1
|
Santos HLC, Pereira GL, do Reis RB, Rodrigues IC, Masini d’Avila C, Vidal VE. Using Acanthamoeba spp. as a cell model to evaluate Leishmania infections. PLoS Negl Trop Dis 2024; 18:e0012517. [PMID: 39356724 PMCID: PMC11472918 DOI: 10.1371/journal.pntd.0012517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/14/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024] Open
Abstract
Leishmaniasis represents a severe global health problem. In the last decades, there have been significant challenges in controlling this disease due to the unavailability of licensed vaccines, the high toxicity of the available drugs, and an unrestrained surge of drug-resistant parasites, and human immunodeficiency virus (HIV)-Leishmania co-infections. Leishmania spp. preferentially invade macrophage lineage cells of vertebrates for replication after subverting cellular functions of humans and other mammals. These early events in host-parasite interactions are likely to influence the future course of the disease. Thus, there is a continuing need to discover a simple cellular model that reproduces the in vivo pathogenesis. Acanthamoeba spp. are non-mammalian phagocytic amoeba with remarkable similarity to the cellular and functional aspects of macrophages. We aimed to assess whether the similarity reported between macrophages and Acanthamoeba spp. is sufficient to reproduce the infectivity of Leishmania spp. Herein, we analyzed co-cultures of Acanthamoeba castellanii or Acanthamoeba polyphaga with Leishmania infantum, Leishmania amazonensis, Leishmania major, and Leishmania braziliensis. Light and fluorescence microscopy revealed that the flagellated promastigotes attach to the A. castellanii and/or A. polyphaga in a bipolar and or random manner, which initiates their uptake via pseudopods. Once inside the cells, the promastigotes undergo significant changes, which result in the obligatory amastigote-like intracellular form. There was a productive infection with a continuous increase in intracellular parasites. However, we frequently observed intracellular amastigotes in vacuoles, phagolysosomes, and the cytosol of Acanthamoeba spp. Our findings corroborate that Leishmania spp. infects Acanthamoeba spp. and replicates in them but does not cause their rapid degeneration or lysis. Overall, the evidence presented here confirms that Acanthamoeba spp. have all prerequisites and can help elucidate how Leishmania spp. infect mammalian cells. Future work exposing the mechanisms of these interactions should yield novel insights into how these pathogens exploit amoebae.
Collapse
Affiliation(s)
- Helena Lúcia Carneiro Santos
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
- Coleção de Protozoários da FIOCRUZ, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Parasitologia, Departamento de Patologia da Faculdade de Medicina da Universidade Federal Fluminense, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriela Linhares Pereira
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rhagner Bonono do Reis
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Igor Cardoso Rodrigues
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Masini d’Avila
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vitor Ennes Vidal
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Bamra T, Shafi T, Das S, Kumar M, Das P. Leishmania donovani mevalonate kinase regulates host actin for inducing phagocytosis. Biochimie 2024; 220:31-38. [PMID: 38123120 DOI: 10.1016/j.biochi.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Despite the well-established role of macrophages in phagocytosing Leishmania, the contribution of the parasite to this process is not well understood. Present study provides insights into the mechanism underlying the MVK-induced entry of L. donovani and improve our knowledge of host-pathogen interactions. We have discussed Mevalonate kinase (MVK)-induced actin reorganization, modulation of signaling pathways and host cell functions. Our results show that LdMVK gains access to macrophage cytosol and induces actin assembly modulation through the activation of actin-related proteins: VASP, Src and ERM. We have also demonstrated that LdMVK induces Ca2+ signaling and Akt pathway in macrophages, which are critical components of Leishmania survival and proliferation. Interestingly, we found that antibodies against LdMVK can kill Leishmania-infected macrophages in culture by forming extracellular traps, highlighting the potential of LdMVK in inhibiting parasite death. Overall, LdMVK is a virulent factor in Leishmania that mediates parasite internalization and host modulation by targeting host proteins phosphorylation and calcium homeostasis having significant implications in disease progression.
Collapse
Affiliation(s)
- Tanvir Bamra
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India.
| | - Taj Shafi
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India.
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Phulwarisharif, Patna, Bihar, 801 507, India.
| | - Manjay Kumar
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India.
| | - Pradeep Das
- Department of Molecular Biology, ICMR- Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, Bihar, 800 007, India; Division of Parasitology, ICMR-National Institute of Cholera and Enteric Diseases, Beleghata, Kolkata, West Bengal, 700 010, India.
| |
Collapse
|
3
|
Shrivastava S, Sarkar P, Preira P, Salomé L, Chattopadhyay A. Cholesterol-Dependent Dynamics of the Serotonin 1A Receptor Utilizing Single Particle Tracking: Analysis of Diffusion Modes. J Phys Chem B 2022; 126:6682-6690. [PMID: 35973070 DOI: 10.1021/acs.jpcb.2c03941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
G protein-coupled receptors (GPCRs) are signaling hubs in cell membranes that regulate a wide range of physiological processes and are popular drug targets. Serotonin1A receptors are important members of the GPCR family and are implicated in neuropsychiatric disorders. Cholesterol is a key constituent of higher eukaryotic membranes and is believed to contribute to the segregated distribution of membrane constituents into domains. To explore the role of cholesterol in lateral dynamics of GPCRs, we utilized single particle tracking (SPT) to monitor diffusion of serotonin1A receptors under acute and chronic cholesterol-depleted conditions. Our results show that the short-term diffusion coefficient of the receptor decreases upon cholesterol depletion, irrespective of the method of cholesterol depletion. Analysis of SPT trajectories revealed that relative populations of receptors undergoing various modes of diffusion change upon cholesterol depletion. Notably, in cholesterol-depleted cells, we observed an increase in the confined population of the receptor accompanied by a reduction in diffusion coefficient for chronic cholesterol depletion. These results are supported by our recent work and present observations that show polymerization of G-actin in response to chronic cholesterol depletion. Taken together, our results bring out the interdependence of cholesterol and actin cytoskeleton in regulating diffusion of GPCRs in membranes.
Collapse
Affiliation(s)
- Sandeep Shrivastava
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Pascal Preira
- Institut de Pharmacologie et de Biologie Structurale, CNRS, Université de Toulouse (UPS), 31 077 Toulouse, France
| | - Laurence Salomé
- Institut de Pharmacologie et de Biologie Structurale, CNRS, Université de Toulouse (UPS), 31 077 Toulouse, France
| | | |
Collapse
|
4
|
Sarkar P, Chattopadhyay A. Statin-induced Increase in Actin Polymerization Modulates GPCR Dynamics and Compartmentalization. Biophys J 2022:S0006-3495(22)00708-1. [DOI: 10.1016/j.bpj.2022.08.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/16/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
|
5
|
Sarkar P, Kumar GA, Shrivastava S, Chattopadhyay A. Chronic cholesterol depletion increases F-actin levels and induces cytoskeletal reorganization via a dual mechanism. J Lipid Res 2022; 63:100206. [PMID: 35390404 PMCID: PMC9096963 DOI: 10.1016/j.jlr.2022.100206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Previous work from us and others has suggested that cholesterol is an important lipid in the context of the organization of the actin cytoskeleton. However, reorganization of the actin cytoskeleton upon modulation of membrane cholesterol is rarely addressed in the literature. In this work, we explored the signaling crosstalk between cholesterol and the actin cytoskeleton by using a high-resolution confocal microscopic approach to quantitatively measure changes in F-actin content upon cholesterol depletion. Our results show that F-actin content significantly increases upon chronic cholesterol depletion, but not during acute cholesterol depletion. In addition, utilizing inhibitors targeting the cholesterol biosynthetic pathway at different steps, we show that reorganization of the actin cytoskeleton could occur due to the synergistic effect of multiple pathways, including prenylated Rho GTPases and availability of membrane phosphatidylinositol 4,5-bisphosphate. These results constitute one of the first comprehensive dissections of the mechanistic basis underlying the interplay between cellular actin levels and cholesterol biosynthesis. We envision these results will be relevant for future understating of the remodeling of the actin cytoskeleton in pathological conditions with altered cholesterol.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
6
|
Dutta A, Mukku RP, Kumar GA, Jafurulla M, Raghunand TR, Chattopadhyay A. Integrity of the Actin Cytoskeleton of Host Macrophages is Necessary for Mycobacterial Entry. J Membr Biol 2022; 255:623-632. [PMID: 35166859 PMCID: PMC8852914 DOI: 10.1007/s00232-022-00217-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/27/2022] [Indexed: 11/16/2022]
Abstract
Macrophages are the primary hosts for Mycobacterium tuberculosis (M. tb), an intracellular pathogen, and the causative organism of tuberculosis (TB) in humans. While M. tb has the ability to enter and survive in host macrophages, the precise mechanism of its internalization, and factors that control this essential process are poorly defined. We have previously demonstrated that perturbations in levels of cholesterol and sphingolipids in macrophages lead to significant reduction in the entry of Mycobacterium smegmatis (M. smegmatis), a surrogate model for mycobacterial internalization, signifying a role for these plasma membrane lipids in interactions at the host–pathogen interface. In this work, we investigated the role of the host actin cytoskeleton, a critical protein framework underlying the plasma membrane, in the entry of M. smegmatis into human macrophages. Our results show that cytochalasin D mediated destabilization of the actin cytoskeleton of host macrophages results in a dose-dependent reduction in the entry of mycobacteria. Notably, the internalization of Escherichia coli remained invariant upon actin destabilization of host cells, implying a specific involvement of the actin cytoskeleton in mycobacterial infection. By monitoring the F-actin content of macrophages utilizing a quantitative confocal microscopy-based technique, we observed a close correlation between the entry of mycobacteria into host macrophages with cellular F-actin content. Our results constitute the first quantitative analysis of the role of the actin cytoskeleton of human macrophages in the entry of mycobacteria, and highlight actin-mediated mycobacterial entry as a potential target for future anti-TB therapeutics.
Collapse
Affiliation(s)
- Aritri Dutta
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Ravi Prasad Mukku
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Md Jafurulla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Tirumalai R Raghunand
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India.
| | | |
Collapse
|
7
|
Proteomic Analysis of Leishmania donovani Membrane Components Reveals the Role of Activated Protein C Kinase in Host-Parasite Interaction. Pathogens 2021; 10:pathogens10091194. [PMID: 34578226 PMCID: PMC8465321 DOI: 10.3390/pathogens10091194] [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: 04/08/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/05/2022] Open
Abstract
Visceral leishmaniasis (VL), mainly caused by the Leishmania donovani parasitic infection, constitutes a potentially fatal disease, for which treatment is primarily dependent on chemotherapy. The emergence of a resistant parasite towards current antileishmanial agents and increasing reports of relapses are the major concerns. Detailed research on the molecular interaction at the host-parasite interface may provide the identification of the parasite and the host-related factors operating during disease development. Genomic and proteomic studies highlighted several essential secretory and cytosolic proteins that play vital roles during Leishmania pathogenesis. The aim of this study was to identify membrane proteins from the Leishmania donovani parasite and the host macrophage that interact with each other using 2-DE/MALDI-TOF/MS. We identified membrane proteins including activated protein C kinase, peroxidoxin, small myristoylated protein 1 (SMP-1), and cytochrome C oxidase from the parasite, while identifying filamin A interacting protein 1(FILIP1) and β-actin from macrophages. We further investigated parasite replication and persistence within macrophages following the macrophage-amastigote model in the presence or absence of withaferin (WA), an inhibitor of activated C kinase. WA significantly reduced Leishmania donovani replication within host macrophages. This study sheds light on the important interacting proteins for parasite proliferation and virulence, and the establishment of infection within host cells, which can be targeted further to develop a strategy for chemotherapeutic intervention.
Collapse
|
8
|
Sharma A, Kumar GA, Chattopadhyay A. Late endosomal/lysosomal accumulation of a neurotransmitter receptor in a cellular model of Smith-Lemli-Opitz syndrome. Traffic 2021; 22:332-344. [PMID: 34418249 DOI: 10.1111/tra.12811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022]
Abstract
Smith-Lemli-Opitz syndrome (SLOS) is a congenital and developmental malformation syndrome associated with defective cholesterol biosynthesis. It is characterized by accumulation of 7-dehydrocholesterol (the immediate biosynthetic precursor of cholesterol in the Kandutsch-Russell pathway) and an altered cholesterol to total sterol ratio. Because SLOS is associated with neurological malfunction, exploring the function and trafficking of neuronal receptors and their interaction with membrane lipids under these conditions assume significance. In this work, we generated a cellular model of SLOS in HEK-293 cells stably expressing the human serotonin1A receptor (an important neurotransmitter G-protein coupled receptor) using AY 9944, an inhibitor for the enzyme 3β-hydroxy-steroid-∆7 -reductase (7-DHCR). Using a quantitative flow cytometry based assay, we show that the plasma membrane population of serotonin1A receptors was considerably reduced under these conditions without any change in total cellular expression of the receptor. Interestingly, the receptors were trafficked to sterol-enriched LysoTracker positive compartments, which accumulated under these conditions. To the best of our knowledge, our results constitute one of the first reports demonstrating intracellular accumulation and misregulated traffic of a neurotransmitter GPCR in SLOS-like conditions. We believe these results assume relevance in our overall understanding of the molecular basis underlying the functional relevance of neurotransmitter receptors in SLOS.
Collapse
Affiliation(s)
- Ashwani Sharma
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
9
|
Kumar A, Sarkar P, Chattopadhyay A. Metabolic Depletion of Sphingolipids Reduces Cell Surface Population of the Human Serotonin 1A Receptor due to Impaired Trafficking. ACS Chem Neurosci 2021; 12:1189-1196. [PMID: 33760584 DOI: 10.1021/acschemneuro.1c00017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sphingolipids and their metabolites are increasingly implicated in the pathogenesis of many metabolic and neurological diseases. It has been postulated that sphingolipids coalesce with cholesterol to form laterally segregated lipid domains that are involved in protein sorting and trafficking. In this work, we have explored the effect of metabolic depletion of sphingolipids on cell surface expression of the human serotonin1A receptor, a neurotransmitter G protein-coupled receptor. We used fumonisin B1 (FB1), a fungal mycotoxin, to inhibit sphingolipid biosynthesis in HEK-293 cells stably expressing the human serotonin1A receptor. Our results obtained using flow cytometric analysis and confocal microscopic imaging show that the cell surface population of the serotonin1A receptor is reduced under sphingolipid-depleted condition. Western blot analysis confirmed that there was no significant difference in total cellular level of the serotonin1A receptor upon depletion of sphingolipids. Interestingly, the effect of FB1 on serotonin1A receptor population was reversed upon replenishment with sphingolipids. These results indicate that sphingolipid depletion does not alter total cellular receptor levels, but impairs serotonin1A receptor trafficking to the cellular plasma membrane. These results could provide mechanistic insights into the role of sphingolipids in modulation of neurotransmitter receptor signaling and trafficking in health and disease.
Collapse
Affiliation(s)
- Abhishek Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
- Amity Institute of Biotechnology, Amity University Haryana, Amity Education Valley, Gurugram 122 413, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
10
|
Pimentel PMDO, de Assis DRR, Gualdrón-Lopez M, Barroso A, Brant F, Leite PG, de Lima Oliveira BC, Esper L, McKinnie SMK, Vederas JC, do Nascimento Cordeiro M, Dos Reis PVM, Teixeira MM, de Castro Pimenta AM, Borges MH, de Lima ME, Machado FS. Tityus serrulatus scorpion venom as a potential drug source for Chagas' disease: Trypanocidal and immunomodulatory activity. Clin Immunol 2021; 226:108713. [PMID: 33711450 DOI: 10.1016/j.clim.2021.108713] [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: 08/19/2020] [Revised: 01/19/2021] [Accepted: 03/06/2021] [Indexed: 11/19/2022]
Abstract
Current chemical therapies for Chagas Disease (CD) lack ability to clear Trypanosoma cruzi (Tc) parasites and cause severe side effects, making search for new strategies extremely necessary. We evaluated the action of Tityus serrulatus venom (TsV) components during Tc infection. TsV treatment increased nitric oxide and pro-inflammatory cytokine production by Tc-infected macrophages (MØ), decreased intracellular parasite replication and trypomastigotes release, also triggering ERK1/2, JNK1/2 and p38 activation. Ts7 demonstrated the highest anti-Tc activity, inducing high levels of TNF and IL-6 in infected MØ. TsV/Ts7 presented synergistic effect on p38 activation when incubated with Tc antigen. KPP-treatment of MØ also decreased trypomastigotes releasing, partially due to p38 activation. TsV/Ts7-pre-incubation of Tc demonstrated a direct effect on parasite decreasing MØ-trypomastigotes releasing. In vivo KPP-treatment of Tc-infected mice resulted in decreased parasitemia. Summarizing, this study opens perspectives for new bioactive molecules as CD-therapeutic treatment, demonstrating the TsV/Ts7/KPP-trypanocidal and immunomodulatory activity during Tc infection.
Collapse
Affiliation(s)
| | - Diego Rodney Rodrigues de Assis
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Melisa Gualdrón-Lopez
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Andréia Barroso
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fátima Brant
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paulo Gaio Leite
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Cabral de Lima Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lisia Esper
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Shaun M K McKinnie
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada; Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA, USA
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Pablo Victor Mendes Dos Reis
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adriano Monteiro de Castro Pimenta
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Maria Elena de Lima
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Santa Casa BH: Instituto de Ensino e Pesquisa, Belo Horizonte, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Science, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
11
|
Kumar GA, Chattopadhyay A. Membrane cholesterol regulates endocytosis and trafficking of the serotonin 1A receptor: Insights from acute cholesterol depletion. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158882. [PMID: 33429076 DOI: 10.1016/j.bbalip.2021.158882] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/24/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis and intracellular trafficking constitute important regulatory features associated with G protein-coupled receptor (GPCR) function. GPCR endocytosis involves several remodeling events at the plasma membrane orchestrated by a concerted interplay of a large number of proteins and membrane lipids. Although considerable literature exists on the protein framework underlying GPCR endocytosis, the role of membrane lipids in this process remains largely unexplored. In order to explore the role of membrane cholesterol (an essential and important lipid in higher eukaryotes) in GPCR endocytosis, we monitored the effect of acute cholesterol depletion using methyl-β-cyclodextrin (MβCD) on endocytosis and intracellular trafficking of the serotonin1A receptor, an important neurotransmitter GPCR. Our results show that the serotonin1A receptor exhibits agonist-induced clathrin-mediated endocytosis with a concentration-dependent inhibition in internalization with increasing concentrations of MβCD, which was restored upon cholesterol replenishment. Interestingly, subsequent to internalization under these conditions, serotonin1A receptors were re-routed toward lysosomal degradation, instead of endosomal recycling observed under normal conditions, thereby implicating membrane cholesterol in modulation of intracellular trafficking of the receptor. This raises the possibility of a novel cholesterol-dependent role of intracellular sorting proteins in GPCR trafficking. These results differ from our previous observations on the endocytosis of the serotonin1A receptor upon statin-induced chronic cholesterol depletion, in terms of endocytic pathway. We conclude that analysis of complex cellular trafficking events such as GPCR endocytosis under acute and chronic cholesterol depletion conditions should be carried out with caution due to fundamental differences underlying these processes.
Collapse
Affiliation(s)
- G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
12
|
Mishra M, Dadhich R, Mogha P, Kapoor S. Mycobacterium Lipids Modulate Host Cell Membrane Mechanics, Lipid Diffusivity, and Cytoskeleton in a Virulence-Selective Manner. ACS Infect Dis 2020; 6:2386-2399. [PMID: 32786287 DOI: 10.1021/acsinfecdis.0c00128] [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] [Indexed: 12/20/2022]
Abstract
Microbial lipids play a critical role in the pathogenesis of infectious diseases by modulating the host cell membrane properties, including lipid/protein diffusion and membrane organization. Mycobacterium tuberculosis (Mtb) synthesizes various chemically distinct lipids that are exposed on its outer membrane and interact with host cell membranes. However, the effects of the structurally diverse Mtb lipids on the host cell membrane properties to fine-tune the host cellular response remain unknown. In this study, we employed membrane biophysics and cell biology to assess the effects of different Mtb lipids on cell membrane mechanics, lipid diffusion, and the cytoskeleton of THP-1 macrophages. We found that Mtb lipids modulate macrophage membrane properties, actin cytoskeleton, and biochemical processes, such as protein phosphorylation and lipid peroxidation, in a virulence lipid-selective manner. These results emphasize that Mtb can fine-tune its interactions with the host cells governed by modulating the lipid profile on its surface. These observations provide a novel lipid-centric paradigm of Mtb pathogenesis that is amenable to pharmacological inhibition and could promote the development of robust biomarkers of Mtb infection and pathogenesis.
Collapse
Affiliation(s)
- Manjari Mishra
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Pankaj Mogha
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
13
|
Horta MF, Andrade LO, Martins-Duarte ÉS, Castro-Gomes T. Cell invasion by intracellular parasites - the many roads to infection. J Cell Sci 2020; 133:133/4/jcs232488. [PMID: 32079731 DOI: 10.1242/jcs.232488] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracellular parasites from the genera Toxoplasma, Plasmodium, Trypanosoma, Leishmania and from the phylum Microsporidia are, respectively, the causative agents of toxoplasmosis, malaria, Chagas disease, leishmaniasis and microsporidiosis, illnesses that kill millions of people around the globe. Crossing the host cell plasma membrane (PM) is an obstacle these parasites must overcome to establish themselves intracellularly and so cause diseases. The mechanisms of cell invasion are quite diverse and include (1) formation of moving junctions that drive parasites into host cells, as for the protozoans Toxoplasma gondii and Plasmodium spp., (2) subversion of endocytic pathways used by the host cell to repair PM, as for Trypanosoma cruzi and Leishmania, (3) induction of phagocytosis as for Leishmania or (4) endocytosis of parasites induced by specialized structures, such as the polar tubes present in microsporidian species. Understanding the early steps of cell entry is essential for the development of vaccines and drugs for the prevention or treatment of these diseases, and thus enormous research efforts have been made to unveil their underlying biological mechanisms. This Review will focus on these mechanisms and the factors involved, with an emphasis on the recent insights into the cell biology of invasion by these pathogens.
Collapse
Affiliation(s)
- Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Luciana Oliveira Andrade
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Érica Santos Martins-Duarte
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
14
|
Karmakar J, Roy S, Mandal C. Modulation of TLR4 Sialylation Mediated by a Sialidase Neu1 and Impairment of Its Signaling in Leishmania donovani Infected Macrophages. Front Immunol 2019; 10:2360. [PMID: 31649671 PMCID: PMC6794462 DOI: 10.3389/fimmu.2019.02360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 09/19/2019] [Indexed: 12/12/2022] Open
Abstract
Altered sialylation is generally maintained by a fine balance between sialidases and sialyltransferases, which plays an essential role during disease pathogenesis. TLR4 is a membrane-bound highly sialylated glycoprotein predominantly having α2,3-linked sialic acids. It is one of the most important client molecules in the anti-leishmanial innate immune arm. Here, we initiated a comprehensive study on the modulation of TLR4 sialylation in Leishmania donovani (L. d)-infected macrophages by a mammalian sialidase/neuraminidase-1 (Neu1) having substrate specificity toward α2,3-linked sialic acids. We observed reduced membrane-associated Neu1 with its decreased enzyme activity in infected macrophages. Moreover, we demonstrated reduced association of Neu1 with TLR4 leading to enhanced sialylation of TLR4 in these infected cells. Conversely, Neu1 over expression exhibited enhanced association of TLR4 with Neu1 leading to reduced sialylation which possibly linked to increased association of TLR4 with its downstream adaptor protein, MyD88. This, in turn, activated downstream MAP kinase signaling pathway, with enhanced nuclear translocation of NFκB that resulted in increased genetic and protein levels expression of Th1 cytokines and effector molecule nitric oxide secretion which ultimately leads to reduced parasite burden in macrophages. This was further validated by Neu1 silencing in infected macrophages which reversed such a situation. Such events strongly confirm the importance of Neu1 in modulation of TLR4 sialylation during parasite infection resulting in impairment of innate immune response. Furthermore, decreased membrane-bound Neu1 in infected macrophages could be attributed to its reduced tyrosine-phosphorylation as well as diminished association with cathepsin A. Both these phenomenon possibly play significant roles in inhibiting translocation of the sialidase from cytosol to membrane. Taken together, our study first time demonstrated impaired translocation of cytosolic Neu1 to the membrane of L. donovani-infected macrophages due to impaired phosphorylation of this enzyme. This novel finding establishes a link between enhanced α2,3-linked sialic acids on TLR4 and reduced membrane-bound Neu1 which plays a significant role for inhibiting downstream signaling to establish successful infection in the host cells.
Collapse
Affiliation(s)
- Joyshree Karmakar
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Saptarshi Roy
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
15
|
Mishra M, Adhyapak P, Dadhich R, Kapoor S. Dynamic Remodeling of the Host Cell Membrane by Virulent Mycobacterial Sulfoglycolipid-1. Sci Rep 2019; 9:12844. [PMID: 31492926 PMCID: PMC6731295 DOI: 10.1038/s41598-019-49343-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 08/23/2019] [Indexed: 12/31/2022] Open
Abstract
Lipids dictate membrane properties to modulate lateral membrane organization, lipid/protein diffusion and lipid-protein interactions, thereby underpinning proper functioning of cells. Mycobacterium tuberculosis harnesses the power of its atypical cell wall lipids to impact immune surveillance machinery centered at the host cell membrane. However, the role of specific virulent lipids in altering host cellular functions by modulating membrane organization and the associated signaling response are still pertinent unresolved questions. Here, combining membrane biophysics and cell biology, we elucidate how virulent Mtb sulfoglycolipids hijack the host cell membrane, affecting its order, fluidity, and stiffness along with manipulating the linked cytoskeleton. The functional outcome of this perturbation was assayed by monitoring membrane-associated autophagy signaling. These actions form a part of the overall response to commandeer host membrane-associated immune processes during infection. The findings on the mechanism of action of Mtb lipids on host cell membrane structure and downstream signaling will deepen the collective understanding of their functional aspects in membrane-dictated bacterial survival, pathogenesis and drug resistance and reveal suitable membrane driven-therapeutic intervention points and diagnostic tools.
Collapse
Affiliation(s)
- Manjari Mishra
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India
| | - Pranav Adhyapak
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India
| | - Ruchika Dadhich
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, India.
| |
Collapse
|
16
|
Shrivastava S, Sarkar P, Preira P, Salomé L, Chattopadhyay A. Role of Actin Cytoskeleton in Dynamics and Function of the Serotonin 1A Receptor. Biophys J 2019; 118:944-956. [PMID: 31606121 DOI: 10.1016/j.bpj.2019.08.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/13/2019] [Accepted: 08/29/2019] [Indexed: 12/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are important membrane proteins in higher eukaryotes that carry out a vast array of cellular signaling and act as major drug targets. The serotonin1A receptor is a prototypical member of the GPCR family and is implicated in neuropsychiatric disorders such as anxiety and depression, besides serving as an important drug target. With an overall goal of exploring the functional consequence of altered receptor dynamics, in this work, we probed the role of the actin cytoskeleton in the dynamics, ligand binding, and signaling of the serotonin1A receptor. We monitored receptor dynamics utilizing single particle tracking, which provides information on relative distribution of receptors in various diffusion modes in addition to diffusion coefficient. We show here that the short-term diffusion coefficient of the receptor increases upon actin destabilization by cytochalasin D. In addition, analysis of individual trajectories shows that there are changes in relative populations of receptors undergoing various types of diffusion upon actin destabilization. The release of dynamic constraint was evident by an increase in the radius of confinement of the receptor upon actin destabilization. The functional implication of such actin destabilization was manifested as an increase in specific agonist binding and downstream signaling, monitored by measuring reduction in cellular cAMP levels. These results bring out the interdependence of GPCR dynamics with cellular signaling.
Collapse
Affiliation(s)
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Pascal Preira
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Laurence Salomé
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.
| | | |
Collapse
|
17
|
Kumar GA, Karmakar J, Mandal C, Chattopadhyay A. Leishmania donovani Internalizes into Host Cells via Caveolin-mediated Endocytosis. Sci Rep 2019; 9:12636. [PMID: 31477757 PMCID: PMC6718660 DOI: 10.1038/s41598-019-49007-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 08/19/2019] [Indexed: 02/08/2023] Open
Abstract
Leishmania donovani is an intracellular protozoan parasite that causes visceral leishmaniasis, a major cause of mortality and morbidity worldwide. The host plasma membrane serves as the portal of entry for Leishmania to gain access to the cellular interior. Although several host cell membrane receptors have been shown to be involved in the entry of Leishmania donovani into host cells, the endocytic pathway involved in the internalization of the parasite is not known. In this work, we explored the endocytic pathway involved in the entry of Leishmania donovani into host macrophages, utilizing specific inhibitors against two major pathways of internalization, i.e., clathrin- and caveolin-mediated endocytosis. We show that pitstop 2, an inhibitor for clathrin-mediated endocytosis, does not affect the entry of Leishmania donovani promastigotes into host macrophages. Interestingly, a significant reduction in internalization was observed upon treatment with genistein, an inhibitor for caveolin-mediated endocytosis. These results are supported by a similar trend in intracellular amastigote load within host macrophages. These results suggest that Leishmania donovani utilizes caveolin-mediated endocytosis to internalize into host cells. Our results provide novel insight into the mechanism of phagocytosis of Leishmania donovani into host cells and assume relevance in the development of novel therapeutics against leishmanial infection.
Collapse
Affiliation(s)
- G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500 007, India
| | - Joyshree Karmakar
- CSIR-Indian Institute of Chemical Biology, Raja S.C. Mullick Road, Kolkata, 700 032, India
| | - Chitra Mandal
- CSIR-Indian Institute of Chemical Biology, Raja S.C. Mullick Road, Kolkata, 700 032, India.
| | | |
Collapse
|
18
|
Need for sustainable approaches in antileishmanial drug discovery. Parasitol Res 2019; 118:2743-2752. [DOI: 10.1007/s00436-019-06443-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
|
19
|
Kumar GA, Sarkar P, Jafurulla M, Singh SP, Srinivas G, Pande G, Chattopadhyay A. Exploring Endocytosis and Intracellular Trafficking of the Human Serotonin1A Receptor. Biochemistry 2019; 58:2628-2641. [DOI: 10.1021/acs.biochem.9b00033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- G. Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Md. Jafurulla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Shishu Pal Singh
- National Centre for Biological Sciences, UAS-GKVK Campus, Bellary Road, Bangalore 560 065, India
| | - Gunda Srinivas
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Gopal Pande
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
20
|
Pereira MA, Alexandre-Pires G, Câmara M, Santos M, Martins C, Rodrigues A, Adriana J, Passero LFD, Pereira da Fonseca I, Santos-Gomes G. Canine neutrophils cooperate with macrophages in the early stages of Leishmania infantum in vitro infection. Parasite Immunol 2019; 41:e12617. [PMID: 30735568 DOI: 10.1111/pim.12617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/31/2019] [Indexed: 11/28/2022]
Abstract
Leishmania infantum is the aetiological agent of human visceral leishmaniasis and canine leishmaniasis, both systemic and potentially fatal diseases. Polymorphonuclear neutrophils (PMN) are the first cells to phagocyte this parasite at the inoculation site, but macrophages (MØ) are the definitive host cells, ensuring parasite replication. The interaction between dog MØ, PMN and L infantum promastigotes was in vitro investigated. It was observed that promastigotes establish contact with blood monocyte-derived MØ mainly by the tip of the flagellum. These cells, that efficiently bind and internalize parasites, underwent major morphological changes, produced nitric oxide (NO) and released histone H1 in order to inactivate the parasite. Transfer of intracellular parasites from PMN to MØ was confirmed by flow cytometry, using L infantum expressing a green fluorescent protein. The interaction of MØ with L infantum-infected PMN lead to NO production and release of extracellular traps, which may contribute to parasite containment and inactivation. This study highlights for the first time the diversity of cellular and molecular events triggered by the interaction between canine PMN and MØ, which can promote a reduction of parasite burden in the early phase of L infantum infection.
Collapse
Affiliation(s)
- Maria A Pereira
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisboa, Portugal.,Instituto Politécnico de Portalegre (IPP), Portalegre, Portugal
| | - Graça Alexandre-Pires
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Lisboa, Portugal
| | - Margarida Câmara
- Câmara Municipal de Évora, Serviço Veterinário Municipal, Évora, Portugal
| | - Marcos Santos
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Lisboa, Portugal
| | - Catarina Martins
- CEDOC-Chronic Diseases Research Center, Immunology, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Armanda Rodrigues
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisboa, Portugal
| | - Jéssica Adriana
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, São Paulo, Brazil
| | - Luiz Felipe D Passero
- São Paulo State University (UNESP), Institute of Biosciences, São Paulo, Brazil.,São Paulo State University (UNESP), Institute for Advanced Studies of Ocean, São Paulo, Brazil
| | - Isabel Pereira da Fonseca
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Lisboa, Portugal
| | - Gabriela Santos-Gomes
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisboa, Portugal
| |
Collapse
|
21
|
Shadab M, Das S, Banerjee A, Sinha R, Asad M, Kamran M, Maji M, Jha B, Deepthi M, Kumar M, Tripathi A, Kumar B, Chakrabarti S, Ali N. RNA-Seq Revealed Expression of Many Novel Genes Associated With Leishmania donovani Persistence and Clearance in the Host Macrophage. Front Cell Infect Microbiol 2019; 9:17. [PMID: 30805314 PMCID: PMC6370631 DOI: 10.3389/fcimb.2019.00017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
Host- as well as parasite-specific factors are equally crucial in allowing either the Leishmania parasites to dominate, or host macrophages to resist infection. To identify such factors, we infected murine peritoneal macrophages with either the virulent (vAG83) or the non-virulent (nvAG83) parasites of L. donovani. Then, through dual RNA-seq, we simultaneously elucidated the transcriptomic changes occurring both in the host and the parasites. Through Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the differentially expressed (DE) genes, we showed that the vAG83-infected macrophages exhibit biased anti-inflammatory responses compared to the macrophages infected with the nvAG83. Moreover, the vAG83-infected macrophages displayed suppression of many important cellular processes, including protein synthesis. Further, through protein-protein interaction study, we showed significant downregulation in the expression of many hubs and hub-bottleneck genes in macrophages infected with vAG83 as compared to nvAG83. Cell signaling study showed that these two parasites activated the MAPK and PI3K-AKT signaling pathways differentially in the host cells. Through gene ontology analyses of the parasite-specific genes, we discovered that the genes for virulent factors and parasite survival were significantly upregulated in the intracellular amastigotes of vAG83. In contrast, genes involved in the immune stimulations, and those involved in negative regulation of the cell cycle and transcriptional regulation, were upregulated in the nvAG83. Collectively, these results depicted a differential regulation in the host and the parasite-specific molecules during in vitro persistence and clearance of the parasites.
Collapse
Affiliation(s)
- Mohammad Shadab
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Anindyajit Banerjee
- Structural Biology and Bio-Informatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Roma Sinha
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mohammad Asad
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Mithun Maji
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Baijayanti Jha
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Makaraju Deepthi
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | | | | | - Bipin Kumar
- Nucleome Informatics Pvt. Ltd., Hyderabad, India
| | - Saikat Chakrabarti
- Structural Biology and Bio-Informatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
22
|
Taenia solium glutathione transferase fraction activates macrophages and favors the development of Th1-type response. Biosci Rep 2019; 39:BSR20181132. [PMID: 30538171 PMCID: PMC6340957 DOI: 10.1042/bsr20181132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Abstract
Glutathione (GSH) transferase (GST) is an essential enzyme in cestodes for the detoxification of xenobiotics. In Taenia solium, two GSTs (Ts25GST and Ts26GST kDa) were isolated as a fraction (SGSTF) by GSH-Sepharose-4B. Both are located on the tegument. Immunization assays with SGSTF reduced up to 90% of the parasitic load in a murine model of cysticercosis. It prompted us to investigate how SGSTF induces this protective immune response. To test it, we exposed peritoneal macrophages to SGSTF for 24 h; such exposure favored the production of IL-12, TNF, and IL-10 as well as the expression of nitric oxide synthase 2 inducible (Nos2) and CD86, but did not induce the expression of chitinase-like 3 (Chil3). Confocal microscopy showed that the macrophages internalize the SGSTF which co-localized after 1 h with MHC-II in their plasma membranes. Macrophages exposed to SGSTF and co-cultured with anti-CD3 pre-activated T CD4+ cells, enhanced the proliferation of CD4+ cells, induced high interferon-γ (IFN-γ) secretion, and elevated the expression of CD25 and CD69, molecules associated with cell activation. Similar assay using T CD4+ cells from DO11.10 mice and ovalbumin (OVA) peptide+SGSTF as stimuli, showed enhanced cell proliferation and OVA-specific IFN-γ secretion. These data are in-line with those indicating that the P1, P5, and P6 peptides of Schistosoma japonicum 28GST highly promote T-cell proliferation and Th1 response in vitro. We found that such peptides are also present on Ts25GST and Ts26GST. It suggests that SGSTF activates peritoneal macrophages to a classically activated-like phenotype, and that these macrophages induce the differentiation of T CD4+ cells toward a Th1-type response.
Collapse
|
23
|
Sarkar P, Chattopadhyay A. Exploring membrane organization at varying spatiotemporal resolutions utilizing fluorescence-based approaches: implications in membrane biology. Phys Chem Chem Phys 2019; 21:11554-11563. [DOI: 10.1039/c9cp02087j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Representative experimental approaches based on dynamic fluorescence microscopy to analyze organization and dynamics of membrane lipids and proteins.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology
- Hyderabad 500 007
- India
| | | |
Collapse
|
24
|
Galuppo MK, de Rezende E, Forti FL, Cortez M, Cruz MC, Teixeira AA, Giordano RJ, Stolf BS. CD100/Sema4D Increases Macrophage Infection by Leishmania (Leishmania) amazonensis in a CD72 Dependent Manner. Front Microbiol 2018; 9:1177. [PMID: 29922261 PMCID: PMC5996280 DOI: 10.3389/fmicb.2018.01177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 05/15/2018] [Indexed: 11/13/2022] Open
Abstract
Leishmaniasis is caused by trypanosomatid protozoa of the genus Leishmania, which infect preferentially macrophages. The disease affects 12 million people worldwide, who may present cutaneous, mucocutaneous or visceral forms. Several factors influence the form and severity of the disease, and the main ones are the Leishmania species and the host immune response. CD100 is a membrane bound protein that can also be shed. It was first identified in T lymphocytes and latter shown to be induced in macrophages by inflammatory stimuli. The soluble CD100 (sCD100) reduces migration and expression of inflammatory cytokines in human monocytes and dendritic cells, as well as the intake of oxidized low-density lipoprotein (oxLDL) by human macrophages. Considering the importance of macrophages in Leishmania infection and the potential role of sCD100 in the modulation of macrophage phagocytosis and activation, we analyzed the expression and distribution of CD100 in murine macrophages and the effects of sCD100 on macrophage infection by Leishmania (Leishmania) amazonensis. Here we show that CD100 expression in murine macrophages increases after infection with Leishmania. sCD100 augments infection and phagocytosis of Leishmania (L.) amazonensis promastigotes by macrophages, an effect dependent on macrophage CD72 receptor. Besides, sCD100 enhances phagocytosis of zymosan particles and infection by Trypanosoma cruzi.
Collapse
Affiliation(s)
- Mariana K Galuppo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eloiza de Rezende
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fabio L Forti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Mauro Cortez
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mario C Cruz
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andre A Teixeira
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Ricardo J Giordano
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Beatriz S Stolf
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Cota BB, Tunes LG, Maia DNB, Ramos JP, Oliveira DMD, Kohlhoff M, Alves TMDA, Souza-Fagundes EM, Campos FF, Zani CL. Leishmanicidal compounds of Nectria pseudotrichia, an endophytic fungus isolated from the plant Caesalpinia echinata (Brazilwood). Mem Inst Oswaldo Cruz 2018; 113:102-110. [PMID: 29236928 PMCID: PMC5722265 DOI: 10.1590/0074-02760170217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/22/2017] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In a screen of extracts from plants and fungi to detect antileishmanial activity, we found that the ethyl acetate extract of the fungus Nectria pseudotrichia, isolated from the tree Caesalpinia echinata (Brazilwood), is a promising source of bioactive compounds. OBJECTIVES The aims of this study were to isolate and determine the chemical structures of the compounds responsible for the antileishmanial activity of the organic extract from N. pseudotrichia. METHODS Compounds were isolated by chromatographic fractionation using semi-preparative high-performance liquid chromatography, and their chemical structures were determined by analytical and spectral data and by comparison with published data. The antileishmanial activity of the isolated compounds was evaluated in intracellular amastigote forms of Leishmania (Viannia) braziliensis expressing firefly luciferase as reporter gene, and cytotoxicity was determined in Vero and THP-1 mammalian cell lines by MTT assay. FINDINGS Fractionation of the extract yielded seven compounds: 10-acetyl trichoderonic acid A (1), 6'-acetoxy-piliformic acid (2), 5',6'-dehydropiliformic acid (3), piliformic acid (4), hydroheptelidic acid (5), xylaric acid D (6), and cytochalasin D (7). Compounds 1, 2 and 3 are reported here for the first time. Compounds 1, 2, and 5 were more active, with IC50 values of 21.4, 28.3, and 24.8 µM, respectively, and showed low toxicity to Vero and THP-1 cells. MAIN CONCLUSIONS N. pseudotrichia produces secondary metabolites that are more toxic to intracellular amastigote forms of L. (V.) braziliensis than to mammalian cells.
Collapse
Affiliation(s)
- Betania Barros Cota
- Laboratório de Química de Produtos Naturais Bioativos, Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brasil
| | - Luiza Guimarães Tunes
- Laboratório de Genômica Funcional e Proteômica de Leishmania spp. e Trypanosoma cruzi, Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brasil
| | - Daniela Nabak Bueno Maia
- Laboratório de Química de Produtos Naturais Bioativos, Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brasil
| | - Jonas Pereira Ramos
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | | | - Markus Kohlhoff
- Laboratório de Química de Produtos Naturais Bioativos, Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brasil
| | - Tânia Maria de Almeida Alves
- Laboratório de Química de Produtos Naturais Bioativos, Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brasil
| | | | - Fernanda Fraga Campos
- Departamento de Ciências Biológicas e da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Carlos Leomar Zani
- Laboratório de Química de Produtos Naturais Bioativos, Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brasil
| |
Collapse
|
26
|
Differential Membrane Dipolar Orientation Induced by Acute and Chronic Cholesterol Depletion. Sci Rep 2017; 7:4484. [PMID: 28667339 PMCID: PMC5493612 DOI: 10.1038/s41598-017-04769-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/22/2017] [Indexed: 02/02/2023] Open
Abstract
Cholesterol plays a crucial role in cell membrane organization, dynamics and function. Depletion of cholesterol represents a popular approach to explore cholesterol-sensitivity of membrane proteins. An emerging body of literature shows that the consequence of membrane cholesterol depletion often depends on the actual process (acute or chronic), although the molecular mechanism underlying the difference is not clear. Acute depletion, using cyclodextrin-type carriers, is faster relative to chronic depletion, in which inhibitors of cholesterol biosynthesis are used. With the overall goal of addressing molecular differences underlying these processes, we monitored membrane dipole potential under conditions of acute and chronic cholesterol depletion in CHO-K1 cells, using a voltage-sensitive fluorescent dye in dual wavelength ratiometric mode. Our results show that the observed membrane dipole potential exhibits difference under acute and chronic cholesterol depletion conditions, even when cholesterol content was identical. To the best of our knowledge, these results provide, for the first time, molecular insight highlighting differences in dipolar reorganization in these processes. A comprehensive understanding of processes in which membrane cholesterol gets modulated would provide novel insight in its interaction with membrane proteins and receptors, thereby allowing us to understand the role of cholesterol in cellular physiology associated with health and disease.
Collapse
|
27
|
Roy S, Dutta D, Satyavarapu EM, Yadav PK, Mandal C, Kar S, Mandal C. Mahanine exerts in vitro and in vivo antileishmanial activity by modulation of redox homeostasis. Sci Rep 2017; 7:4141. [PMID: 28646156 PMCID: PMC5482887 DOI: 10.1038/s41598-017-03943-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/12/2017] [Indexed: 11/09/2022] Open
Abstract
Earlier we have established a carbazole alkaloid (mahanine) isolated from an Indian edible medicinal plant as an anticancer agent with minimal effect on normal cells. Here we report for the first time that mahanine-treated drug resistant and sensitive virulent Leishmania donovani promastigotes underwent apoptosis through phosphatidylserine externalization, DNA fragmentation and cell cycle arrest. An early induction of reactive oxygen species (ROS) suggests that the mahanine-induced apoptosis was mediated by oxidative stress. Additionally, mahanine-treated Leishmania-infected macrophages exhibited anti-amastigote activity by nitric oxide (NO)/ROS generation along with suppression of uncoupling protein 2 and Th1-biased cytokines response through modulating STAT pathway. Moreover, we have demonstrated the interaction of a few antioxidant enzymes present in parasite with mahanine through molecular modeling. Reduced genetic and protein level expression of one such enzyme namely ascorbate peroxidase was also observed in mahanine-treated promastigotes. Furthermore, oral administration of mahanine in acute murine model exhibited almost complete reduction of parasite burden, upregulation of NO/iNOS/ROS/IL-12 and T cell proliferation. Taken together, we have established a new function of mahanine as a potent antileishmanial molecule, capable of inducing ROS and exploit antioxidant enzymes in parasite along with modulation of host's immune response which could be developed as an inexpensive and nontoxic therapeutics either alone or in combination.
Collapse
Affiliation(s)
- Saptarshi Roy
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Devawati Dutta
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Eswara M Satyavarapu
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| | - Pawan K Yadav
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, 226001, India
| | - Chhabinath Mandal
- National Institute of Pharmaceutical Education and Research, Kolkata, 4, Raja S. C. Mullick Road, Kolkata, 700032, India
| | - Susanta Kar
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, 226001, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India.
| |
Collapse
|
28
|
Gallic and ellagic acids: two natural immunomodulator compounds solve infection of macrophages by Leishmania major. Naunyn Schmiedebergs Arch Pharmacol 2017. [DOI: 10.1007/s00210-017-1387-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
29
|
Roy S, Mandal C. Leishmania donovani Utilize Sialic Acids for Binding and Phagocytosis in the Macrophages through Selective Utilization of Siglecs and Impair the Innate Immune Arm. PLoS Negl Trop Dis 2016; 10:e0004904. [PMID: 27494323 PMCID: PMC4975436 DOI: 10.1371/journal.pntd.0004904] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 07/14/2016] [Indexed: 12/15/2022] Open
Abstract
Background Leishmania donovani, belonging to a unicellular protozoan parasite, display the differential level of linkage-specific sialic acids on their surface. Sialic acids binding immunoglobulin-like lectins (siglecs) are a class of membrane-bound receptors present in the haematopoetic cell lineages interact with the linkage-specific sialic acids. Here we aimed to explore the utilization of sialic acids by Leishmania donovani for siglec-mediated binding, phagocytosis, modulation of innate immune response and signaling pathways for establishment of successful infection in the host. Methodology/Principle Findings We have found enhanced binding of high sialic acids containing virulent strains (AG83+Sias) with siglec-1 and siglec-5 present on macrophages compared to sialidase treated AG83+Sias (AG83-Sias) and low sialic acids-containing avirulent strain (UR6) by flow cytometry. This specific receptor-ligand interaction between sialic acids and siglecs were further confirmed by confocal microscopy. Sialic acids-siglec-1-mediated interaction of AG83+Sias with macrophages induced enhanced phagocytosis. Additionally, sialic acids-siglec-5 interaction demonstrated reduced ROS, NO generation and Th2 dominant cytokine response upon infection with AG83+Sias in contrast to AG83-Sias and UR6. Sialic acids-siglecs binding also facilitated multiplication of intracellular amastigotes. Moreover, AG83+Sias induced sialic acids-siglec-5-mediated upregulation of host phosphatase SHP-1. Such sialic acids-siglec interaction was responsible for further downregulation of MAPKs (p38, ERK and JNK) and PI3K/Akt pathways followed by the reduced translocation of p65 subunit of NF-κβ to the nucleus from cytosol in the downstream signaling pathways. This sequence of events was reversed in AG83-Sias and UR6-infected macrophages. Besides, siglec-knockdown macrophages also showed the reversal of AG83+Sias infection-induced effector functions and downstream signaling events. Conclusions/Significances Taken together, this study demonstrated that virulent parasite (AG83+Sias) establish a unique sialic acids-mediated binding and subsequent phagocytosis in the host cell through the selective exploitation of siglec-1. Additionally, sialic acids-siglec-5 interaction altered the downstream signaling pathways which contributed impairment of immune effector functions of macrophages. To the best of our knowledge, this is a comprehensive report describing sialic acids-siglec interactions and their role in facilitating uptake of the virulent parasite within the host. Sialic acids are nine carbon sugars present on terminal cell surface glycoproteins and glycolipids. Siglec is a membrane receptor that belongs to an immunoglobulin super family present in almost all the haematopoetic cell lineages. There are 14 different types of siglecs present on human immune cells that take an active part in balancing the magnitude of immunological reactions. In general, these siglecs bind with sialic acids and negatively regulate the immune response. Leishmania contains sialic acids on its surface. Virulent parasites utilize this sugar to bind with macrophages through siglec-1 and siglec-5 compared to low sialic acids containing avirulent parasites. Such sialic acids-siglec-mediated interactions exhibited a suppressed host immune response which helped them to establish successful infection compared to desialylated virulent and avirulent parasites, as well as, siglec-depleted macrophages. Interestingly, interaction between sialic acids and siglec-1 induced enhanced phagocytosis, while sialic acids-siglec-5 interaction upregulated the phosphatase SHP-1. This interaction with the virulent strain exhibited deactivation of various downstream signaling pathways and ultimately controlled translocation of a functional component of transcription factor NF-κβ for regulation of cytokines and other effector molecules in infected macrophages. Thus, the interaction between the parasite and the host cells through sialic acids-siglec binding is clearly a newly identified mechanism by which parasites can establish successful infection by subverting the host’s innate immune response.
Collapse
Affiliation(s)
- Saptarshi Roy
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- * E-mail:
| |
Collapse
|
30
|
Kumar GA, Roy S, Jafurulla M, Mandal C, Chattopadhyay A. Statin-induced chronic cholesterol depletion inhibits Leishmania donovani infection: Relevance of optimum host membrane cholesterol. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2088-2096. [PMID: 27319380 DOI: 10.1016/j.bbamem.2016.06.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 06/08/2016] [Accepted: 06/15/2016] [Indexed: 12/15/2022]
Abstract
Leishmania are obligate intracellular protozoan parasites that invade and survive within host macrophages leading to leishmaniasis, a major cause of mortality and morbidity worldwide, particularly among economically weaker sections in tropical and subtropical regions. Visceral leishmaniasis is a potent disease caused by Leishmania donovani. The detailed mechanism of internalization of Leishmania is poorly understood. A basic step in the entry of Leishmania involves interaction of the parasite with the host plasma membrane. In this work, we have explored the effect of chronic metabolic cholesterol depletion using lovastatin on the entry and survival of Leishmania donovani in host macrophages. We show here that chronic cholesterol depletion of host macrophages results in reduction in the attachment of Leishmania promastigotes, along with a concomitant reduction in the intracellular amastigote load. These results assume further relevance since chronic cholesterol depletion is believed to mimic physiological cholesterol modulation. Interestingly, the reduction in the ability of Leishmania to enter host macrophages could be reversed upon metabolic replenishment of cholesterol. Importantly, enrichment of host membrane cholesterol resulted in reduction in the entry and survival of Leishmania in host macrophages. As a control, the binding of Escherichia coli to host macrophages remained invariant under these conditions, thereby implying specificity of cholesterol requirement for effective leishmanial infection. To the best of our knowledge, these results constitute the first comprehensive demonstration that an optimum content of host membrane cholesterol is necessary for leishmanial infection. Our results assume relevance in the context of developing novel therapeutic strategies targeting cholesterol-mediated leishmanial infection.
Collapse
Affiliation(s)
- G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Saptarshi Roy
- CSIR-Indian Institute of Chemical Biology, Raja S.C. Mullick Road, Kolkata 700 032, India
| | - Md Jafurulla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Chitra Mandal
- CSIR-Indian Institute of Chemical Biology, Raja S.C. Mullick Road, Kolkata 700 032, India.
| | | |
Collapse
|
31
|
Richter E, Ventz K, Harms M, Mostertz J, Hochgräfe F. Induction of Macrophage Function in Human THP-1 Cells Is Associated with Rewiring of MAPK Signaling and Activation of MAP3K7 (TAK1) Protein Kinase. Front Cell Dev Biol 2016; 4:21. [PMID: 27066479 PMCID: PMC4811913 DOI: 10.3389/fcell.2016.00021] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022] Open
Abstract
Macrophages represent the primary human host response to pathogen infection and link the immediate defense to the adaptive immune system. Mature tissue macrophages convert from circulating monocyte precursor cells by terminal differentiation in a process that is not fully understood. Here, we analyzed the protein kinases of the human monocytic cell line THP-1 before and after induction of macrophage differentiation by using kinomics and phosphoproteomics. When comparing the macrophage-like state with the monocytic precursor, 50% of the kinome was altered in expression and even 71% of covered kinase phosphorylation sites were affected. Kinome rearrangements are for example characterized by a shift of overrepresented cyclin-dependent kinases associated with cell cycle control in monocytes to calmodulin-dependent kinases and kinases involved in proinflammatory signaling. Eventually, we show that monocyte-to-macrophage differentiation is associated with major rewiring of mitogen-activated protein kinase signaling networks and demonstrate that protein kinase MAP3K7 (TAK1) acts as the key signaling hub in bacterial killing, chemokine production and differentiation. Our study proves the fundamental role of protein kinases and cellular signaling as major drivers of macrophage differentiation and function. The finding that MAP3K7 is central to macrophage function suggests MAP3K7 and its networking partners as promising targets in host-directed therapy for macrophage-associated disease.
Collapse
Affiliation(s)
- Erik Richter
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Katharina Ventz
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Manuela Harms
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Jörg Mostertz
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| | - Falko Hochgräfe
- Junior Research Group Pathoproteomics, Competence Center Functional Genomics, University of Greifswald Greifswald, Germany
| |
Collapse
|
32
|
Kumar GA, Jafurulla M, Chattopadhyay A. The membrane as the gatekeeper of infection: Cholesterol in host-pathogen interaction. Chem Phys Lipids 2016; 199:179-185. [PMID: 26902688 DOI: 10.1016/j.chemphyslip.2016.02.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 02/13/2016] [Accepted: 02/15/2016] [Indexed: 11/30/2022]
Abstract
The cellular plasma membrane serves as a portal for the entry of intracellular pathogens. An essential step for an intracellular pathogen to gain entry into a host cell therefore is to be able to cross the cell membrane. In this review, we highlight the role of host membrane cholesterol in regulating the entry of intracellular pathogens using insights obtained from work on the interaction of Leishmania and Mycobacterium with host cells. The entry of these pathogens is known to be dependent on host membrane cholesterol. Importantly, pathogen entry is inhibited either upon depletion (or complexation), or enrichment of membrane cholesterol. In other words, an optimum level of host membrane cholesterol is necessary for efficient infection by pathogens. In this overall context, we propose a general mechanism, based on cholesterol-induced conformational changes, involving cholesterol binding sites in host cell surface receptors that are implicated in this process. A therapeutic strategy targeting modulation of membrane cholesterol would have the advantage of avoiding the commonly encountered problem of drug resistance in tackling infection by intracellular pathogens. Insights into the role of host membrane cholesterol in pathogen entry would be instrumental in the development of novel therapeutic strategies to effectively tackle intracellular pathogenesis.
Collapse
Affiliation(s)
- G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Md Jafurulla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
33
|
Liévin-Le Moal V, Loiseau PM. Leishmania hijacking of the macrophage intracellular compartments. FEBS J 2015; 283:598-607. [PMID: 26588037 DOI: 10.1111/febs.13601] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/08/2015] [Accepted: 11/13/2015] [Indexed: 12/15/2022]
Abstract
Leishmania spp., transmitted to humans by the bite of the sandfly vector, are responsible for the three major forms of leishmaniasis, cutaneous, diffuse mucocutaneous and visceral. Leishmania spp. interact with membrane receptors of neutrophils and macrophages. In macrophages, the parasite is internalized within a parasitophorous vacuole and engages in a particular intracellular lifestyle in which the flagellated, motile Leishmania promastigote metacyclic form differentiates into non-motile, metacyclic amastigote form. This phenomenon is induced by Leishmania-triggered events leading to the fusion of the parasitophorous vacuole with vesicular members of the host cell endocytic pathway including recycling endosomes, late endosomes and the endoplasmic reticulum. Maturation of the parasitophorous vacuole leads to the intracellular proliferation of the Leishmania amastigote forms by acquisition of host cell nutrients while escaping host defense responses.
Collapse
Affiliation(s)
- Vanessa Liévin-Le Moal
- Anti-Parasitic Chemotherapy, Faculté de Pharmacie, CNRS, UMR 8076 BioCIS, Châtenay-Malabry, France.,Université Paris-Sud, Orsay, France.,Faculté de Pharmacie, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LabEx LERMIT), Châtenay-Malabry, France
| | - Philippe M Loiseau
- Anti-Parasitic Chemotherapy, Faculté de Pharmacie, CNRS, UMR 8076 BioCIS, Châtenay-Malabry, France.,Université Paris-Sud, Orsay, France.,Faculté de Pharmacie, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LabEx LERMIT), Châtenay-Malabry, France
| |
Collapse
|
34
|
Viswanathan G, Jafurulla M, Kumar GA, Raghunand TR, Chattopadhyay A. Dissecting the membrane cholesterol requirement for mycobacterial entry into host cells. Chem Phys Lipids 2015; 189:19-27. [DOI: 10.1016/j.chemphyslip.2015.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/19/2015] [Accepted: 05/21/2015] [Indexed: 12/28/2022]
|
35
|
Chattopadhyay A, Rao BD, Jafurulla M. Solubilization of G Protein-Coupled Receptors. Methods Enzymol 2015; 557:117-34. [DOI: 10.1016/bs.mie.2015.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
36
|
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: 11.7] [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
|
37
|
Abstract
G protein-coupled receptors (GPCRs) are the largest class of molecules involved in signal transduction across cell membranes and represent major targets in the development of novel drug candidates in all clinical areas. Although there have been some recent leads, structural information on GPCRs is relatively rare due to the difficulty associated with crystallization. A specific reason for this is the intrinsic flexibility displayed by GPCRs, which is necessary for their functional diversity. Since GPCRs are integral membrane proteins, interaction of membrane lipids with them constitutes an important area of research in GPCR biology. In particular, membrane cholesterol has been reported to have a modulatory role in the function of a number of GPCRs. The role of membrane cholesterol in GPCR function is discussed with specific example of the serotonin1A receptor. Recent results show that GPCRs are characterized with structural motifs that preferentially associate with cholesterol. An emerging and important concept is oligomerization of GPCRs and its role in GPCR function and signaling. The role of membrane cholesterol in GPCR oligomerization is highlighted. Future research in GPCR biology would offer novel insight in basic biology and provide new avenues for drug discovery.
Collapse
|