1
|
Chaúque BJM, da Silva TCB, Rott EB, Rott FB, Leite APMC, Benitez GB, Neuana NF, Goldim JR, Rott MB, Zanette RA. Effectiveness of phytoproducts against pathogenic free-living amoebae - A scoping and critical review paving the way toward plant-based pharmaceuticals. Fitoterapia 2025; 182:106404. [PMID: 39922391 DOI: 10.1016/j.fitote.2025.106404] [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: 08/17/2024] [Revised: 01/10/2025] [Accepted: 02/01/2025] [Indexed: 02/10/2025]
Abstract
Infections caused by free-living amoebae (FLA) have increased worldwide and are expected to worsen. The lack of drugs that are effective (especially against cysts), affordable, and safe to treat these infections exacerbates the concern. Plants present a promising source of bioactive compounds for developing effective drugs; however, the scientific literature on this topic has yet to be adequately synthesized. This work provides a critical scoping review summarizing the amoebicidal performance of plant-derived products and their potential for developing effective drugs to treat FLA infections. Out of 5889 articles retrieved from multiple databases, 119 articles were selected, from which data on 180 plant species belonging to 127 genera and 62 families were extracted. The extracts, essential oils, and compounds from these plants exhibited a diverse range of potency against cysts and trophozoites. Among the compounds studied, periglaucine A, kolavenic acid, and (+)-elatol are promising cysticidal drug candidates due to their high potency, as well as their known low toxicity to non-target cells. Tovophillin A, gartinin, 8-deoxygartinin, garcinone E, 9-hydroxycalabaxanthone, γ-mangostin, and borneol also exhibit high cysticidal potency, but their selectivity profile is unknown. Resveratrol, rosmarinic acid, β-amyrin, and vanillic acid stand out for their high potency against trophozoites and low toxicity to mammalian cells. Another group of compounds with similarly high trophocidal potency includes (-)-epicatechin, (-)-epigallocatechin, apigenin, costunolide, demethoxycurcumin, kaempferol, methyl-β-orcinolcarboxylate, sakuraetin, (+)-elatol, debromolaurinterol, luteolin, (-)-rogiolol, cystomexicone B, epigallocatechin gallate, quercetin, and α-bisabolol. These compounds are priority candidates for further studies on in vivo efficacy, safety, pharmacokinetics, and pharmacodynamics.
Collapse
Affiliation(s)
- Beni Jequicene Mussengue Chaúque
- Postgraduate Program in Biological Sciences, Pharmacology and Therapeutics, UFRGS, Rio Grande do Sul, Brazil; Postdoctoral fellow at Master's Program in Clinical Research (MPPC) at the Hospital de Clínicas de Porto Alegre (HCPA) (CAPES Pilot Program), Rio Grande do Sul, Brazil; Center of Studies in Science and Technology (NECET), Biology Course, Universidade Rovuma, Niassa Branch, Lichinga, Mozambique.
| | - Thaisla Cristiane Borella da Silva
- Protozoology Laboratory, Microbiology Immunology and Parasitology Department, Basic Health Sciences Institute, Federal University of Rio Grande do Sul, Ramiro Barcelos Street, N 2600, 90035-002 Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Felipe Brittes Rott
- Faculty of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil
| | | | - Guilherme Brittes Benitez
- Industrial and Systems Engineering Graduate Program, Polytechnic School, Pontifical Catholic University of Parana (PUCPR), Brazil
| | - Neuana Fernando Neuana
- Center of Studies in Science and Technology (NECET), Biology Course, Universidade Rovuma, Niassa Branch, Lichinga, Mozambique; Department of Mechanical and Materials Engineering, Federal University of Santa Catarina, Florianópolis, SC 88040900, Brazil
| | - José Roberto Goldim
- Postdoctoral fellow at Master's Program in Clinical Research (MPPC) at the Hospital de Clínicas de Porto Alegre (HCPA) (CAPES Pilot Program), Rio Grande do Sul, Brazil.
| | - Marilise Brittes Rott
- Protozoology Laboratory, Microbiology Immunology and Parasitology Department, Basic Health Sciences Institute, Federal University of Rio Grande do Sul, Ramiro Barcelos Street, N 2600, 90035-002 Porto Alegre, Rio Grande do Sul, Brazil.
| | - Régis Adriel Zanette
- Postgraduate Program in Biological Sciences, Pharmacology and Therapeutics, UFRGS, Rio Grande do Sul, Brazil.
| |
Collapse
|
2
|
Jiang Y, He P, Sheng K, Peng Y, Wu H, Qian S, Ji W, Guo X, Shan X. The protective roles of eugenol on type 1 diabetes mellitus through NRF2-mediated oxidative stress pathway. eLife 2025; 13:RP96600. [PMID: 39792010 PMCID: PMC11723580 DOI: 10.7554/elife.96600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
Type 1 diabetes mellitus (T1DM), known as insulin-dependent diabetes mellitus, is characterized by persistent hyperglycemia resulting from damage to the pancreatic β cells and an absolute deficiency of insulin, leading to multi-organ involvement and a poor prognosis. The progression of T1DM is significantly influenced by oxidative stress and apoptosis. The natural compound eugenol (EUG) possesses anti-inflammatory, anti-oxidant, and anti-apoptotic properties. However, the potential effects of EUG on T1DM had not been investigated. In this study, we established the streptozotocin (STZ)-induced T1DM mouse model in vivo and STZ-induced pancreatic β cell MIN6 cell model in vitro to investigate the protective effects of EUG on T1DM, and tried to elucidate its potential mechanism. Our findings demonstrated that the intervention of EUG could effectively induce the activation of nuclear factor E2-related factor 2 (NRF2), leading to an up-regulation in the expressions of downstream proteins NQO1 and HMOX1, which are regulated by NRF2. Moreover, this intervention exhibited a significant amelioration in pancreatic β cell damage associated with T1DM, accompanied by an elevation in insulin secretion and a reduction in the expression levels of apoptosis and oxidative stress-related markers. Furthermore, ML385, an NRF2 inhibitor, reversed these effects of EUG. The present study suggested that EUG exerted protective effects on pancreatic β cells in T1DM by attenuating apoptosis and oxidative stress through the activation of the NRF2 signaling pathway. Consequently, EUG holds great promise as a potential therapeutic candidate for T1DM.
Collapse
Affiliation(s)
- Yalan Jiang
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Pingping He
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ke Sheng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yongmiao Peng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Huilan Wu
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Songwei Qian
- Department of Genaral Surgery, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s HospitalQuzhouChina
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Weiping Ji
- Department of Genaral Surgery, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s HospitalQuzhouChina
- Department of General Surgery, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaoling Guo
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaoou Shan
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
3
|
Berhe H, Kumar Cinthakunta Sridhar M, Zerihun M, Qvit N. The Potential Use of Peptides in the Fight against Chagas Disease and Leishmaniasis. Pharmaceutics 2024; 16:227. [PMID: 38399281 PMCID: PMC10892537 DOI: 10.3390/pharmaceutics16020227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Chagas disease and leishmaniasis are both neglected tropical diseases that affect millions of people around the world. Leishmaniasis is currently the second most widespread vector-borne parasitic disease after malaria. The World Health Organization records approximately 0.7-1 million newly diagnosed leishmaniasis cases each year, resulting in approximately 20,000-30,000 deaths. Also, 25 million people worldwide are at risk of Chagas disease and an estimated 6 million people are infected with Trypanosoma cruzi. Pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine are currently used to treat leishmaniasis. Also, nifurtimox and benznidazole are two drugs currently used to treat Chagas disease. These drugs are associated with toxicity problems such as nephrotoxicity and cardiotoxicity, in addition to resistance problems. As a result, the discovery of novel therapeutic agents has emerged as a top priority and a promising alternative. Overall, there is a need for new and effective treatments for Chagas disease and leishmaniasis, as the current drugs have significant limitations. Peptide-based drugs are attractive due to their high selectiveness, effectiveness, low toxicity, and ease of production. This paper reviews the potential use of peptides in the treatment of Chagas disease and leishmaniasis. Several studies have demonstrated that peptides are effective against Chagas disease and leishmaniasis, suggesting their use in drug therapy for these diseases. Overall, peptides have the potential to be effective therapeutic agents against Chagas disease and leishmaniasis, but more research is needed to fully investigate their potential.
Collapse
Affiliation(s)
| | | | | | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel; (H.B.); (M.K.C.S.); (M.Z.)
| |
Collapse
|
4
|
Ahmad AA, Maurice MN, Monib MESM, Soliman M, Al-Thagfan SS, Huseein EAM. Eugenol Essential Oil and Nanoemulsion as Antihydatic Agents with Antifibrotic and Immunomodulatory Effects in Cystic Echinococcosis. Trop Med Infect Dis 2023; 8:tropicalmed8050253. [PMID: 37235301 DOI: 10.3390/tropicalmed8050253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Conventional scolicidal agents are still unsatisfactory in combating hydatid disease due to their low efficacy and increased drug side effects. Therefore, novel scolicides are required. This study aimed to evaluate the antihydatic and immunomodulatory effects of eugenol essential oil (Eug) and its nanoemulsion (Eug-NE) in cystic echinococcosis (CE). Eug and Eug-NE were administered orally to CE-infected rats and compared to albendazole (ABZ). Hydatid cyst development was assessed based on organ weight and hypertrophy indicators of the infected organs, along with a histopathological and histochemical evaluation of collagen content. The immunomodulatory effects of treatment on CE were evaluated by serum cytokine levels measurement of interferon-γ (IFN-γ) and interleukin (IL)-4 and immunohistochemical (IHC) analysis of signal transducer and activator of transcription 4 (STAT4) and GATA-binding protein 3 (GATA3) markers. Eug-NE was the most effective in reducing the cyst weights, organ weights, and hypertrophy indicators and improving histopathological lesions with reduced collagen content. Eug and Eug-NE significantly increased the IFN-γ levels and decreased the IL-4 levels, while IHC analysis demonstrated a significant reduction in STAT4 and GATA3 expression in all treated groups. Eug and Eug-NE demonstrated antihydatic and preventative effects, with a substantial decrease in liver fibrosis compared to that of ABZ. Besides their promising immunomodulatory effects, their good treatment response suggests their use as alternatives or complementary scolicidal agents in hydatid cyst treatment.
Collapse
Affiliation(s)
| | - Maria Naged Maurice
- Department of Medical Parasitology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | | | - Mahmoud Soliman
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Sultan S Al-Thagfan
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Al Madinah Al Munawarah 30001, Saudi Arabia
| | | |
Collapse
|
5
|
Liu Y. Integrative network pharmacology and in silico analyses identify the anti-omicron SARS-CoV-2 potential of eugenol. Heliyon 2023; 9:e13853. [PMID: 36845041 PMCID: PMC9937729 DOI: 10.1016/j.heliyon.2023.e13853] [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: 04/03/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Eugenol as a natural product is the source of isoniazid, and purified eugenol is extensively used in the cosmetics industry and the productive processes of edible spices. Accumulating evidence suggested that eugenol exerted potent anti-microorganism and anti-inflammation effects. Application of eugenol effectively reduced the risk of atherosclerosis, arterial embolism, and Type 2 diabetes. A previous study confirmed that treatment with eugenol attenuated lung inflammation and improved heart functions in SARS-CoV-2 spike S1-intoxicated mice. In addition to the study, based on a series of public datasets, computational analyses were conducted to characterize the acting targets of eugenol and the functional roles of these targets in COVID-19. The binding capacities of eugenol to conservative sites of SARS-CoV-2 like RNA-dependent RNA polymerase (RdRp) and mutable site as spike (S) protein, were calculated by using molecular docking following the molecular dynamics simulation with RMSD, RMSF, and MM-GBSA methods. The results of network pharmacology indicated that six targets, including PLAT, HMOX1, NUP88, CTSL, ITGB1 andTMPRSS2 were eugenol-SARS-CoV-2 interacting proteins. The omics results of in-silico study further implicated that eugenol increased the expression of SCARB1, HMOX1 and GDF15, especially HMOX1, which were confirmed the potential interacting targets between eugenol and SARS-CoV-2 antigens. Enrichment analyses indicated that eugenol exerted extensive biological effects such as regulating immune infiltration of macrophage, lipid localization, monooxyenase activity, iron ion binding and PPAR signaling. The results of the integrated analysis of eugenol targets and immunotranscription profile of COVID-19 cases shows that eugenol also plays an important role in strengthen of immunologic functions and regulating cytokine signaling. As a complement to the integrated analysis, the results of molecular docking indicated the potential binding interactions between eugenol and four proteins relating to cytokine production/release and the function of T type lymphocytes, including human TLR-4, TCR, NF-κB, JNK and AP-1. Furthermore, results of molecular docking and molecular dynamics (100ns) simulations implicated that stimulated modification of eugenol to the SARS-CoV-2 Omicron Spike-ACE2 complex, especially for human ACE2, and the molecular interaction of eugenol to SARS-CoV-2 RdRp, were no less favorable than two positive controls, molnupiravir and nilotinib. Dynamics (200ns) simulations indicated that the binding capacities and stabilities of eugenol to finger subdomain of RdRp is no less than molnupiravir. However, the simulated binding capacity of eugenol to SARS-CoV-2 wild type RBD and Omicron mutant RBD were less than nilotinib. Eugenol was predicted to have more favor LD50 value and lower cytotoxicity than two positive controls, and eugenol can pass through the blood-brain barrier (BBB). In a brief, eugenol is helpful for attenuating systemic inflammation induced by SARS-CoV-2 infection, due to the direct interaction of eugenol to SARS-CoV-2 proteins and extensive bio-manipulation of pro-inflammatory factors. This study carefully suggests eugenol is a candidate compound of developing drugs and supplement agents against SARS-CoV-2 and its Omicron variants.
Collapse
Affiliation(s)
- Yang Liu
- Graduated Student of Harbin Medical University, Cardiology. Baojian Road105, Nangang Distinct, Harbin, Heilongjiang, China
| |
Collapse
|
6
|
Jayaraman A, Srinivasan S, Uppuluri KB, Kar Mahapatra S. Unwinding the mechanism of macrophage repolarization potential of Oceanimonas sp. BPMS22-derived protein protease inhibitor through Toll-like receptor 4 against experimental visceral leishmaniasis. Front Cell Infect Microbiol 2023; 13:1120888. [PMID: 37033485 PMCID: PMC10073655 DOI: 10.3389/fcimb.2023.1120888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
The Oceanimonas sp. BPMS22-derived protein protease inhibitor (PPI) has been proven to shift macrophages towards an inflammatory state and reduce Leishmania donovani infection in vitro and in vivo. The current study explored and validated the mechanistic aspects of the PPI and Toll-like receptor (TLR) interaction. The PPI exhibited the upregulation of TLR2, TLR4, and TLR6 during treatment which was proven to orchestrate parasite clearance effectively. An in silico study confirmed the high interaction with TLR4 and PPI. Immune blotting confirmed the significant upregulation of TLR4 in macrophages irrespective of L. donovani infection. Pharmacological inhibition and immune blot study confirmed the involvement of the PPI in TLR4-mediated phosphorylation of p38 MAPK and dephosphorylation of ERK1/2, repolarizing to pro-inflammatory macrophage state against experimental visceral leishmaniasis. In addition, in TLR4 knockdown condition, PPI treatment failed to diminish M2 phenotypical markers (CD68, Fizz1, Ym1, CD206, and MSR-2) and anti-inflammatory cytokines (IL-4, IL-10, and TGF-β). Simultaneously, the PPI failed to upregulate the M1 phenotypical markers and pro-inflammatory cytokines (IL-1β, IL-6, IL-12, and IFN-γ) (p < 0.001) during the TLR4 knockdown condition. In the absence of TLR4, the PPI also failed to reduce the parasite load and T-cell proliferation and impaired the delayed-type hypersensitivity response. The absence of pro-inflammatory cytokines was observed during a co-culture study with PPI-treated macrophages (in the TLR4 knockdown condition) with day 10 T-cell obtained from L. donovani-infected mice. This study supports the immunotherapeutic potential of the PPI as it interacted with TLR4 and promoted macrophage repolarization (M2-M1) to restrict the L. donovani parasite burden and helps in the mounting immune response against experimental visceral leishmaniasis.
Collapse
Affiliation(s)
- Adithyan Jayaraman
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sujatha Srinivasan
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Kiran Babu Uppuluri
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
- *Correspondence: Santanu Kar Mahapatra, ; Kiran Babu Uppuluri,
| | - Santanu Kar Mahapatra
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore, India
- *Correspondence: Santanu Kar Mahapatra, ; Kiran Babu Uppuluri,
| |
Collapse
|
7
|
Mazire PH, Saha B, Roy A. Immunotherapy for visceral leishmaniasis: A trapeze of balancing counteractive forces. Int Immunopharmacol 2022; 110:108969. [PMID: 35738089 DOI: 10.1016/j.intimp.2022.108969] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
The protozoan parasite Leishmania donovani, residing and replicating within the cells of the monocyte-macrophage (mono-mac) lineage, causes visceral leishmaniasis (VL) in humans. While, Leishmania infantum, is the main causative agent for zoonotic VL, where dogs are the main reservoirs of the disease. The chemotherapy is a serious problem because of restricted repertoire of drugs, drug-resistant parasites, drug-toxicity and the requirement for parenteral administration, which is a problem in resource-starved countries. Moreover, immunocompromised individuals, particularly HIV-1 infected are at higher risk of VL due to impairment in T-helper cell and regulatory cell responses. Furthermore, HIV-VL co-infected patients report poor response to conventional chemotherapy. Recent efforts are therefore directed towards devising both prophylactic and therapeutic immunomodulation. As far as prophylaxis is concerned, although canine vaccines for the disease caused by Leishmania infantum or Leishmania chagasi are available, no vaccine is available for use in humans till date. Therefore, anti-leishmanial immunotherapy triggering or manipulating the host's immune response is gaining momentum during the last two decades. Immunomodulators comprised of small molecules, anti-leishmanial peptides, complex ligands for host receptors, cytokines or their agonists and antibodies have been given trials both in experimental models and in humans. However, the success of immunotherapy in humans remains a far-off target. We, therefore, propose that devising a successful immunotherapy is an act of balancing enhanced beneficial Leishmania-specific responses and deleterious immune activation/hyperinflammation just as the swings in a trapeze.
Collapse
Affiliation(s)
- Priyanka H Mazire
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind Road, Pune 411007, India
| | - Amit Roy
- Department of Biotechnology, Savitribai Phule Pune University, Ganeshkhind Road, Pune 411007, India.
| |
Collapse
|
8
|
Flau-A, a naphthoquinone derivative, is a promising therapeutic candidate against visceral leishmaniasis: A preliminary study. Exp Parasitol 2021; 233:108205. [PMID: 34968460 DOI: 10.1016/j.exppara.2021.108205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 11/30/2021] [Accepted: 12/25/2021] [Indexed: 11/21/2022]
Abstract
Visceral leishmaniasis (VL) is a neglected tropical disease found in tropical and subtropical regions in the world. The therapeutics used for the treatment against disease presents problems, mainly related to drug toxicity, route of administration, high cost and/or by emergence of resistant strains. In this context, the search for alternative antileishmanial candidates is desirable. Recently, a naphthoquinone derivative namely 2-(2,3,4-tri-O-acetyl-6-deoxy-β-L-galactopyranosyloxy)-1,4-naphthoquinone or Flau-A showed an effective in vitro biological action against Leishmania infantum. In the present study, the efficacy of this naphthoquinone derivative was evaluated in an in vivo infection model. BALB/c mice (n = 12 per group) were infected and later received saline or were treated with empty micelles (B/Mic), free Flau-A or it incorporated in Poloxamer 407-based micelles (Flau-A/Mic). The products were administered subcutaneously in the infected animals, which were then euthanized one (n = 6 per group) and 15 (n = 6 per group) days post-therapy, when immunological and parasitological evaluations were performed. Results showed that animals treated with Flau-A or Flau-A/Mic produced significantly higher levels of antileishmanial IFN-γ, IL-12, TNF-α, GM-CSF, nitrite and IgG2a isotype antibody, when compared to data found in the control (saline and B/Mic) groups; which showed significantly higher levels of parasite-specific IL-4, IL-10 and IgG1 antibody. In addition, animals receiving free Flau-A or Flau-A/Mic presented also significant reductions in the parasite load in their spleens, livers, bone marrows and draining lymph nodes, when compared to the controls. A low hepatic and renal toxicity was also found. Overall, Flau-A/Mic showed better immunological and parasitological results, when compared to the use of free molecule. In conclusion, preliminary data suggest that this composition could be considered in future studies as promising therapeutic candidate against VL.
Collapse
|
9
|
Kar A, Jayaraman A, Kumar A, Kar Mahapatra S. Dynamicity in Host Metabolic Adaptation Is Influenced by the Synergistic Effect of Eugenol Oleate and Amphotericin B During Leishmania donovani Infection In Vitro. Front Cell Infect Microbiol 2021; 11:709316. [PMID: 34414131 PMCID: PMC8369346 DOI: 10.3389/fcimb.2021.709316] [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: 05/13/2021] [Accepted: 07/22/2021] [Indexed: 11/28/2022] Open
Abstract
Immune metabolic adaptation in macrophages by intracellular parasites is recognized to play a crucial role during Leishmania infection. However, there is little accessible information about changes in a metabolic switch in L. donovani infected macrophages. In previous studies, we have reported on the anti-leishmanial synergic effect of eugenol oleate with amphotericin B. In the present study, we demonstrated that glycolytic enzymes were highly expressed in infected macrophages during combinatorial treatment of eugenol oleate (2.5 µM) and amphotericin B (0.3125 µM). Additionally, we found that the biphasic role in arachidonic acid metabolite, PGE2, and LTB4, is released during this treatment. In vitro data showed that COX-2 mediated PGE2 synthesis increased significantly (p<0.01) in infected macrophages. Not only was the level of prostaglandin synthesis decreased 4.38 fold in infected macrophages after treatment with eugenol oleate with amphotericin B. The mRNA expression of PTGES, MPGES, and PTGER4 were also moderately expressed in infected macrophages, and found to be decreased in combinatorial treatment. In addition, NOS2 expression was activated by the phosphorylation of p38MAPK when combination-treated macrophages were promoted to kill intracellular parasites. The findings of the present study indicate that the synergism between eugenol oleate and amphotericin B could play an important role in immune metabolism adaptation with a concomitant increase in host immune response against the intracellular pathogen, L. donovani.
Collapse
Affiliation(s)
- Amrita Kar
- Department of Biotechnology, School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed to be University, Thanjavur, India
| | - Adithyan Jayaraman
- Department of Biotechnology, School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed to be University, Thanjavur, India
| | - Avanthika Kumar
- Department of Biotechnology, School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed to be University, Thanjavur, India
| | - Santanu Kar Mahapatra
- Department of Biotechnology, School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed to be University, Thanjavur, India.,Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore, India
| |
Collapse
|
10
|
Corydalis saxicola Bunting total alkaloids attenuate paclitaxel-induced peripheral neuropathy through PKCε/p38 MAPK/TRPV1 signaling pathway. Chin Med 2021; 16:58. [PMID: 34281577 PMCID: PMC8287815 DOI: 10.1186/s13020-021-00468-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/11/2021] [Indexed: 11/18/2022] Open
Abstract
Background Corydalis saxicola Bunting, affiliated with the Papaveraceae Juss., has been proven to work well in anti-inflammation, hemostasis, and analgesia. This study was designed to observe the effect and potential mechanism of Corydalis saxicola Bunting total alkaloids (CSBTA) on paclitaxel-induced peripheral neuropathy (PIPN). Materials and methods Rats were injected 2 mg/kg paclitaxel 4 times and administrated with 30 or 120 mg/kg CSBTA. Mechanical and thermal allodynia and hyperalgesia were tested. After 40 days, serum was collected to detect PGE2, TNF-α, and IL-1β by ELISA. The L4-L6 segment spinal cord, DRG, and plantar skin were harvested, and Western-blot or RT-qPCR analyzed protein and gene levels of pro-inflammatory cytokines, p38 MAPK, PKCε, and TRPV1. The PIPN cell model was established with paclitaxel (300 nM, 5 d) in primary DRG neurons. We examined the effect of CSBTA (25 μg/ml or 50 μg/ml) by measuring the mRNA levels in PGE2, TNF-α and CGRP, and the protein expression on the PKCε/p38 MAPK/TRPV1 signaling pathway in the PIPN cell model. Results The results showed that CSBTA effectively ameliorated allodynia and hyperalgesia, and regulated cytokines' contents (PGE2, TNF-α, and IL-1β) and neuropeptides (CGRP and SP) in different tissues in vivo. In addition, CSBTA significantly decreased cytokine gene levels of DRG neurons (PGE2, TNF-α, and CGRP) and the protein expressions of PKCε/p38 MAPK/TRPV1 signaling pathway in vivo and in vitro. Conclusion Therefore, CSBTA has a perspective therapeutic effect on the treatment of paclitaxel-induced peripheral neuropathy.
Collapse
|
11
|
Kar A, Jayaraman A, Charan Raja MR, Srinivasan S, Debnath J, Mahapatra SK. Synergic effect of eugenol oleate with amphotericin B augments anti-leishmanial immune response in experimental visceral leishmaniasis in vitro and in vivo. Int Immunopharmacol 2021; 91:107291. [PMID: 33360084 DOI: 10.1016/j.intimp.2020.107291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022]
Abstract
Present treatment regimen on visceral leishmaniasis has multiple limitations including severe side effects, toxicity, and resistance of Leishmania strains. Amphotericin B is a well-established pharmacologically approved drug; however, mainly toxicity is a foremost issue with that drug. Recently, our group identified eugenol oleate as an anti-leishmanial immunomodulatory compound. The important objectives of this present study was to evaluate the possible synergistic effect of eugenol oleate with amphotericin B to reduce the toxicity of this approved drug. Results obtained from this study signified that combination of eugenol oleate and amphotericin B showed indifferent combinatorial effect against promastigotes with xΣFIC 1.015, while, moderate synergistic activity with xΣFIC 0.456 against amastigotes. It was also notable that eugenol oleate (2.5 μM) with low concentrations of amphotericin B (0.3125 μM) showed 96.45% parasite reduction within L. donovani-infected murine macrophages. Furthermore, eugenol oleate and amphotericin B significantly (p < 0.01) enhanced the nitrite generation, and pro-inflammatory cytokines (IL-12, IFN-γ and TNF-α) in infected macrophages in vitro and in BALB/c mice in vivo. Eugenol oleate (10 mg/Kg b. wt.) with amphotericin B (1 mg/Kg b.wt.) significantly (p < 0.01) controlled the parasite burden in liver by 96.2% and in spleen by 93.12%. Hence, this study strongly suggested the synergic potential of eugenol oleate with low concentration of amphotericin B in experimental visceral leishmaniasis through anti-leishmanial immune response.
Collapse
MESH Headings
- Amphotericin B/pharmacology
- Animals
- Cells, Cultured
- Cytokines/metabolism
- Disease Models, Animal
- Drug Synergism
- Drug Therapy, Combination
- Female
- Host-Parasite Interactions
- Inflammation Mediators/metabolism
- Leishmania donovani/drug effects
- Leishmania donovani/immunology
- Leishmania donovani/pathogenicity
- Leishmaniasis, Visceral/drug therapy
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/metabolism
- Leishmaniasis, Visceral/parasitology
- Liver/drug effects
- Liver/immunology
- Liver/metabolism
- Liver/parasitology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/parasitology
- Mice, Inbred BALB C
- Nitrites/metabolism
- Parasite Load
- Spleen/drug effects
- Spleen/immunology
- Spleen/metabolism
- Spleen/parasitology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th1 Cells/parasitology
- Th1-Th2 Balance
- Th2 Cells/drug effects
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Th2 Cells/parasitology
- Trypanocidal Agents/pharmacology
- Mice
Collapse
Affiliation(s)
- Amrita Kar
- Department of Biotechnology, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India; Department of Chemistry, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Adithyan Jayaraman
- Department of Biotechnology, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India; Department of Chemistry, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Mamilla R Charan Raja
- Department of Biotechnology, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India; Department of Chemistry, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Sujatha Srinivasan
- Department of Biotechnology, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India; Department of Chemistry, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Joy Debnath
- Department of Biotechnology, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India; Department of Chemistry, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India.
| | - Santanu Kar Mahapatra
- Department of Biotechnology, Centre for Research in Infectious Diseases, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India; Department of Chemistry, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India.
| |
Collapse
|