in AmB-treated fungal cells. Consistantly, the hst3Δ mutant displayed high sensitivity to AmB, while addition of NAM, an H3K56 deacetylation inhibitor, resulted in an even severe inhibition in the growth of this strain. These results indicated that AmB could execute antifungal activity via boosting H3K56ac which was mediated by HST3, and the mechanism for the synergistic interaction of NAM and AmB was based on exacerbating this process, which led to even excessive H3K56ac and oxidative damage. This finding provided theoretical basis for better understanding the antifungal mechanisms of AmB and clinical application of this drug.
Collapse
Affiliation(s)
- Yu Yan
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - ZeBin Liao
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Juan Shen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China.,Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - ZhenYu Zhu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - YingYing Cao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Cao P, Chen Q, Shi CX, Wang LW, Gong ZJ. Sirtuin1 attenuates acute liver failure by reducing reactive oxygen species via hypoxia inducible factor 1α. World J Gastroenterol 2022; 28:1798-1813. [PMID: 35633910 PMCID: PMC9099200 DOI: 10.3748/wjg.v28.i17.1798] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/21/2021] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The occurrence and development of acute liver failure (ALF) is closely related to a series of inflammatory reactions, such as the production of reactive oxygen species (ROS). Hypoxia inducible factor 1α (HIF-1α) is a key factor that regulates oxygen homeostasis and redox, and the stability of HIF-1α is related to the ROS level regulated by Sirtuin (Sirt) family. The activation of Sirt1 will lead to a powerful antioxidant defense system and therapeutic effects in liver disease. However, little is known about the relationship between HIF-1α and Sirt1 in the process of ALF and the molecular mechanism.
AIM To investigate whether HIF-1α may be a target of Sirt1 deacetylation and what the effects on ALF are.
METHODS Mice were administrated lipopolysaccharide (LPS)/D-gal and exposed to hypoxic conditions as animal model, and resveratrol was used as an activator of Sirt1. The cellular model was established with L02 cells stimulated by LPS. N-acetyl-L-cysteine was used to remove ROS, and the expression of Sirt1 was inhibited by nicotinamide. Western blotting was used to detect Sirt1 and HIF-1α activity and related protein expression. The possible signaling pathways involved were analyzed by immunofluorescent staining, co-immunoprecipitation, dihydroethidium staining, and Western blotting.
RESULTS Compared with mice stimulated with LPS alone, the expression of Sirt1 decreased, the level of HIF-1α acetylation increased in hypoxic mice, and the levels of carbonic anhydrase 9 and Bcl-2-adenovirus E1B interacting protein 3 increased significantly, which was regulated by HIF-1α, indicating an increase of HIF-1α activity. Under hypoxia, the down-regulation of Sirt1 activated and acetylated HIF-1α in L02 cells. The inhibition of Sirt1 significantly aggravated this effect and the massive production of ROS. The regulation of ROS was partly through peroxisome proliferator-activated receptor alpha or AMP-activated protein kinase. Resveratrol, a Sirt1 activator, effectively relieved ALF aggravated by hypoxia, the production of ROS, and cell apoptosis. It also induced the deacetylation of HIF-1α and inhibited the activity of HIF-1α.
CONCLUSION Sirt1 may have a protective effect on ALF by inducing HIF-1α deacetylation to reduce ROS.
Collapse
Affiliation(s)
- Pan Cao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Qian Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Chun-Xia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Lu-Wen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Zuo-Jiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| |
Collapse
|
4
|
Ghugari R, Tsao S, Schmidt M, Bonneil É, Brenner C, Verreault A. Mechanisms to reduce the cytotoxicity of pharmacological nicotinamide concentrations in the pathogenic fungus Candida albicans. FEBS J 2021; 288:3478-3506. [PMID: 33155404 DOI: 10.1111/febs.15622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/13/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022]
Abstract
Candida albicans is a pathogenic fungus that causes systemic infections and mortality in immunosuppressed individuals. We previously showed that deacetylation of histone H3 lysine 56 by Hst3 is essential for C. albicans viability. Hst3 is a fungal-specific NAD+ -dependent protein deacetylase of the sirtuin family. In vivo, supraphysiological concentrations of nicotinamide (NAM) are required for Hst3 inhibition and cytotoxicity. This underscores the importance of identifying mechanisms by which C. albicans can modulate intracellular NAM concentrations. For the first time in a pathogenic fungus, we combine genetics, heavy isotope labeling, and targeted quantitative metabolomics to identify genes, pathways, and mechanisms by which C. albicans can reduce the cytotoxicity of high NAM concentrations. We discovered three distinct fates for supraphysiological NAM concentrations. First, upon transient exposure to NAM, high intracellular NAM concentrations rapidly return near the physiological levels observed in cells that are not exposed to NAM. Second, during the first step of a fungal-specific NAM salvage pathway, NAM is converted into nicotinic acid, a metabolite that cannot inhibit the sirtuin Hst3. Third, we provide evidence that NAM enters the NAD+ metabolome through a NAM exchange reaction that contributes to NAM-mediated inhibition of sirtuins. However, in contrast to the other fates of NAM, the NAM exchange reaction cannot cause a net decrease in the intracellular concentration of NAM. Therefore, this reaction cannot enhance resistance to NAM. In summary, we demonstrate that C. albicans possesses at least two mechanisms to attenuate the cytotoxicity of pharmacological NAM concentrations. It seems likely that those two mechanisms of resistance to cytotoxic NAM concentrations are conserved in many other pathogenic fungi.
Collapse
Affiliation(s)
- Rahul Ghugari
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
- Programme de Biologie Moléculaire, Université de Montréal, QC, Canada
| | - Sarah Tsao
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
| | - Mark Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, IA, USA
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
| | - Charles Brenner
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Alain Verreault
- Institute for Research in Immunology and Cancer, Université de Montréal, QC, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, QC, Canada
| |
Collapse
|
5
|
Oliaee RT, Sharifi I, Bamorovat M, Keyhani A, Babaei Z, Salarkia E, Tavakoly R, Khosravi A, Mostafavi M, Sharifi F, Mousavi SM. The potential role of nicotinamide on Leishmania tropica: An assessment of inhibitory effect, cytokines gene expression and arginase profiling. Int Immunopharmacol 2020; 86:106704. [PMID: 32590317 DOI: 10.1016/j.intimp.2020.106704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/30/2020] [Accepted: 06/11/2020] [Indexed: 11/26/2022]
Abstract
Leishmaniasis represents a major health concern worldwide which has no effective treatment modality. Nicotinamide (NAm) has been used for a wide range of applications from anticancer to antimicrobial usage. This study aimed to assess the effect of NAm combination on Leishmania tropica Inhibition, as well as on cytokines gene expression and arginase (ARG) activity in L. tropica-infected macrophages in an in vitro model. The leishmanicidal effects of NAm and Glucantime (meglumine antimoniate, MA) alone and in combination (NAm/MA) were evaluated using a colorimetric assay and macrophage model. Additionally, immunomodulatory effects and enzymatic activity were assessed by analyzing Th1 and Th2 cytokines gene expression and ARG level, respectively, in infected macrophages treated with NAm and MA, alone and in combination. Findings indicated that the NAm/MA combination demonstrated greater inhibitory effects on L. tropica promastigotes and amastigotes compared with each drug individually. Docking results proved the affinity of NAm to IFN-γ, which can affirm the increased levels of IFN-γ, IL-12p40 and TNF-α as well as reductions in IL-10 secretion with a dose-response effect, especially in the combination group. The NAm/MA combination also showed a significant reduction in the level of ARG activity at all concentrations used compared to each drug individually. These findings indicate higher effectiveness of NAm plus MA in reducing parasite growth, promoting immune response and inhibiting ARG level. This combination should be considered as a potential therapeutic regimen for treatment of volunteer patients with anthroponotic cutaneous leishmaniasis (ACL) in future control programs.
Collapse
Affiliation(s)
- Razieh Tavakoli Oliaee
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran; Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Bamorovat
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Alireza Keyhani
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Babaei
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ehsan Salarkia
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Rahele Tavakoly
- Student Research Committee, School of Health, Kerman University of Medical Sciences, Kerman, Iran; Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahshid Mostafavi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Sharifi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Mohammad Mousavi
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
In vitro effectivity of three approved drugs and their synergistic interaction against Leishmania infantum. ACTA ACUST UNITED AC 2020; 40:89-101. [PMID: 32463611 PMCID: PMC7449103 DOI: 10.7705/biomedica.4891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Indexed: 12/24/2022]
Abstract
Introduction: Leishmaniasis remains one of the neglected tropical diseases. Repurposing existing drugs has proven to be successful for treating neglected tropical diseases while combination therapy is a strategic alternative for the treatment of infectious diseases. Auranofin, lopinavir/ritonavir, and sorafenib are FDA approved drugs used in the treatment of diverse diseases by acting on different essential biological enzymes. Objective: To evaluate the effects of monotherapy and combined therapies with the three drugs against Leishmania infantum. Materials and methods: We compared the leishmanicidal effects of the three drugs on promastigotes in vitro as regards the parasite count, the drug concentration providing a half-maximal response, and the ultrastructural changes of the parasite. We determined the fractional inhibitory concentration index of combined drugs in two ways, as well as the activity of the three drugs together to establish their synergetic effect. Results: The monotherapy with the three drugs was effective with auranofin showing the best leishmanicidal effect (EC50=1.5 µM), whereas sorafinib reduced parasite growth at EC50=2.5 µM. The scanning electron microscopy of promastigotes from all treated media showed distortion in the shape with loss of flagella and bleb formation. Acidocalcinosis was evident by transmission electron microscopy with all treatments suggesting apoptosis. Treatment with lopinavir/ritonavir showed signs of autophagy. The two-way combination of the drugs led to additive interactions while the combination of the three drugs showed synergistic action. Conclusion: Each drug when used as monotherapy against Leishmania spp. was effective, but the combination therapy was more effective than the individual drugs due to the additive or synergistic effects.
Collapse
|
7
|
Nicotinamide induces G2 cell cycle arrest in Giardia duodenalis trophozoites and promotes changes in sirtuins transcriptional expression. Exp Parasitol 2020; 209:107822. [DOI: 10.1016/j.exppara.2019.107822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 11/15/2022]
|
8
|
The superfamily keeps growing: Identification in trypanosomatids of RibJ, the first riboflavin transporter family in protists. PLoS Negl Trop Dis 2017; 11:e0005513. [PMID: 28406895 PMCID: PMC5404878 DOI: 10.1371/journal.pntd.0005513] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/25/2017] [Accepted: 03/21/2017] [Indexed: 01/07/2023] Open
Abstract
Background Trypanosomatid parasites represent a major health issue affecting hundreds of million people worldwide, with clinical treatments that are partially effective and/or very toxic. They are responsible for serious human and plant diseases including Trypanosoma cruzi (Chagas disease), Trypanosoma brucei (Sleeping sickness), Leishmania spp. (Leishmaniasis), and Phytomonas spp. (phytoparasites). Both, animals and trypanosomatids lack the biosynthetic riboflavin (vitamin B2) pathway, the vital precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) cofactors. While metazoans obtain riboflavin from the diet through RFVT/SLC52 transporters, the riboflavin transport mechanisms in trypanosomatids still remain unknown. Methodology/Principal findings Here, we show that riboflavin is imported with high affinity in Trypanosoma cruzi, Trypanosoma brucei, Leishmania (Leishmania) mexicana, Crithidia fasciculata and Phytomonas Jma using radiolabeled riboflavin transport assays. The vitamin is incorporated through a saturable carrier-mediated process. Effective competitive uptake occurs with riboflavin analogs roseoflavin, lumiflavin and lumichrome, and co-factor derivatives FMN and FAD. Moreover, important biological processes evaluated in T. cruzi (i.e. proliferation, metacyclogenesis and amastigote replication) are dependent on riboflavin availability. In addition, the riboflavin competitive analogs were found to interfere with parasite physiology on riboflavin-dependent processes. By means of bioinformatics analyses we identified a novel family of riboflavin transporters (RibJ) in trypanosomatids. Two RibJ members, TcRibJ and TbRibJ from T. cruzi and T. brucei respectively, were functionally characterized using homologous and/or heterologous expression systems. Conclusions/Significance The RibJ family represents the first riboflavin transporters found in protists and the third eukaryotic family known to date. The essentiality of riboflavin for trypanosomatids, and the structural/biochemical differences that RFVT/SLC52 and RibJ present, make the riboflavin transporter -and its downstream metabolism- a potential trypanocidal drug target. In this work, we show that riboflavin plays a key role in the trypanosomatid life cycles and describe a novel family of riboflavin transporters (RibJ) with uptake function. Despite the vital importance of riboflavin for all living cells, RibJ are the first transporters described in protists. We functionally characterized the T. cruzi and T. brucei RibJ members and the effect of riboflavin analogs on parasite physiology. The structural and biochemical differences presented between human transporters and RibJ members make riboflavin transport and downstream metabolism, attractive and potential trypanosomatid targets.
Collapse
|
9
|
Mwololo SW, Mutiso JM, Macharia JC, Bourdichon AJ, Gicheru MM. In vitro activity and in vivo efficacy of a combination therapy of diminazene and chloroquine against murine visceral leishmaniasis. J Biomed Res 2015; 29:214-23. [PMID: 26060445 PMCID: PMC4449489 DOI: 10.7555/jbr.29.20140072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/17/2014] [Accepted: 12/12/2014] [Indexed: 11/24/2022] Open
Abstract
The present study evaluated the in vitro activity and in vivo efficacy of diminazene combined with chloroquine as a potential drug against Leishmania donovani. Amphotericin B was used as a positive control drug. In vitro activity involved incubation of various drug concentrations with promastigotes or vero cells in culture before determination of parasite growth inhibition or cell death while in vivo evaluations involved infection of various mice groups with virulent L. donovani parasites and treatment with test drug compounds following disease establishment. Weight changes in experimental mice were also evaluated before infection and throughout the experiment. The results indicated that the diminazene–chloroquine combination was at least nine times more efficacious than individual drugs in killing promastigotes in culture. The diminazene–chloroquine combination was safer (Ld50 = 0.03±0.04) than Amphotericin B (Ld50 = 0.02±0.01). Body weight in infected mice increased significantly (P = 0.0007) from day 7 to day 37 following infection (P = 0.026). However, body weight remained comparable in all mice groups during treatment (P = 0.16). The diminazene–chloroquine combination significantly reduced splenic parasite numbers as compared to individual drug therapies (P = 0.0001) although Amphotericin B was still more efficacious than any other treatment (P = 0.0001). Amongst the test compounds, the diminazene–chloroquine combination showed the lowest level of IgG antibody responses with results indicating significant negative correlation between antileishmanial antibody responses and protection against disease. These findings demonstrate the positive advantage and the potential use of a combined therapy of diminazene–chloroquine over the constituent drugs. Further evaluation is recommended to determine the most efficacious combination ratio of the two compounds.
Collapse
Affiliation(s)
- Samuel W Mwololo
- Department of Zoological Sciences, Kenyatta University, P. O. Box 43844-00100, Nairobi, Kenya. ; Department of Tropical and Infectious Diseases, Institute of Primate Research, P. O. Box 24481-00502, Karen, Nairobi, Kenya
| | - Joshua M Mutiso
- Department of Zoological Sciences, Kenyatta University, P. O. Box 43844-00100, Nairobi, Kenya
| | - John C Macharia
- Department of Tropical and Infectious Diseases, Institute of Primate Research, P. O. Box 24481-00502, Karen, Nairobi, Kenya
| | - Alain J Bourdichon
- BPM Bulk Medicine and Pharmaceuticals Production, TROPMED GMbH, Neuhofer Welche 48, D-19370 Parchtm, Germany
| | - Michael M Gicheru
- Department of Zoological Sciences, Kenyatta University, P. O. Box 43844-00100, Nairobi, Kenya
| |
Collapse
|
10
|
Ghosh AK, Sardar AH, Mandal A, Saini S, Abhishek K, Kumar A, Purkait B, Singh R, Das S, Mukhopadhyay R, Roy S, Das P. Metabolic reconfiguration of the central glucose metabolism: a crucial strategy of Leishmania donovani for its survival during oxidative stress. FASEB J 2015; 29:2081-98. [PMID: 25690656 DOI: 10.1096/fj.14-258624] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/09/2015] [Indexed: 12/15/2022]
Abstract
Understanding the mechanism that allows the intracellular protozoan parasite Leishmania donovani (Ld) to respond to reactive oxygen species (ROS) is of increasing therapeutic importance because of the continuing resistance toward antileishmanial drugs and for determining the illusive survival strategy of these parasites. A shift in primary carbon metabolism is the fastest response to oxidative stress. A (14)CO2 evolution study, expression of glucose transporters together with consumption assays, indicated a shift in metabolic flux of the parasites from glycolysis toward pentose phosphate pathway (PPP) when exposed to different oxidants in vitro/ex vivo. Changes in gene expression, protein levels, and enzyme activities all pointed to a metabolic reconfiguration of the central glucose metabolism in response to oxidants. Generation of glucose-6-phosphate dehydrogenase (G6PDH) (∼5-fold) and transaldolase (TAL) (∼4.2-fold) overexpressing Ld cells reaffirmed that lethal doses of ROS were counterbalanced by effective manipulation of NADPH:NADP(+) ratio and stringent maintenance of reduced thiol content. The extent of protein carbonylation and accumulation of lipid peroxidized products were also found to be less in overexpressed cell lines. Interestingly, the LD50 of sodium antimony gluconate (SAG), amphotericin-B (AmB), and miltefosine were significantly high toward overexpressing parasites. Consequently, this study illustrates that Ld strategizes a metabolic reconfiguration for replenishment of NADPH pool to encounter oxidative challenges.
Collapse
Affiliation(s)
- Ayan K Ghosh
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Abul H Sardar
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Abhishek Mandal
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Savita Saini
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Kumar Abhishek
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Ashish Kumar
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Bidyut Purkait
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Ruby Singh
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sushmita Das
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Rupkatha Mukhopadhyay
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Syamal Roy
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Pradeep Das
- *Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (Indian Council of Medical Research), Agamkuan, Patna, Bihar, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park, Hajipur, Vaishali, Bihar, India; Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, India; and Division of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| |
Collapse
|
11
|
Sousa MC, Varandas R, Santos RC, Santos-Rosa M, Alves V, Salvador JAR. Antileishmanial activity of semisynthetic lupane triterpenoids betulin and betulinic acid derivatives: synergistic effects with miltefosine. PLoS One 2014; 9:e89939. [PMID: 24643019 PMCID: PMC3958361 DOI: 10.1371/journal.pone.0089939] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/23/2014] [Indexed: 11/21/2022] Open
Abstract
Leishmaniasis is a neglected tropical disease (NTDs), endemic in 88 countries, affecting more than 12 million people. The treatment consists in pentavalent antimony compounds, amphotericin B, pentamidine and miltefosine, among others. However, these current drugs are limited due to their toxicity, development of biological resistance, length of treatment and high cost. Thus, it is important to continue the search for new effective and less toxic treatments. The anti-Leishmania activity of sixteen semisynthetic lupane triterpenoids derivatives of betulin (BT01 to BT09) and betulinic acid (AB10 to AB16) were evaluated. Drug interactions between the active compounds and one current antileishmanial drug, miltefosine, were assessed using the fixed ratio isobologram method. In addition, effects on the cell cycle, apoptosis/necrosis events, morphology and DNA integrity were studied. The derivatives BT06 (3β-Hydroxy-(20R)-lupan-29-oxo-28-yl-1H-imidazole-1-carboxylate) and AB13 (28-(1H-imidazole-1-yl)-3,28-dioxo-lup-1,20(29)-dien-2-yl-1H-imidazole-1-carboxylate) were found to be the most active, with IC50 values of 50.8 µM and 25.8 µM, respectively. Interactions between these two compounds and miltefosine were classified as synergistic, with the most effective association being between AB13 and miltefosine, where decreases of IC50 values to 6 µM were observed, similar to the miltefosine activity alone. AB13 induced significant morphological changes, while both derivatives produced anti-proliferative activity through cell cycle arrest at the G0/G1 phase. Neither of these derivatives induced significant apoptosis/necrosis, as indicated by phosphatidylserine externalization and DNA fragmentation assays. In addition, neither of the derivatives induced death in macrophage cell lines. Thus, they do not present any potential risk of toxicity for the host cells. This study has identified the betulin derivative BT06 and the betulinic acid derivative AB13 as promising molecules in the development of new alternative therapies for leishmaniasis, including those involving combined-therapy with miltefosine.
Collapse
Affiliation(s)
- Maria C. Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
- Centre of Pharmaceutical Studies, Faculty of Pharmacy of University of Coimbra (CEF/FFUC), Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
- * E-mail: (MCS); (JARS)
| | - Raquel Varandas
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Rita C. Santos
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Manuel Santos-Rosa
- Institute of Immunology, Faculty of Medicine, University of Coimbra, Pólo I, Rua Larga, Coimbra, Portugal
| | - Vera Alves
- Institute of Immunology, Faculty of Medicine, University of Coimbra, Pólo I, Rua Larga, Coimbra, Portugal
| | - Jorge A. R. Salvador
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
- CNC- Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- * E-mail: (MCS); (JARS)
| |
Collapse
|
12
|
Unciti-Broceta JD, Maceira J, Morales S, García-Pérez A, Muñóz-Torres ME, Garcia-Salcedo JA. Nicotinamide inhibits the lysosomal cathepsin b-like protease and kills African trypanosomes. J Biol Chem 2013; 288:10548-57. [PMID: 23443665 DOI: 10.1074/jbc.m112.449207] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide, a soluble compound of the vitamin B3 group, has antimicrobial activity against several microorganisms ranging from viruses to parasite protozoans. However, the mode of action of this antimicrobial activity is unknown. Here, we investigate the trypanocidal activity of nicotinamide on Trypanosoma brucei, the causative agent of African trypanosomiasis. Incubation of trypanosomes with nicotinamide causes deleterious defects in endocytic traffic, disruption of the lysosome, failure of cytokinesis, and, ultimately, cell death. At the same concentrations there was no effect on a cultured mammalian cell line. The effects on endocytosis and vesicle traffic were visible within 3 h and can be attributed to inhibition of lysosomal cathepsin b-like protease activity. The inhibitory effect of nicotinamide was confirmed by a direct activity assay of recombinant cathepsin b-like protein. Taken together, these data demonstrate that inhibition of the lysosomal protease cathepsin b-like blocks endocytosis, causing cell death. In addition, these results demonstrate for the first time the inhibitory effect of nicotinamide on a protease.
Collapse
Affiliation(s)
- Juan D Unciti-Broceta
- Unidad de Enfermedades Infecciosas, Instituto de Investigación Biosanitaria de Granada (IBIG), Av. Dr. Olóriz 16, 18012 Granada, Granada, Spain
| | | | | | | | | | | |
Collapse
|
13
|
Soares MB, Silva CV, Bastos TM, Guimarães ET, Figueira CP, Smirlis D, Azevedo WF. Anti-Trypanosoma cruzi activity of nicotinamide. Acta Trop 2012; 122:224-9. [PMID: 22281243 DOI: 10.1016/j.actatropica.2012.01.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 12/13/2011] [Accepted: 01/01/2012] [Indexed: 11/25/2022]
Abstract
Inhibition of Trypanosoma brucei and Leishmania spp. sirtuins has shown promising antiparasitic activity, indicating that these enzymes may be used as targets for drug discovery against trypanosomatid infections. In the present work we carried out a virtual screening focused on the C pocket of Sir2 from Trypanosoma cruzi. Using this approach, the best ligand found was nicotinamide. In vitro tests confirmed the anti-T. cruzi activity of nicotinamide on epimastigote and trypomastigote forms. Moreover, treatment of T. cruzi-infected macrophages with nicotinamide caused a significant reduction in the number of amastigotes. In addition, alterations in the mitochondria and an increase in the vacuolization in the cytoplasm were observed in epimastigotes treated with nicotinamide. Analysis of the complex of Sir2 and nicotinamide revealed the details of the possible ligand-target interaction. Our data reveal a potential use of TcSir2 as a target for anti-T. cruzi drug discovery.
Collapse
|
14
|
Michels PAM, Avilán L. The NAD+ metabolism of Leishmania, notably the enzyme nicotinamidase involved in NAD+ salvage, offers prospects for development of anti-parasite chemotherapy. Mol Microbiol 2011; 82:4-8. [PMID: 21854468 DOI: 10.1111/j.1365-2958.2011.07810.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
NAD+ plays multiple, essential roles in the cell. As a cofactor in many redox reactions it is key in the cellular energy metabolism and as a substrate it participates in many reactions leading to a variety of covalent modifications of enzymes with major roles in regulation of expression and metabolism. Cells may have the ability to produce this metabolite either via alternative de novo synthesis pathways and/or by different salvage pathways. In this issue of Molecular Microbiology, Gazanion et al. (2011) demonstrate that Leishmania species can only rely on the salvage of NAD+ building blocks. One of the enzymes involved, nicotinamidase, is absent from human cells. The enzyme is important for growth of Leishmania infantum and essential for establishing an infection. The crystal structure of the parasite protein has been solved and shows prospects for design of inhibitors to be used as leads for development of new drugs. Indeed, NAD+ metabolism is currently being considered as a promising drug target in various diseases and the vulnerability of Leishmania for interference of this metabolism has been proved in previous work by the same group, by showing that administration of NAD+ precursors has detrimental effect on the pathogenic, amastigote stage of this parasite.
Collapse
Affiliation(s)
- Paul A M Michels
- Research Unit for Tropical Diseases, de Duve Institute and Laboratory of Biochemistry, Université catholique de Louvain, Avenue Hippocrate 74, Postal Box B1.74.01, B-1200 Brussels, Belgium.
| | | |
Collapse
|
15
|
Gazanion E, Garcia D, Silvestre R, Gérard C, Guichou JF, Labesse G, Seveno M, Cordeiro-Da-Silva A, Ouaissi A, Sereno D, Vergnes B. The Leishmania nicotinamidase is essential for NAD+ production and parasite proliferation. Mol Microbiol 2011; 82:21-38. [PMID: 21819459 DOI: 10.1111/j.1365-2958.2011.07799.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
NAD+ is a central cofactor that plays important roles in cellular metabolism and energy production in all living cells. Genomics-based reconstruction of NAD+ metabolism revealed that Leishmania protozoan parasites are NAD+ auxotrophs. Consequently, these parasites require assimilating NAD+ precursors (nicotinamide, nicotinic acid, nicotinamide riboside) from their host environment to synthesize NAD+ by a salvage pathway. Nicotinamidase is a key enzyme of this salvage pathway that catalyses conversion of nicotinamide (NAm) to nicotinic acid (Na), and that is absent in higher eukaryotes. We present here the biochemical and functional characterizations of the Leishmania infantum nicotinamidase (LiPNC1). Generation of Lipnc1 null mutants leads to a decrease in NAD+ content, associated with a metabolic shutdown-like phenotype with an extensive lag phase of growth. Both phenotypes could be rescued by an add-back construct or by addition of exogenous Na. In addition, Lipnc1 null mutants were unable to establish a sustained infection in a murine experimental model. Altogether, these results illustrate that NAD+ homeostasis is a fundamental component of Leishmania biology and virulence, and that NAm constitutes its main NAD+ source in the mammalian host. The crystal structure of LiPNC1 we solved allows now the design of rational inhibitors against this new promising therapeutic target.
Collapse
Affiliation(s)
- E Gazanion
- MIVEGEC (UM1-CNRS 5290-IRD 224), Institut de Recherche pour le Développement (IRD), BP 64501, 34394 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|