1
|
Chen ZX, Qin YS, Shi BH, Gao BY, Tao RC, Yong XZ. Effects of Curcumin on Radiation/Chemotherapy-Induced Oral Mucositis: Combined Meta-Analysis, Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulation. Curr Issues Mol Biol 2024; 46:10545-10569. [PMID: 39329977 PMCID: PMC11431004 DOI: 10.3390/cimb46090625] [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: 09/05/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
The study aims to investigate the effects of curcumin on radiation/chemotherapy-induced oral mucositis (R/CIOM) and preliminarily explore its mechanism. Randomized controlled trials were identified from the PubMed, Embase, Web of Science, Cochrane Library, Medline, and Google Scholar databases. RevMan 5.4 was used for statistical analysis to calculate the combined risk ratios (RRs). The mechanism was analyzed through network pharmacology, molecular docking, and a molecular dynamics simulation. The targets of curcumin were collected in HERB, PharmMapper, Targetnet, Swiss Target Prediction, and SuperPred. OMIM, GeneCards, and Disgenet were used to collect relevant targets for R/CIOM. Cytoscape software 3.8.0 was used to construct the component-target-pathway network. Protein-Protein Interaction (PPI) networks were constructed using the STRING database. GO and KEGG enrichment analyses were performed by Metascape. AutoDock Vina 4.2 software was used for molecular docking. The molecular dynamics simulation was performed by Gromacs v2022.03. It is found that 12 studies involving 565 patients were included. Meta-analyses showed that curcumin reduced the incidence of severe R/CIOM (RR 0.42 [0.24, 0.75]) and the mean severity of R/CIOM (MD -0.93 [-1.34, -0.52]). Eleven core target genes were identified in the treatment of R/CIOM with curcumin. The results of molecular docking and the molecular dynamics simulation showed that curcumin had strong binding energy and stability with target proteins including MAPK3, SRC, and TNF. Overall, these findings suggest curcumin can effectively improve severe R/CIOM, perhaps by affecting MAPK3, SRC, and TNF.
Collapse
Affiliation(s)
- Zhi-Xing Chen
- College of Stomatology, Guangxi Medical University, Nanning 530021, China; (Z.-X.C.); (Y.-S.Q.); (B.-H.S.); (B.-Y.G.)
| | - Ya-Shi Qin
- College of Stomatology, Guangxi Medical University, Nanning 530021, China; (Z.-X.C.); (Y.-S.Q.); (B.-H.S.); (B.-Y.G.)
| | - Bang-Hui Shi
- College of Stomatology, Guangxi Medical University, Nanning 530021, China; (Z.-X.C.); (Y.-S.Q.); (B.-H.S.); (B.-Y.G.)
| | - Bi-Yun Gao
- College of Stomatology, Guangxi Medical University, Nanning 530021, China; (Z.-X.C.); (Y.-S.Q.); (B.-H.S.); (B.-Y.G.)
| | - Ren-Chuan Tao
- College of Stomatology, Guangxi Medical University, Nanning 530021, China; (Z.-X.C.); (Y.-S.Q.); (B.-H.S.); (B.-Y.G.)
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning 530021, China
| | - Xiang-Zhi Yong
- College of Stomatology, Guangxi Medical University, Nanning 530021, China; (Z.-X.C.); (Y.-S.Q.); (B.-H.S.); (B.-Y.G.)
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning 530021, China
| |
Collapse
|
2
|
Bogdan C, Islam NAK, Barinberg D, Soulat D, Schleicher U, Rai B. The immunomicrotope of Leishmania control and persistence. Trends Parasitol 2024; 40:788-804. [PMID: 39174373 DOI: 10.1016/j.pt.2024.07.013] [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: 06/20/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/24/2024]
Abstract
Leishmania is an intracellular protozoan transmitted by sand fly vectors; it causes cutaneous, mucocutaneous, or visceral disease. Its growth and survival are impeded by type 1 T helper cell responses, which entail interferon (IFN)-γ-mediated macrophage activation. Leishmania partially escapes this host defense by triggering immune cell and cytokine responses that favor parasite replication rather than killing. Novel methods for in situ analyses have revealed that the pathways of immune control and microbial evasion are strongly influenced by the tissue context, the micro milieu factors, and the metabolism at the site of infection, which we collectively term the 'immunomicrotope'. Understanding the components and the impact of the immunomicrotope will enable the development of novel strategies for the treatment of chronic leishmaniasis.
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany.
| | - Noor-A-Kasida Islam
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - David Barinberg
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany; FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Baplu Rai
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Wasserturmstraße 3/5, D-91054 Erlangen, Germany
| |
Collapse
|
3
|
Khandibharad S, Singh S. Mechanistic study of inhibitory peptides with SHP-1 in hypertonic environment for infection model. Biochim Biophys Acta Gen Subj 2024; 1868:130670. [PMID: 38996989 DOI: 10.1016/j.bbagen.2024.130670] [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: 05/19/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Cutaneous Leishmaniasis, an infectious disease is globally the most prevalent form of leishmaniasis accounting for approximately 1 million cases every year as per world health organization. Infected individuals develop skin lesion which has been reported to be infiltrated by immune cells and parasite with high sodium accumulation creating hypertonic environment. In our work, we tried to mimic the hypertonic environment in virtual environment to study dynamicity of SHP-1 and NFAT5 along with their interactions through molecular dynamics simulation. We validated the SHP-1 and NFAT5 dynamics in infection and HSD conditions to study the impact of hypertonicity derived NFAT5 mediated response to L.major infection. We also evaluated our therapeutic peptides for their binding to SHP-1 and to form stable complex. Membrane stability with the peptides was analyzed to understand their ability to sustain mammalian membrane. We identified PepA to be a potential candidate to interact with SHP-1. Inhibition of SHP-1 through PepA to discern IL-10 and IL-12 reciprocity may be assessed in future and furnish us with a potential therapeutic molecule. HSD mice exhibited high pro-inflammatory response to L.major infection which resulted in reduced lesion size. Contrary to observations in HSD mice, infection model exhibited low pro-inflammatory response and increased lesion size with high parasite load. Thus, increase in NFAT5 expression and reduced SHP-1 expression may result in disease resolving effect which can be further studied through incorporation of synthetic circuit using PepA to modulate IL-10 and IL-12 reciprocity.
Collapse
Affiliation(s)
- Shweta Khandibharad
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SPPU Campus, Pune 411007, INDIA
| | - Shailza Singh
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SPPU Campus, Pune 411007, INDIA.
| |
Collapse
|
4
|
Reyaz E, Puri N, Selvapandiyan A. Global Remodeling of Host Proteome in Response to Leishmania Infection. ACS Infect Dis 2024; 10:5-19. [PMID: 38084821 DOI: 10.1021/acsinfecdis.3c00492] [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: 01/13/2024]
Abstract
The protozoan parasite Leishmania possesses an intrinsic ability to modulate a multitude of pathways in the host, toward aiding its own proliferation. In response, the host reprograms its cellular, immunological, and metabolic machinery to evade the parasite's lethal impact. Besides inducing various antioxidant signaling pathways to counter the elevated stress response proteins like heme oxygenase-1 (HO-1), Leishmania also attempts to delay host cell apoptosis by promoting anti-apoptotic proteins like Bcl-2. The downstream modulation of apoptotic proteins is regulated by effector pathways, including the PI3K/Akt survival pathway, the mitogen-activated protein kinases (MAPKs) signaling pathway, and STAT phosphorylation. In addition, Leishmania assists in its infection in a time-dependent manner by modulating the level of various proteins of autophagic machinery. Immune effector cells, such as mast cells and neutrophils, entrap and kill the pathogen by secreting various granular proteins. In contrast, the host macrophages exert their leishmanicidal effect by secreting various cytokines, such as IL-2, IL-12, etc. An interplay of various signaling pathways occurs in an organized network that is highly specific to both pathogen and host species. This Review analyzes the modulation of expression of proteins, including the cytokines, providing a realistic approach toward understanding the pathophysiology of disease and predicting some prominent markers for disease intervention and vaccine support strategies.
Collapse
Affiliation(s)
- Enam Reyaz
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | | |
Collapse
|
5
|
Lai Y, Yong X, Wei Y, Wei S, Su Z, Tao R. Network Pharmacology and Molecular Docking Reveal the Mechanism of Corydalis saxicola Bunting Total Alkaloids in Treating Radiation-Induced Oral Mucositis. Chem Biodivers 2023; 20:e202300028. [PMID: 37390332 DOI: 10.1002/cbdv.202300028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/02/2023]
Abstract
The study aims to explore the effect and mechanism of total alkaloids of Corydalis saxicola Bunting (CSBTA) in the treatment of radiation induced oral mucositis (RIOM) through network pharmacology and molecular docking. The components and corresponding targets of Corydalis saxicola Bunting were screened by literature review. RIOM related targets were obtained in GeneCards. Cytoscape software was used to construct the component-target-pathway network. Protein-Protein Interaction (PPI) networks was constructed by String database. GO and KEGG enrichment analyses were performed by Metascape. AutoDock Vina 4.2 software was used for molecular docking. There were 26 components of CSBTA targeting 61 genes related to RIOM. Through Cytoscape and PPI analysis, 15 core target genes of CSBTA for treating RIOM were identified. GO functional analysis indicated that CSBTA might play a role through kinase binding and protein kinase activation. KEGG pathway analysis showed that the core targets of CSBTA were mainly focused on cancer and reactive oxygen species (ROS) pathway. The results of molecular docking showed that CSBTA had strong binding energy with target protein including SRC, AKT and EGFR. The study demonstrates that CSBTA may treat RIOM by affecting SRC, AKT and EGFR through ROS pathway.
Collapse
Affiliation(s)
- Yiyuan Lai
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, China
| | - Xiangzhi Yong
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, China
| | - Yongzheng Wei
- Guangxi University of Chinese Medicine, Nanning, China
| | - Shanni Wei
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, China
| | - Zhiheng Su
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Renchuan Tao
- Department of Periodontics and Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention and Treatment for Oral Infectious Diseases, Nanning, China
| |
Collapse
|
6
|
Maksoud S, El Hokayem J. The cytokine/chemokine response in Leishmania/HIV infection and co-infection. Heliyon 2023; 9:e15055. [PMID: 37082641 PMCID: PMC10112040 DOI: 10.1016/j.heliyon.2023.e15055] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/04/2023] Open
Abstract
HIV infection progressively weakens the immune system by infecting and destroying cells involved in host defense. Viral infection symptoms are generated and aggravated as immunosuppression progresses, triggered by the presence of opportunistic infections: among these is leishmaniasis, a disease caused by the intracellular parasite Leishmania. The incidence of this co-infection is growing progressively due to the geographic distribution overlap. Both pathogens infect monocytes/macrophages and dendritic cells, although they can also modulate the activity of other cells without co-infecting, such as T and B lymphocytes. Leishmania/HIV co-infection could be described as a system comprising modulations of cell surface molecule expression, production of soluble factors, and intracellular death activities, leading ultimately to the potentiation of infectivity, replication, and spread of both pathogens. This review describes the cytokine/chemokine response in Leishmania/HIV infection and co-infection, discussing how these molecules modulate the course of the disease and analyzing the therapeutic potential of targeting this network.
Collapse
|
7
|
Alizadeh Z, Omidnia P, Altalbawy FMA, Gabr GA, Obaid RF, Rostami N, Aslani S, Heidari A, Mohammadi H. Unraveling the role of natural killer cells in leishmaniasis. Int Immunopharmacol 2023; 114:109596. [PMID: 36700775 DOI: 10.1016/j.intimp.2022.109596] [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: 10/06/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022]
Abstract
NK cells are known as frontline responders that are efficient in combating several maladies as well as leishmaniasis caused by Leishmania spp. As such they are being investigated to be used for adoptive transfer therapy and vaccine. In spite of the lack of antigen-specific receptors at their surface, NK cells can selectively recognize pathogens, accomplished by the activation of the receptors on the NK cell surface and also as the result of their effector functions. Activation of NK cells can occur through interaction between TLR-2 expressed on NK cells and. LPG of Leishmania parasites. In addition, NK cell activation can occur by cytokines (e.g., IFN-γ and IL-12) that also lead to producing cytokines and chemokines and lysis of target cells. This review summarizes several evidences that support NK cells activation for controlling leishmaniasis and the potentially lucrative roles of NK cells during leishmaniasis. Furthermore, we discuss strategies of Leishmania parasites in inhibiting NK cell functions. Leishmania LPG can utilizes TLR2 to evade host-immune responses. Also, Leishmania GP63 can directly binds to NK cells and modulates NK cell phenotype. Finally, this review analyzes the potentialities to harness NK cells effectiveness in therapy regimens and vaccinations.
Collapse
Affiliation(s)
- Zahra Alizadeh
- Department of Parasitology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Farag M A Altalbawy
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt; Department of Chemistry, University College of Duba, University of Tabuk, Duba 71911, Saudi Arabia
| | - Gamal A Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center, Giza, Egypt
| | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | - Narges Rostami
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliehsan Heidari
- Department of Parasitology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
8
|
Immune Responses in Leishmaniases: An Overview. Trop Med Infect Dis 2022; 7:tropicalmed7040054. [PMID: 35448829 PMCID: PMC9029249 DOI: 10.3390/tropicalmed7040054] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a parasitic, widespread, and neglected disease that affects more than 90 countries in the world. More than 20 Leishmania species cause different forms of leishmaniasis that range in severity from cutaneous lesions to systemic infection. The diversity of leishmaniasis forms is due to the species of parasite, vector, environmental and social factors, genetic background, nutritional status, as well as immunocompetence of the host. Here, we discuss the role of the immune system, its molecules, and responses in the establishment, development, and outcome of Leishmaniasis, focusing on innate immune cells and Leishmania major interactions.
Collapse
|
9
|
Bettadapura M, Roys H, Bowlin A, Venugopal G, Washam CL, Fry L, Murdock S, Wanjala H, Byrum SD, Weinkopff T. HIF-α Activation Impacts Macrophage Function during Murine Leishmania major Infection. Pathogens 2021; 10:pathogens10121584. [PMID: 34959539 PMCID: PMC8706659 DOI: 10.3390/pathogens10121584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/21/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Leishmanial skin lesions are characterized by inflammatory hypoxia alongside the activation of hypoxia-inducible factors, HIF-1α and HIF-2α, and subsequent expression of the HIF-α target VEGF-A during Leishmania major infection. However, the factors responsible for HIF-α activation are not known. We hypothesize that hypoxia and proinflammatory stimuli contribute to HIF-α activation during infection. RNA-Seq of leishmanial lesions revealed that transcripts associated with HIF-1α signaling were induced. To determine whether hypoxia contributes to HIF-α activation, we followed the fate of myeloid cells infiltrating from the blood and into hypoxic lesions. Recruited myeloid cells experienced hypoxia when they entered inflamed lesions, and the length of time in lesions increased their hypoxic signature. To determine whether proinflammatory stimuli in the inflamed tissue can also influence HIF-α activation, we subjected macrophages to various proinflammatory stimuli and measured VEGF-A. While parasites alone did not induce VEGF-A, and proinflammatory stimuli only modestly induced VEGF-A, HIF-α stabilization increased VEGF-A during infection. HIF-α stabilization did not impact parasite entry, growth, or killing. Conversely, the absence of ARNT/HIF-α signaling enhanced parasite internalization. Altogether, these findings suggest that HIF-α is active during infection, and while macrophage HIF-α activation promotes lymphatic remodeling through VEGF-A production, HIF-α activation does not impact parasite internalization or control.
Collapse
Affiliation(s)
- Manjunath Bettadapura
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Hayden Roys
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Anne Bowlin
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Gopinath Venugopal
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (C.L.W.); (S.D.B.)
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Lucy Fry
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Steven Murdock
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Humphrey Wanjala
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (C.L.W.); (S.D.B.)
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Tiffany Weinkopff
- Department of Microbiology and Immunology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (M.B.); (H.R.); (A.B.); (G.V.); (L.F.); (S.M.); (H.W.)
- Correspondence: ; Tel.: +1-501-686-5518
| |
Collapse
|
10
|
Aghaei M, Khanahmad H, Jalali A, Aghaei S, Narimani M, Hosseini SM, Namdar F, Hejazi SH. Effect of transgenic Leishmania major expressing mLLO-Bax-Smac fusion gene in the apoptosis of the infected macrophages. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1666-1675. [PMID: 35432807 PMCID: PMC8976903 DOI: 10.22038/ijbms.2021.56960.12701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 10/31/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Leishmaniasis is a complex infection against which no confirmed vaccine has been reported so far. Transgenic expression of proteins involved in macrophage apoptosis-like BAX through the parasite itself accelerates infected macrophage apoptosis and prevents Leishmania differentiation. So, in the present research, the impact of the transgenic Leishmania major including mLLO-BAX-SMAC proapoptotic proteins was assayed in macrophage apoptosis acceleration. MATERIALS AND METHODS The coding sequence mLLO-Bax-Smac was designed and integrated into the pLexyNeo2 plasmid. The designed sequence was inserted under the 18srRNA locus into the L. major genome using homologous recombination. Then, mLLO-BAX-SMAC expression was studied using the Western blot, and the transgenic parasite pathogenesis was investigated compared with wild-type L. major in vitro and also in vivo. RESULTS Western blot and PCR results approved mLLO-BAX-SMAC expression and proper integration of the mLLO-Bax-Smac fragment under the 18srRNA locus of L. major, respectively. The flow cytometry results revealed faster apoptosis of transgenic Leishmania-infected macrophages compared with wild-type parasite-infected macrophages. Also, the mild lesion with the less parasitic burden of the spleen was observed only in transgenic Leishmania-infected mice. The delayed progression of leishmaniasis was obtained in transgenic strain-injected mice after challenging with wild-type Leishmania. CONCLUSION This study recommended transgenic L. major including mLLO-BAX-SMAC construct as a pilot model for providing a protective vaccine against leishmaniasis.
Collapse
Affiliation(s)
- Maryam Aghaei
- Skin Diseases and Leishmaniasis Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Akram Jalali
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrzad Aghaei
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Manizheh Narimani
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sayed Mohsen Hosseini
- Department of Biostatistics & Epidemiology, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Namdar
- Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran,Skin Diseases and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Corresponding author: Seyed Hossein Hejazi. Skin Diseases and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
11
|
Zayats R, Uzonna JE, Murooka TT. Visualizing the In Vivo Dynamics of Anti- Leishmania Immunity: Discoveries and Challenges. Front Immunol 2021; 12:671582. [PMID: 34093571 PMCID: PMC8172142 DOI: 10.3389/fimmu.2021.671582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/07/2021] [Indexed: 11/20/2022] Open
Abstract
Intravital microscopy, such as 2-photon microscopy, is now a mainstay in immunological research to visually characterize immune cell dynamics during homeostasis and pathogen infections. This approach has been especially beneficial in describing the complex process of host immune responses to parasitic infections in vivo, such as Leishmania. Human-parasite co-evolution has endowed parasites with multiple strategies to subvert host immunity in order to establish chronic infections and ensure human-to-human transmission. While much focus has been placed on viral and bacterial infections, intravital microscopy studies during parasitic infections have been comparatively sparse. In this review, we will discuss how in vivo microscopy has provided important insights into the generation of innate and adaptive immunity in various organs during parasitic infections, with a primary focus on Leishmania. We highlight how microscopy-based approaches may be key to providing mechanistic insights into Leishmania persistence in vivo and to devise strategies for better parasite control.
Collapse
Affiliation(s)
- Romaniya Zayats
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Jude E. Uzonna
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Thomas T. Murooka
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
Holanda VN, Silva WVD, Nascimento PHD, Silva SRB, Cabral Filho PE, Assis SPDO, Silva CAD, Oliveira RND, Figueiredo RCBQD, Lima VLDM. Antileishmanial activity of 4-phenyl-1-[2-(phthalimido-2-yl)ethyl]-1H-1,2,3-triazole (PT4) derivative on Leishmania amazonensis and Leishmania braziliensis: In silico ADMET, in vitro activity, docking and molecular dynamic simulations. Bioorg Chem 2020; 105:104437. [PMID: 33339081 DOI: 10.1016/j.bioorg.2020.104437] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/30/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022]
Abstract
Organic compounds obtained by click chemistry reactions have demonstrated a broad spectrum of biological activities being widely applied for the development of molecules against pathogens of medical and veterinary importance. Cutaneous leishmaniasis (CL), caused by intracellular protozoa parasite of genus Leishmania, comprises a complex of clinical manifestations that affect the skin and mucous membranes. The available drugs for the treatment are toxic and costly, with long periods of treatment, and the emergence of resistant strains has been reported. In this study we investigated the in vitro effects of a phthalimide-1,2,3-triazole derivative, the 4-Phenyl-1-[2-(phthalimido-2-yl)ethyl]-1H-1,2,3-triazole (PT4) obtained by click chemistry, on mammalian cells and on L. amazonensis and L. braziliensis, the causative agents of CL in Brazil. In silico ADMET evaluation of PT4 showed that this molecule has good pharmacokinetic properties with no violation of Lipinski's rules. The in vitro assays showed that PT4 was more selective for both Leishmania species than to mammalian cells. This compound also presented low cytotoxicity to mammalian cells with CC50 > 500 μM. Treatment of promastigote forms with different concentrations of PT4 resulted in ultrastructural alterations, such as plasma membrane wrinkling, shortening of cell body, increased cell volume and cell rupture. The molecular dynamic simulations showed that PT4 interacts with Lanosterol 14 α-demethylase from Leishmania, an essential enzyme of lipid synthesis pathway in this parasite. Our results demonstrated PT4 was effective against both species of Leishmania. PT4 caused a decrease of mitochondrial membrane potential and increased production of reactive oxygen species, which may lead to parasite death. Taken together, our results pointed PT4 as promissing therapeutic agent against CL.
Collapse
Affiliation(s)
- Vanderlan Nogueira Holanda
- Laboratório de Lipídios e Aplicação de Biomoléculas em Doenças Prevalentes e Negligenciadas. Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil; Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil
| | - Welson Vicente da Silva
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil
| | - Pedro Henrique do Nascimento
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil
| | - Sérgio Ruschi Bergamachi Silva
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Av. Nascimento de Castro, 2155 - Morro Branco, 59056-450 Natal, RN, Brazil
| | - Paulo Euzébio Cabral Filho
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil
| | - Shalom Porto de Oliveira Assis
- Núcleo de Pesquisas em Ciências Ambientais e Biotecnologia, Universidade Católica de Pernambuco, Rua do Príncipe, 526, 50050-900 Recife, PE, Brazil
| | - César Augusto da Silva
- Colegiado de Medicina, Universidade Federal do Vale do São Francisco, Avenida José de Sá Maniçoba, s/n - Campus Universitário, 56304-205 Petrolina, PE, Brazil
| | - Ronaldo Nascimento de Oliveira
- Laboratório de Síntese de Compostos Bioativos, Departamento de Química, Universidade Federal Rural de Pernambuco, Rua Dom Manuel de Medeiros, s/n - Dois Irmãos, 52171-900 Recife, PE, Brazil
| | - Regina Celia Bressan Queiroz de Figueiredo
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil
| | - Vera Lucia de Menezes Lima
- Laboratório de Lipídios e Aplicação de Biomoléculas em Doenças Prevalentes e Negligenciadas. Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Avenida Professor Moraes Rego, 1235, 50670-901 Recife, PE, Brazil.
| |
Collapse
|
13
|
Arana-Argáez VE, Ceballos-Góngora E, Alvarez-Sánchez ME, Euan-Canto A, Lara-Riegos J, Torres-Romero JC. In Vitro Activation of Macrophages by an MHC Class II-restricted Trichomonas Vaginalis TvZIP8-derived Synthetic Peptide. Immunol Invest 2020; 51:88-102. [DOI: 10.1080/08820139.2020.1810703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Emanuel Ceballos-Góngora
- Laboratorio De Farmacología, Facultad De Química, Universidad Autónoma De Yucatán, Mérida, México
- Laboratorio De Bioquímica Y Genética Molecular, Facultad De Química, Universidad Autónoma De Yucatán, Mérida, México
| | | | - Antonio Euan-Canto
- Laboratorio De Bioquímica Y Genética Molecular, Facultad De Química, Universidad Autónoma De Yucatán, Mérida, México
| | - Julio Lara-Riegos
- Laboratorio De Bioquímica Y Genética Molecular, Facultad De Química, Universidad Autónoma De Yucatán, Mérida, México
| | - Julio César Torres-Romero
- Laboratorio De Bioquímica Y Genética Molecular, Facultad De Química, Universidad Autónoma De Yucatán, Mérida, México
| |
Collapse
|
14
|
Aghaei M, KhanAhmad H, Aghaei S, Ali Nilforoushzadeh M, Mohaghegh MA, Hejazi SH. The role of Bax in the apoptosis of Leishmania-infected macrophages. Microb Pathog 2019; 139:103892. [PMID: 31778755 DOI: 10.1016/j.micpath.2019.103892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 10/30/2019] [Accepted: 11/23/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Leishmania is a protozoan parasite that nests in macrophages and is responsible for the Leishmaniasis disease. In spite of different defense pathways, last strategy of macrophage for killing parasite is apoptosis process. By permeableizing the mitochondrial outer membrane (MOM). As breaching MOM releases apoptogenic factors like cytochrome-c which activate caspases that result in the destruction of the cell. In this review, we summarized the appropriate manuscripts regarding the bax includes, its different types and the effect of bax on the apoptosis of Leishmania and parasite-infected macrophages. METHODS Information about the role of BAX in the apoptosis of parasite-infected macrophage of recent articles were surveyed by searching computerized bibliographic database PubMed and Google Scholar entering the keywords BAX and leishmaniasis. RESULTS The common studies revealed Leishmania use different survival strategies for inhibiting macrophage apoptosis. As Leishmania by preventing homooligomerization or upregulating the anti-apoptotic molecule Bcl-2 can prohibits proteins of host-cell apoptosis such as Bax that is required for mitochondrial permeabilisation during apoptosis. CONCLUSION With regard to the supportive role of bax in apoptosis and the preventive role of Leishmania in its function, it seems that expression of bax gene in parasite by technologies like transgenic or down regulating of anti-apoptotic molecule Bcl-2 by miRNA could be prompted the apoptosis process of infected-macrophages and inhibited extensive spread of Leishmania and the resulting lesions.
Collapse
Affiliation(s)
- Maryam Aghaei
- Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein KhanAhmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrzad Aghaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Mohammad-Ali Mohaghegh
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Skin Disease and Leishmaniasis Research Center, Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
15
|
Münck NA, Roth J, Sunderkötter C, Ehrchen J. Aryl Hydrocarbon Receptor-Signaling Regulates Early Leishmania major-Induced Cytokine Expression. Front Immunol 2019; 10:2442. [PMID: 31749794 PMCID: PMC6843081 DOI: 10.3389/fimmu.2019.02442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023] Open
Abstract
The early inflammatory skin micromilieu affects resistance in experimental infection with Leishmania major. We pursue the concept that macrophages, which take up parasites during early infection, exert decisive influence on the inflammatory micromilieu after infection. In order to analyze their distinctive potential, we identified differentially regulated genes of murine granuloma macrophages (GMΦ) from resistant and susceptible mice after their infection with metacyclic Leishmania major. We found induction of several cytokines in GMΦ from both strains and a stronger upregulation of the transcription factor aryl hydrocarbon receptor (AhR) in GMΦ from resistant mice. Using both an AhR agonist and antagonist we demonstrated that AhR is involved in Leishmania-induced production of TNF in macrophages. In vivo, single local injection of an AhR agonist in early lesions of susceptible mice caused an increased induction of Tnf and other cytokines in the skin. Importantly, local agonist treatment led to a reduction of disease severity, reduced parasite loads and a weaker Th2 response. Our results demonstrate that local activation of AhR has a beneficial effect in experimental leishmaniasis.
Collapse
Affiliation(s)
- Niels-Arne Münck
- Institute of Immunology, University of Münster, Münster, Germany.,Department of Translational Dermatoinfectiology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Cord Sunderkötter
- Department of Translational Dermatoinfectiology, University of Münster, Münster, Germany.,Department of Dermatology, University of Münster, Münster, Germany.,Department of Dermatology and Venereology, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Jan Ehrchen
- Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
16
|
Mendoza-Oliveros T, Arana-Argáez V, Alvaréz-Sánchez LC, Lara-Riegos J, Alvaréz-Sánchez ME, Torres-Romero JC. Immune Response of BALB/c Mice toward Putative Calcium Transporter Recombinant Protein of Trichomonas vaginalis. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:33-38. [PMID: 30840797 PMCID: PMC6409216 DOI: 10.3347/kjp.2019.57.1.33] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 12/23/2018] [Indexed: 12/12/2022]
Abstract
Trichomoniasis is a common sexually transmitted infection caused by Trichomonas vaginalis, which actually does not exist a vaccine for control or prevention. Thus, the identification of new and potent immunogens in T. vaginalis, which can contribute to the development of a vaccine against this parasite, is necessary. Therefore, the aim of this work was to evaluate the potential of a recombinant Transient Receptor Potential-like channel of T. vaginalis (TvTRPV), as a promising immunogen in BALB/c mice. First, TvTRPV was cloned and expressed as a recombinant protein in Escherichia coli BL21 cells and purified by nickel affinity. Next, BALB/c mice were immunized and the antibody levels in mice serum and cytokines from the supernatant of macrophages and from co-culture systems were evaluated. Recombinant TvTRPV triggered high levels of specific total IgG in sera from the immunized mice. Also, a statistically significant increase of cytokines: IL-1β, IL-6, and TNF-α after stimulation with the corresponding antigens in vitro, was identified. Moreover, co-cultures using CD4+ T cells from immunized mice were able to identify higher levels of IL-10 and IFN-γ. These results were useful to validate the immunogenicity of TvTRPV in BALB/c mice, where IL-10-IFN-γ-secreting cells could play a role in infection control, supporting the potential of TvTRPV as a promising target for vaccine against T. vaginalis.
Collapse
Affiliation(s)
- Tahali Mendoza-Oliveros
- Laboratorio de Bioquímica y Genética Molecular, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida 97069, México
| | - Victor Arana-Argáez
- Laboratorio de Farmacología, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida 97069, México
| | - Leidi C Alvaréz-Sánchez
- Laboratorio de Virología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi" de la Universidad Autónoma de Yucatán, Mérida 97069, México
| | - Julio Lara-Riegos
- Laboratorio de Bioquímica y Genética Molecular, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida 97069, México
| | | | - Julio C Torres-Romero
- Laboratorio de Bioquímica y Genética Molecular, Facultad de Química de la Universidad Autónoma de Yucatán, Mérida 97069, México
| |
Collapse
|
17
|
Arens K, Filippis C, Kleinfelder H, Goetzee A, Reichmann G, Crauwels P, Waibler Z, Bagola K, van Zandbergen G. Anti-Tumor Necrosis Factor α Therapeutics Differentially Affect Leishmania Infection of Human Macrophages. Front Immunol 2018; 9:1772. [PMID: 30108591 PMCID: PMC6079256 DOI: 10.3389/fimmu.2018.01772] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/17/2018] [Indexed: 01/18/2023] Open
Abstract
Tumor necrosis factor α (TNFα) drives the pathophysiology of human autoimmune diseases and consequently, neutralizing antibodies (Abs) or Ab-derived molecules directed against TNFα are essential therapeutics. As treatment with several TNFα blockers has been reported to entail a higher risk of infectious diseases such as leishmaniasis, we established an in vitro model based on Leishmania-infected human macrophages, co-cultured with autologous T-cells, for the analysis and comparison of anti-TNFα therapeutics. We demonstrate that neutralization of soluble TNFα (sTNFα) by the anti-TNFα Abs Humira®, Remicade®, and its biosimilar Remsima® negatively affects infection as treatment with these agents significantly reduces Leishmania-induced T-cell proliferation and increases the number of infected macrophages. By contrast, we show that blockade of sTNFα by Cimzia® does not affect T-cell proliferation and infection rates. Moreover, compared to Remicade®, treatment with Cimzia® does not impair the expression of cytolytic effector proteins in proliferating T-cells. Our data demonstrate that Cimzia® supports parasite control through its conjugated polyethylene glycol (PEG) moiety as PEGylation of Remicade® improves the clearance of intracellular Leishmania. This effect can be linked to complement activation, with levels of complement component C5a being increased upon treatment with Cimzia® or a PEGylated form of Remicade®. Taken together, we provide an in vitro model of human leishmaniasis that allows direct comparison of different anti-TNFα agents. Our results enhance the understanding of the efficacy and adverse effects of TNFα blockers and they contribute to evaluate anti-TNFα therapy for patients living in countries with a high prevalence of leishmaniasis.
Collapse
Affiliation(s)
- Katharina Arens
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | | | | | - Arthur Goetzee
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Peter Crauwels
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Zoe Waibler
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Katrin Bagola
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | - Ger van Zandbergen
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany.,Institute of Immunology, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
18
|
Coêlho EDS, Lopes GLN, Pinheiro IM, Holanda JNPD, Alves MMDM, Carvalho Nogueira N, Carvalho FADA, Carvalho ALM. Emulgel based on amphotericin B and bacuri butter (Platonia insignis Mart.) for the treatment of cutaneous leishmaniasis: characterization and in vitro assays. Drug Dev Ind Pharm 2018; 44:1713-1723. [DOI: 10.1080/03639045.2018.1492610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Elvilene de Sousa Coêlho
- Postgraduate Program of Pharmaceutical Sciences (PPGCF), Federal University of Piauí, Teresina, Brazil
| | - Gláucia Laís Nunes Lopes
- Postgraduate Program of Pharmaceutical Sciences (PPGCF), Federal University of Piauí, Teresina, Brazil
| | - Iluska Martins Pinheiro
- Postgraduate Program of Pharmaceutical Sciences (PPGCF), Federal University of Piauí, Teresina, Brazil
| | | | | | | | | | | |
Collapse
|
19
|
Guimarães-Pinto K, Nascimento DO, Corrêa-Ferreira A, Morrot A, Freire-de-Lima CG, Lopes MF, DosReis GA, Filardy AA. Trypanosoma cruzi Infection Induces Cellular Stress Response and Senescence-Like Phenotype in Murine Fibroblasts. Front Immunol 2018; 9:1569. [PMID: 30038622 PMCID: PMC6047053 DOI: 10.3389/fimmu.2018.01569] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/25/2018] [Indexed: 12/20/2022] Open
Abstract
Trypanosoma cruzi infects and replicates within a wide variety of immune and non-immune cells. Here, we investigated early cellular responses induced in NIH-3T3 fibroblasts upon infection with trypomastigote forms of T. cruzi. We show that fibroblasts were susceptible to T. cruzi infection and started to release trypomastigotes to the culture medium after 4 days of infection. Also, we found that T. cruzi infection reduced the number of fibroblasts in 3-day cell cultures, by altering fibroblast proliferation. Infected fibroblasts displayed distinctive phenotypic alterations, including enlarged and flattened morphology with a nuclei accumulation of senescence-associated heterochromatin foci. In addition, infection induced an overexpression of the enzyme senescence-associated β-galactosidase (SA-β-gal), an activation marker of the cellular senescence program, as well as the production of cytokines and chemokines involved with the senescence-associated secretory phenotype (SASP) such as IL-6, TNF-α, IL-1β, and MCP-1. Infected fibroblasts released increased amounts of stress-associated factors nitric oxide (NO) and reactive oxygen species (ROS), and the treatment with antioxidants deferoxamine (DFO) and N-acetylcysteine reduced ROS generation, secretion of SASP-related cytokine IL-6, SA-β-gal activity, and parasite load by infected fibroblasts. Taken together, our data suggest that T. cruzi infection triggers a rapid cellular stress response followed by induction of a senescent-like phenotype in NIH-3T3 fibroblasts, enabling them to act as reservoirs of parasites during the early stages of the Chagas disease.
Collapse
Affiliation(s)
- Kamila Guimarães-Pinto
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Antonia Corrêa-Ferreira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre Morrot
- Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Celio G Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcela F Lopes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - George A DosReis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional para Pesquisa Translacional em Saúde e Ambiente na Região Amazônica, Conselho Nacional de Desenvolvimento Científico e Tecnológico, Rio de Janeiro, Brazil
| | - Alessandra A Filardy
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Lei Q, Li L, Zhang S, Li T, Zhang X, Ding X, Qin B. HEV ORF3 downregulates TLR7 to inhibit the generation of type I interferon via impairment of multiple signaling pathways. Sci Rep 2018; 8:8585. [PMID: 29872132 PMCID: PMC5988675 DOI: 10.1038/s41598-018-26975-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Hepatitis E is the most common type of acute hepatitis prevalent worldwide. The open reading frame 3 protein of HEV (HEV ORF3) is proposed to create a favorable environment for viral replication and pathogenesis. However, the mechanisms by which HEV overcomes the effects of host immunity, particularly the role of ORF3, remain to be established. Expression of IFNα and IFNβ in supernatant and cell samples was examined via ELISA and quantitative RT-PCR. The protein levels of specific signaling factors in cells overexpressing HEV ORF3 were examined via western blot. Analyses of cells transfected with vectors expressing ORF3 demonstrated that HEV ORF3 significantly impairs the generation of endogenous type I interferon through downregulating TLR3 and TLR7 as well as their corresponding downstream signaling pathways. Moreover, inhibition of NFκB, JAK/STAT and JNK/MAPK signaling pathways contributed significantly to suppression of increased levels of TLR7. Levels of p-P65, p-STAT1 and p-JNK were markedly impaired in ORF3-expressing cells, even upon treatment with the respective agonists. HEV ORF3 inhibits the production of endogenous type I interferon through downregulation of TLR3 and TLR7. Furthermore, suppression of TLR7 is achieved through impairment of multiple signaling pathways, including NFκB, JAK/STAT and JNK/MAPK.
Collapse
Affiliation(s)
- Qingsong Lei
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases,Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lin Li
- Department of hepatic diseases, Chongqing Tranditional Chinese Medicine Hospital, Chongqing, 400011, China
| | - Shujun Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases,Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tianju Li
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases,Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases,Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaolin Ding
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases,Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bo Qin
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases,Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
21
|
ELIKAEE S, MOHEBALI M, ESLAMI H, REZAEI S, NAJAFIAN HR, KAZEMI-RAD E, KESHAVARZ H, ESHRAGHIAN MR, HAJJARAN H, OSHAGHI MA, AYAZIAN MAVI S. Comparison of p27 Gene Expression of Promastigote and Amastigote Forms of Leishmania major (MRHO/IR/75/ER) by Real-time RT-PCR. IRANIAN JOURNAL OF PARASITOLOGY 2018; 13:186-192. [PMID: 30069202 PMCID: PMC6068362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
BACKGROUND Cutaneous leishmaniasis (CL) is one of the world health problems. Leishmania major is the etiological agent of zoonotic cutaneous leishmaniasis (ZCL). Promastigote and amastigote are two morphological forms of Leishmania parasites that express different proteins and p27 is an important gene encoding cytochrome c oxidase (COX) component. P27 gene expresses a 27 kDa protein that essential in ATP synthesis. This study aimed to compare p27 gene expression in promastigote and amastigote forms in Iranian strain of L. major (MRHO/IR/75/ER). METHODS This study was conducted in 2015. Clinical isolates of CL patients from north, center, west and south parts of Iran were collected and identified by PCRRFLP. After RNA extraction of promastigotes and amastigotes and cDNA synthesis, the expression level of p27 gene was compared by real-time RT-PCR. RESULTS By comparison of expression level between amastigote and promastigote forms of Iranian strain of L. major, up-regulation of p27 gene (2.73 fold) was observed in amastigotes. Moreover, there was no significant difference in p27 gene expression between L. major isolates. CONCLUSION p27 gene and protein can be considered as a target in recombinant vaccine production and treatment process.
Collapse
Affiliation(s)
- Samira ELIKAEE
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi MOHEBALI
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran, Center for Endemic Parasites of Iran, Tehran University of Medical Sciences, Tehran, Iran,Correspondence
| | - Hamid ESLAMI
- Dept. of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sassan REZAEI
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza NAJAFIAN
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein KESHAVARZ
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza ESHRAGHIAN
- Dept. of Biostatistics and Epidemiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa HAJJARAN
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali OSHAGHI
- Dept. of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara AYAZIAN MAVI
- Dept. of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
von Stebut E, Tenzer S. Cutaneous leishmaniasis: Distinct functions of dendritic cells and macrophages in the interaction of the host immune system with Leishmania major. Int J Med Microbiol 2017; 308:206-214. [PMID: 29129568 DOI: 10.1016/j.ijmm.2017.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/30/2017] [Accepted: 11/05/2017] [Indexed: 12/12/2022] Open
Abstract
Leishmaniasis is transmitted by sand flies leading to parasite inoculation into skin. In the mammalian host, the parasite primarily resides in skin macrophages (MΦ) and dendritic cells (DC). MΦ are silently invaded by the parasite eliciting a stress response, whereas DC become activated, release IL-12, and prime antigen-specific T cells. Here we review the basics of the immune response against this human pathogen and elucidate the role and function DC and MΦ for establishment of protective immunity against leishmaniasis. We focus on cell type-specific differences in parasite uptake, phagocyte activation and processing of parasite antigens to facilitate an understanding how their respective function may be modulated e.g. under therapeutic considerations.
Collapse
Affiliation(s)
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
23
|
Kiessling S, Dubeau-Laramée G, Ohm H, Labrecque N, Olivier M, Cermakian N. The circadian clock in immune cells controls the magnitude of Leishmania parasite infection. Sci Rep 2017; 7:10892. [PMID: 28883509 PMCID: PMC5589941 DOI: 10.1038/s41598-017-11297-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/22/2017] [Indexed: 11/27/2022] Open
Abstract
The intracellular parasite Leishmania uses neutrophils and macrophages as host cells upon infection. These immune cells harbour their own intrinsic circadian clocks, known to influence many aspects of their functions. Therefore, we tested whether the host circadian clocks regulate the magnitude of Leishmania major infection in mice. The extent of parasitic infection varied over 24 h in bone marrow-derived macrophages in vitro and in two different in vivo models, footpad and peritoneal cavity infection. In vivo this was paralleled by time of day-dependent neutrophil and macrophage infiltration to the infection site and rhythmic chemokine expression. Thus, rhythmic parasitic infection observed in vivo was likely initiated by the circadian expression of chemoattractants and the subsequent rhythmic infiltration of neutrophils and macrophages. Importantly, all rhythms were abolished in clock-deficient macrophages and when mice lacking the circadian clock in immune cells were infected. Therefore we demonstrated a critical role for the circadian clocks in immune cells in modulating the magnitude of Leishmania infection. To our knowledge this is the first report showing that the circadian clock controls infection by protozoan parasites in mammals. Understanding the timed regulation of host-parasite interactions will allow developing better prophylactic and therapeutic strategies to fight off vector-borne diseases.
Collapse
Affiliation(s)
- Silke Kiessling
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada.,Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | | | - Hyejee Ohm
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Centre, Department of Medicine and Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Martin Olivier
- Department of Medicine, Microbiology and Immunology, McGill University, and Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
24
|
Deep DK, Singh R, Bhandari V, Verma A, Sharma V, Wajid S, Sundar S, Ramesh V, Dujardin JC, Salotra P. Increased miltefosine tolerance in clinical isolates of Leishmania donovani is associated with reduced drug accumulation, increased infectivity and resistance to oxidative stress. PLoS Negl Trop Dis 2017; 11:e0005641. [PMID: 28575060 PMCID: PMC5470736 DOI: 10.1371/journal.pntd.0005641] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 06/14/2017] [Accepted: 05/13/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Miltefosine (MIL) is an oral antileishmanial drug used for treatment of visceral leishmaniasis (VL) in the Indian subcontinent. Recent reports indicate a significant decline in its efficacy with a high rate of relapse in VL as well as post kala-azar dermal leishmaniasis (PKDL). We investigated the parasitic factors apparently involved in miltefosine unresponsiveness in clinical isolates of Leishmania donovani. METHODOLOGY L. donovani isolated from patients of VL and PKDL at pretreatment stage (LdPreTx, n = 9), patients that relapsed after MIL treatment (LdRelapse, n = 7) and parasites made experimentally resistant to MIL (LdM30) were included in this study. MIL uptake was estimated using liquid chromatography coupled mass spectrometry. Reactive oxygen species and intracellular thiol content were measured fluorometrically. Q-PCR was used to assess the differential expression of genes associated with MIL resistance. RESULTS LdRelapse parasites exhibited higher IC50 both at promastigote level (7.92 ± 1.30 μM) and at intracellular amastigote level (11.35 ± 6.48 μM) when compared with LdPreTx parasites (3.27 ± 1.52 μM) and (3.85 ± 3.11 μM), respectively. The percent infectivity (72 hrs post infection) of LdRelapse parasites was significantly higher (80.71 ± 5.67%, P<0.001) in comparison to LdPreTx (60.44 ± 2.80%). MIL accumulation was significantly lower in LdRelapse parasites (1.7 fold, P<0.001) and in LdM30 parasites (2.4 fold, P<0.001) when compared with LdPreTx parasites. MIL induced ROS levels were significantly lower (p<0.05) in macrophages infected with LdRelapse while intracellular thiol content were significantly higher in LdRelapse compared to LdPreTx, indicating a better tolerance for oxidative stress in LdRelapse isolates. Genes associated with oxidative stress, metabolic processes and transporters showed modulated expression in LdRelapse and LdM30 parasites in comparison with LdPreTx parasites. CONCLUSION The present study highlights the parasitic factors and pathways responsible for miltefosine unresponsiveness in VL and PKDL.
Collapse
Affiliation(s)
- Deepak Kumar Deep
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
- Department of Biotechnology, Faculty of Science, Jamia Hamdard, New Delhi, India
| | - Ruchi Singh
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
| | - Vasundhra Bhandari
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
| | - Aditya Verma
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
| | - Vanila Sharma
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
| | - Saima Wajid
- Department of Biotechnology, Faculty of Science, Jamia Hamdard, New Delhi, India
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - V. Ramesh
- Dermatology Department, Safdarjung Hospital and Vardhman Mahavir Medical College (VMMC), New Delhi, India
| | - Jean Claude Dujardin
- Unit of Molecular Parasitology, Department of Parasitology, Institute of Tropical Medicine, Antwerp, Belgium
| | - Poonam Salotra
- National Institute of Pathology (ICMR), Safdarjung Hospital Campus, New Delhi, India
| |
Collapse
|
25
|
Endoplasmic reticulum stress and unfolded protein response in infection by intracellular parasites. Future Sci OA 2017; 3:FSO198. [PMID: 28883998 PMCID: PMC5583660 DOI: 10.4155/fsoa-2017-0020] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/21/2017] [Indexed: 12/30/2022] Open
Abstract
Perturbations of the physiological status of the endoplasmic reticulum (ER) trigger a specific response known as the ER stress response or unfolded protein response (UPR). In mammalian cells, the UPR is mediated by three ER transmembrane proteins (IRE1, PERK and ATF6) which activate three signaling cascades to restore ER homeostasis. In recent years, a cross-talk between UPR, inflammatory and microbial sensing pathways has been elucidated. Pathogen infection can lead to UPR activation; moreover, several pathogens subvert the UPR to promote their survival and replication. While the UPR in viral and bacterial infection has been characterized, little is known about the role of UPR in intracellular parasite infection. Here, we review recent findings on UPR induction/modulation by intracellular parasites in host cells.
Collapse
|
26
|
Tomiotto-Pellissier F, Cataneo AHD, Orsini TM, Thomazelli APFDS, Dalevedo GA, de Oliveira AG, Panagio LA, Costa IN, Conchon-Costa I, Pavanelli WR, Almeida RS. Galleria mellonella hemocytes: A novel phagocytic assay for Leishmania (Viannia) braziliensis. J Microbiol Methods 2016; 131:45-50. [DOI: 10.1016/j.mimet.2016.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/23/2016] [Accepted: 10/01/2016] [Indexed: 11/28/2022]
|
27
|
Grogg MW, Braydich-Stolle LK, Maurer-Gardner EI, Hill NT, Sakaram S, Kadakia MP, Hussain SM. Modulation of miRNA-155 alters manganese nanoparticle-induced inflammatory response. Toxicol Res (Camb) 2016; 5:1733-1743. [PMID: 30090472 DOI: 10.1039/c6tx00208k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/26/2016] [Indexed: 01/02/2023] Open
Abstract
Regulation of gene expression by non-coding RNAs, such as microRNAs (miRNAs), is increasingly being examined in a variety of disciplines. Here we evaluated changes in miRNA expression following metallic nanoparticle (NP) exposure in a mouse neuronal co-culture model. Exposure to manganese (Mn) NPs resulted in oxidative stress, inflammation, and toxicity. Next-generation sequencing (NGS) following an 8 h exposure to Mn NPs (low and high doses) revealed several miRNA candidates that modulate NP induced responses. The lead candidate identified was miR-155, which showed a dose dependent decrease in expression upon Mn exposure. Introduction of a miR-155 mimic into the co-culture to restore miR-155 expression completely abrogated the Mn NP-induced gene and protein expression of inflammatory markers TNF-α and IL-6. Taken together, this study is the first report where global NP-induced miRNA expression changes were used to identify and then modulate negative impacts of metallic NP exposure in a neuronal model. These findings demonstrate that unique miRNA expression profiles provide novel targets for manipulating gene and protein expression, and therefore provide the potential of modifying cellular responses to NP exposure.
Collapse
Affiliation(s)
- Matthew W Grogg
- Molecular Bioeffects Branch , Bioeffects Division , Airman Systems Directorate , AFRL , 711 HPW/RHDJ , Wright-Patterson Air Force Base , Dayton , OH 45433 , USA . ; Tel: +1 (937) 904-9517
| | - Laura K Braydich-Stolle
- Molecular Bioeffects Branch , Bioeffects Division , Airman Systems Directorate , AFRL , 711 HPW/RHDJ , Wright-Patterson Air Force Base , Dayton , OH 45433 , USA . ; Tel: +1 (937) 904-9517
| | - Elizabeth I Maurer-Gardner
- Molecular Bioeffects Branch , Bioeffects Division , Airman Systems Directorate , AFRL , 711 HPW/RHDJ , Wright-Patterson Air Force Base , Dayton , OH 45433 , USA . ; Tel: +1 (937) 904-9517
| | - Natasha T Hill
- Department of Biochemistry and Molecular Biology , Boonshoft School of Medicine , Wright State University , Dayton , OH 45435 , USA
| | - Suraj Sakaram
- Department of Biochemistry and Molecular Biology , Boonshoft School of Medicine , Wright State University , Dayton , OH 45435 , USA
| | - Madhavi P Kadakia
- Department of Biochemistry and Molecular Biology , Boonshoft School of Medicine , Wright State University , Dayton , OH 45435 , USA
| | - Saber M Hussain
- Molecular Bioeffects Branch , Bioeffects Division , Airman Systems Directorate , AFRL , 711 HPW/RHDJ , Wright-Patterson Air Force Base , Dayton , OH 45433 , USA . ; Tel: +1 (937) 904-9517
| |
Collapse
|
28
|
Dos-Santos A, Carvalho-Kelly L, Dick C, Meyer-Fernandes J. Innate immunomodulation to trypanosomatid parasite infections. Exp Parasitol 2016; 167:67-75. [DOI: 10.1016/j.exppara.2016.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 05/13/2016] [Accepted: 05/21/2016] [Indexed: 01/05/2023]
|
29
|
JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol Mol Biol Rev 2016; 80:793-835. [PMID: 27466283 DOI: 10.1128/mmbr.00043-14] [Citation(s) in RCA: 343] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs), as members of the mitogen-activated protein kinase (MAPK) family, mediate eukaryotic cell responses to a wide range of abiotic and biotic stress insults. JNKs also regulate important physiological processes, including neuronal functions, immunological actions, and embryonic development, via their impact on gene expression, cytoskeletal protein dynamics, and cell death/survival pathways. Although the JNK pathway has been under study for >20 years, its complexity is still perplexing, with multiple protein partners of JNKs underlying the diversity of actions. Here we review the current knowledge of JNK structure and isoforms as well as the partnerships of JNKs with a range of intracellular proteins. Many of these proteins are direct substrates of the JNKs. We analyzed almost 100 of these target proteins in detail within a framework of their classification based on their regulation by JNKs. Examples of these JNK substrates include a diverse assortment of nuclear transcription factors (Jun, ATF2, Myc, Elk1), cytoplasmic proteins involved in cytoskeleton regulation (DCX, Tau, WDR62) or vesicular transport (JIP1, JIP3), cell membrane receptors (BMPR2), and mitochondrial proteins (Mcl1, Bim). In addition, because upstream signaling components impact JNK activity, we critically assessed the involvement of signaling scaffolds and the roles of feedback mechanisms in the JNK pathway. Despite a clarification of many regulatory events in JNK-dependent signaling during the past decade, many other structural and mechanistic insights are just beginning to be revealed. These advances open new opportunities to understand the role of JNK signaling in diverse physiological and pathophysiological states.
Collapse
|
30
|
Castañeda-Lopez ME, Garza-Veloz I, Lopez-Hernandez Y, Barbosa-Cisneros OY, Martinez-Fierro ML. Anti-Inflammatory Effects of Modified Adenoviral Vectors for Gene Therapy: A View through Animal Models Tested. Immunol Invest 2016; 45:450-70. [PMID: 27245510 DOI: 10.3109/08820139.2016.1168831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The central dogma of gene therapy relies on the application of novel therapeutic genes to treat or prevent diseases. The main types of vectors used for gene transfer are adenovirus, retrovirus, lentivirus, liposome, and adeno-associated virus vectors. Gene therapy has emerged as a promising alternative for the treatment of inflammatory diseases. The main targets are cytokines, co-stimulatory molecules, and different types of cells from hematological and mesenchymal sources. In this review, we focus on molecules with anti-inflammatory effects used for in vivo gene therapy mediated by adenoviral gene transfer in the treatment of immune-mediated inflammatory diseases, with particular emphasis on autoinflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- M E Castañeda-Lopez
- a Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico.,b Centro de Innovacion Tecnologica e Industrial, Unidad Academica de Ingenieria Electrica , Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| | - I Garza-Veloz
- a Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico.,b Centro de Innovacion Tecnologica e Industrial, Unidad Academica de Ingenieria Electrica , Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| | - Y Lopez-Hernandez
- c CONACyT Research Fellow, Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud , Universidad Autonoma de Zacatecas , Mexico
| | - O Y Barbosa-Cisneros
- d Laboratory of Cell and Molecular Biology, Unidad Academica de Ciencias Quimicas de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| | - M L Martinez-Fierro
- a Molecular Medicine Laboratory, Unidad Academica de Medicina Humana y Ciencias de la Salud de la Universidad Autonoma de Zacatecas , Zacatecas , Mexico.,b Centro de Innovacion Tecnologica e Industrial, Unidad Academica de Ingenieria Electrica , Universidad Autonoma de Zacatecas , Zacatecas , Mexico
| |
Collapse
|
31
|
Zeng J, Chen QW, Yu ZY, Zhang JR, Chen DL, Song C, Luo J, Zhang C, Wang SL, Chen JP. Regulation of intrinsic apoptosis in cycloheximide-treated macrophages by the Sichuan human strain of Chinese Leishmania isolates. Acta Trop 2016; 153:101-10. [PMID: 26482137 DOI: 10.1016/j.actatropica.2015.10.010] [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: 03/29/2015] [Revised: 08/27/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
Abstract
Leishmania spp. are able to survive and proliferate inside mammals' mononuclear phagocytes, causing Leishmaniasis. Previous studies have noted that the regulation of apoptosis in host cells by these parasites may contribute to their ability to evade the immune system. However, current results remain unclear about whether the parasites can promote or delay the apoptotic process in host cells, because the regulatory effect of Leishmania was assumed to be strain-, species- and even infection time-dependent. The aim of this study was to investigate whether the Sichuan isolates of Chinese Leishmania (SC10H2) can alter the process of intrinsic apoptosis induced by cycloheximide in different types of macrophage cell lines and to determine in which steps of the signaling pathway the parasites were involved. Human THP-1 and mouse RAW264.7 macrophages were infected by SC10H2 promastigotes followed by cycloheximide stimulation to assess the alteration of intrinsic apoptosis in these cells. The results indicated that SC10H2 infection of human THP-1 macrophages could promote the initiation of intrinsic apoptosis, but completely opposite results were found in mouse RAW264.7 macrophages. Nevertheless, the expression of Bcl-2 and the DNA fragmentation rates were not altered by SC10H2 infection in the cell lines used in the experiments. This study suggests that SC10H2 promastigote infection is able to promote and delay the transduction of early apoptotic signals induced by cycloheximide in THP-1 and RAW264.7 macrophages, revealing that the regulation of intrinsic apoptosis in host cells by SC10H2 in vitro occurs in a host cell-dependent manner. The data from this study might play a significant role in further understanding the relationship between Leishmania and different host cells.
Collapse
|
32
|
Dameshghi S, Zavaran-Hosseini A, Soudi S, Shirazi FJ, Nojehdehi S, Hashemi SM. Mesenchymal stem cells alter macrophage immune responses to Leishmania major infection in both susceptible and resistance mice. Immunol Lett 2015; 170:15-26. [PMID: 26703818 DOI: 10.1016/j.imlet.2015.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/30/2015] [Accepted: 12/04/2015] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are attracted to inflammation site and switch immune system to modulate inflammatory responses. This ability makes MSCs the best candidate cells for stem cell therapy of infection diseases. Therefore, we aimed to evaluate the modulatory effect of adipose-derived MSCs (AD-MSCs) on macrophages in Leishmania (L.) major infection. Macrophages and MSCs were isolated from both susceptible (BALB/c) and resistance (C57BL/6) strains. After co-culture of AD-MSCs with macrophages using a transwell system, we assessed MSCs-educated macrophage responses to L. major infection. Our results indicated suppression in levels of tumor necrosis factor α (TNF-α) and interleukin 10 (IL-10) of MSCs co-cultured macrophages in response to L. major infection. To clarify the effects of this suppression on inflammatory conditions, TNF-α/IL-10 ratio was calculated, indicating an increase in TNF-α/IL-10 ratio in MSCs co-cultured groups. The higher TNF-α/IL-10 ratio was observed in BALB/c macrophages co-cultured with BALB/c MSCs. Nitric oxide (NO) assay presented a significant reduction in the supernatant of all MSCs co-cultured groups compared to control. We observed a significant reduction in phagocytosis of MSCs co-cultured groups in response to L. major infection without any significant differences in the phagocytic index. In conclusion, our results represented a new spectrum of immunomodulation induced by MSCs co-cultured with macrophages in response to L. major infection. The magnitude of immunoregulation was different between BALB/c and C57BL/6 strains. Our findings also showed that MSCs exerted potential effect of M1 polarization due to unequal decrease in levels of TNF-α and IL-10 when we considered TNF-α and IL-10as representatives of M1 and M2 phenotypes, respectively. Induction of inflammatory cytokine milieu and reduction in level of IL-10 provides a new hope for stem cell therapy of leishmaniasis in susceptible models.
Collapse
Affiliation(s)
- Safura Dameshghi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ahmad Zavaran-Hosseini
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fatemeh Jalali Shirazi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shahrzad Nojehdehi
- Immunology Department, Stem Cell Technology Research Center, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Chen R, Xi L, Huang X, Ma T, Ren H, Ji G. Effect of Jun N-terminal kinase 1 and 2 on the replication of Penicillium marneffei in human macrophages. Microb Pathog 2015; 82:1-6. [PMID: 25792289 DOI: 10.1016/j.micpath.2015.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 11/15/2022]
Abstract
Penicillium marneffei (P. marneffei) is a human pathogen which persists in macrophages and threatens the immunocompromised patients. To clarify the mechanisms involved, we evaluated the effect of c-Jun N-terminal kinase 1 and 2 (JNK1/2) on cytokine expression, phagosomal maturation and multiplication of P. marneffei in P. marneffei-stimulated human macrophages. P. marneffei induced the rapid phosphorylation of JNK1/2. Using the specific inhibitor of JNK1/2 (SP600125), we found that the inhibition of JNK1/2 suppressed P. marneffei-induced tumor necrosis factor-α and IL-10 production. In addition, the presence of SP600125 increased phagosomal acidification and maturation and decreased intracellular replication. These data suggest that JNK1/2 may play an important role in promoting the replication of P. marneffei. Our findings further indicate that the pathogen through the JNK1/2 pathway may attenuate the immune response and macrophage antifungal function.
Collapse
Affiliation(s)
- Renqiong Chen
- Department of Dermatology, Lianyungang First People's Hospital, Lianyungang 222002, China
| | - Liyan Xi
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaowen Huang
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tuan Ma
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hong Ren
- Department of Dermatology, Lianyungang First People's Hospital, Lianyungang 222002, China
| | - Guangquan Ji
- Department of Dermatology, Lianyungang First People's Hospital, Lianyungang 222002, China.
| |
Collapse
|
34
|
Podinovskaia M, Descoteaux A. Leishmania and the macrophage: a multifaceted interaction. Future Microbiol 2015; 10:111-29. [DOI: 10.2217/fmb.14.103] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
ABSTRACT Leishmania, the causative agent of leishmaniases, is an intracellular parasite of macrophages, transmitted to humans via the bite of its sand fly vector. This protozoan organism has evolved strategies for efficient uptake into macrophages and is able to regulate phagosome maturation in order to make the phagosome more hospitable for parasite growth and to avoid destruction. As a result, macrophage defenses such as oxidative damage, antigen presentation, immune activation and apoptosis are compromised whereas nutrient availability is improved. Many Leishmania survival factors are involved in shaping the phagosome and reprogramming the macrophage to promote infection. This review details the complexity of the host–parasite interactions and summarizes our latest understanding of key events that make Leishmania such a successful intracellular parasite.
Collapse
Affiliation(s)
- Maria Podinovskaia
- INRS – Institut Armand-Frappier & Center for Host–Parasite Interactions, 531 boul. des Prairies, Laval, Quebec, H7V 1B7, Canada
| | - Albert Descoteaux
- INRS – Institut Armand-Frappier & Center for Host–Parasite Interactions, 531 boul. des Prairies, Laval, Quebec, H7V 1B7, Canada
| |
Collapse
|
35
|
Daftarian PM, Ager AL, Stone G. A Targeted and Adjuvanted Nanoparticle for Immunochemotherapy of Leishmania Infections. CURRENT TROPICAL MEDICINE REPORTS 2014. [DOI: 10.1007/s40475-014-0023-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Innate immunity to Leishmania infection: within phagocytes. Mediators Inflamm 2014; 2014:754965. [PMID: 25110400 PMCID: PMC4119695 DOI: 10.1155/2014/754965] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/24/2022] Open
Abstract
Infection by Leishmania takes place in the context of inflammation and tissue repair. Besides tissue resident macrophages, inflammatory macrophages and neutrophils are recruited to the infection site and serve both as host cells and as effectors against infection. Recent studies suggest additional important roles for monocytes and dendritic cells. This paper addresses recent experimental findings regarding the regulation of Leishmania major infection by these major phagocyte populations. In addition, the role of IL-4 on dendritic cells and monocytes is discussed.
Collapse
|