1
|
Chauhan R, Tiwari M, Chaudhary A, Sharan Thakur R, Pande V, Das J. Chemokines: A key driver for inflammation in protozoan infection. Int Rev Immunol 2023; 43:211-228. [PMID: 37980574 DOI: 10.1080/08830185.2023.2281566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/08/2023] [Accepted: 10/16/2023] [Indexed: 11/21/2023]
Abstract
Chemokines belong to the group of small proteins within the cytokine family having strong chemo-attractant properties. In most cases, the strong immuno-modulatory role of chemokines is crucial for generating the immune response against pathogens in various protozoan diseases. In this review, we have given a brief update on the classification, characterization, homeostasis, transcellular migration, and immuno-modulatory role of chemokines. Here we will evaluate the potential role of chemokines and their regulation in various protozoan diseases. There is a significant direct relationship between parasitic infection and the recruitment of effector cells of the immune response. Chemokines play an indispensable role in mediating several defense mechanisms against infection, such as leukocyte recruitment and the generation of innate and cell-mediated immunity that aids in controlling/eliminating the pathogen. This process is controlled by the chemotactic movement of chemokines induced as a primary host immune response. We have also addressed that chemokine expressions during infection are time-dependent and orchestrated in a systematic pattern that ultimately assists in generating a protective immune response. Taken together, this review provides a systematic understanding of the complexity of chemokines profiles during protozoan disease conditions and the rationale of targeting chemokines for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Rubika Chauhan
- Parasite-Host Biology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Mrinalini Tiwari
- Parasite-Host Biology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Amrendra Chaudhary
- Parasite-Host Biology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Reva Sharan Thakur
- Parasite-Host Biology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Veena Pande
- Biotechnology Department, Kumaun University, Nainital, India
| | - Jyoti Das
- Parasite-Host Biology, National Institute of Malaria Research, Dwarka, New Delhi, India
| |
Collapse
|
2
|
Goto Y, Mizobuchi H. Pathological roles of macrophages in Leishmania infections. Parasitol Int 2023; 94:102738. [PMID: 36738983 DOI: 10.1016/j.parint.2023.102738] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Macrophages are the major host cells for Leishmania parasites, and determine the fate of infection by either limiting or allowing growth of the parasites, resulting in development or control of leishmaniasis, respectively. They also play important roles in causing pathological outcomes during Leishmania infection. The pathophysiology is complex and include a wide variety of molecular and cellular responses including enhancement of inflammatory responses by releasing cytokines, causing damages to surrounding cells by reactive oxygen species, or disordered phagocytosis of other cells. It is of note that disease severity in leishmaniasis sometimes does not correlate with parasite burdens, indicating that pathological roles of macrophages are not necessarily linked to their parasite-killing activities that are often defined by M1/M2 status. Here, we review the roles of macrophages in leishmaniasis with a focus on their pathological mechanisms in disease development.
Collapse
Affiliation(s)
- Yasuyuki Goto
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| | - Haruka Mizobuchi
- Laboratory of Molecular Immunology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
3
|
Rojas-Pirela M, Andrade-Alviárez D, Medina L, Castillo C, Liempi A, Guerrero-Muñoz J, Ortega Y, Maya JD, Rojas V, Quiñones W, Michels PA, Kemmerling U. MicroRNAs: master regulators in host-parasitic protist interactions. Open Biol 2022; 12:210395. [PMID: 35702995 PMCID: PMC9198802 DOI: 10.1098/rsob.210395] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of small non-coding RNAs present in a wide diversity of organisms. MiRNAs regulate gene expression at a post-transcriptional level through their interaction with the 3' untranslated regions of target mRNAs, inducing translational inhibition or mRNA destabilization and degradation. Thus, miRNAs regulate key biological processes, such as cell death, signal transduction, development, cellular proliferation and differentiation. The dysregulation of miRNAs biogenesis and function is related to the pathogenesis of diseases, including parasite infection. Moreover, during host-parasite interactions, parasites and host miRNAs determine the probability of infection and progression of the disease. The present review is focused on the possible role of miRNAs in the pathogenesis of diseases of clinical interest caused by parasitic protists. In addition, the potential role of miRNAs as targets for the design of drugs and diagnostic and prognostic markers of parasitic diseases is also discussed.
Collapse
Affiliation(s)
- Maura Rojas-Pirela
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile,Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile,Facultad de Farmacia y Bioanálisis, Universidad de Los Andes, Mérida, Venezuela
| | - Diego Andrade-Alviárez
- Laboratorio de Enzimología de Parásitos, Facultad de Ciencias, Universidad de Los Andes, Mérida, Venezuela
| | - Lisvaneth Medina
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Christian Castillo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile,Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Chile
| | - Ana Liempi
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Jesús Guerrero-Muñoz
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Yessica Ortega
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile,Facultad de Farmacia y Bioanálisis, Universidad de Los Andes, Mérida, Venezuela
| | - Juan Diego Maya
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| | - Verónica Rojas
- Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile
| | - Wilfredo Quiñones
- Laboratorio de Enzimología de Parásitos, Facultad de Ciencias, Universidad de Los Andes, Mérida, Venezuela
| | - Paul A. Michels
- Centre for Immunity, Infection and Evolution and Centre for Translational and Chemical Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile 8380453, Chile
| |
Collapse
|
4
|
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
|
5
|
Sellau J, Puengel T, Hoenow S, Groneberg M, Tacke F, Lotter H. Monocyte dysregulation: consequences for hepatic infections. Semin Immunopathol 2021; 43:493-506. [PMID: 33829283 PMCID: PMC8025899 DOI: 10.1007/s00281-021-00852-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
Liver disorders due to infections are a substantial health concern in underdeveloped and industrialized countries. This includes not only hepatotropic viruses (e.g., hepatitis B, hepatitis C) but also bacterial and parasitic infections such as amebiasis, leishmaniasis, schistosomiasis, or echinococcosis. Recent studies of the immune mechanisms underlying liver disease show that monocytes play an essential role in determining patient outcomes. Monocytes are derived from the mononuclear phagocyte lineage in the bone marrow and are present in nearly all tissues of the body; these cells function as part of the early innate immune response that reacts to challenge by external pathogens. Due to their special ability to develop into tissue macrophages and dendritic cells and to change from an inflammatory to an anti-inflammatory phenotype, monocytes play a pivotal role in infectious and non-infectious liver diseases: they can maintain inflammation and support resolution of inflammation. Therefore, tight regulation of monocyte recruitment and termination of monocyte-driven immune responses in the liver is prerequisite to appropriate healing of organ damage. In this review, we discuss monocyte-dependent immune mechanisms underlying hepatic infectious disorders. Better understanding of these immune mechanisms may lead to development of new interventions to treat acute liver disease and prevent progression to organ failure.
Collapse
Affiliation(s)
- Julie Sellau
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Tobias Puengel
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Stefan Hoenow
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marie Groneberg
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Hannelore Lotter
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
6
|
de Freitas E Silva R, von Stebut E. Unraveling the Role of Immune Checkpoints in Leishmaniasis. Front Immunol 2021; 12:620144. [PMID: 33776999 PMCID: PMC7990902 DOI: 10.3389/fimmu.2021.620144] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
Leishmaniasis are Neglected Tropical Diseases affecting millions of people every year in at least 98 countries and is one of the major unsolved world health issues. Leishmania is a parasitic protozoa which are transmitted by infected sandflies and in the host they mainly infect macrophages. Immunity elicited against those parasites is complex and immune checkpoints play a key role regulating its function. T cell receptors and their respective ligands, such as PD-1, CTLA-4, CD200, CD40, OX40, HVEM, LIGHT, 2B4 and TIM-3 have been characterized for their role in regulating adaptive immunity against different pathogens. However, the exact role those receptors perform during Leishmania infections remains to be better determined. This article addresses the key role immune checkpoints play during Leishmania infections, the limiting factors and translational implications.
Collapse
Affiliation(s)
| | - Esther von Stebut
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
7
|
Understanding the immune responses involved in mediating protection or immunopathology during leishmaniasis. Biochem Soc Trans 2021; 49:297-311. [PMID: 33449103 DOI: 10.1042/bst20200606] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 01/21/2023]
Abstract
Leishmaniasis is a vector-borne Neglected Tropical Disease (NTD) transmitted by the sand fly and is a major public health problem worldwide. Infections caused by Leishmania clinically manifest as a wide range of diseases, such as cutaneous (CL), diffuse cutaneous (DCL), mucosal (MCL) and visceral leishmaniasis (VL). The host innate and adaptative immune responses play critical roles in the defense against leishmaniasis. However, Leishmania parasites also manipulate the host immune response for their survival and replication. In addition, other factors such as sand fly salivary proteins and microbiota also promote disease susceptibility and parasite spread by modulating local immune response. Thus, a complex interplay between parasite, sand fly and the host immunity governs disease severity and outcome. In this review, we discuss the host immune response during Leishmania infection and highlight the factors associated with resistance or susceptibility.
Collapse
|
8
|
Bogdan C. Macrophages as host, effector and immunoregulatory cells in leishmaniasis: Impact of tissue micro-environment and metabolism. Cytokine X 2020; 2:100041. [PMID: 33604563 PMCID: PMC7885870 DOI: 10.1016/j.cytox.2020.100041] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Leishmania are protozoan parasites that predominantly reside in myeloid cells within their mammalian hosts. Monocytes and macrophages play a central role in the pathogenesis of all forms of leishmaniasis, including cutaneous and visceral leishmaniasis. The present review will highlight the diverse roles of macrophages in leishmaniasis as initial replicative niche, antimicrobial effectors, immunoregulators and as safe hideaway for parasites persisting after clinical cure. These multiplex activities are either ascribed to defined subpopulations of macrophages (e.g., Ly6ChighCCR2+ inflammatory monocytes/monocyte-derived dendritic cells) or result from different activation statuses of tissue macrophages (e.g., macrophages carrying markers of of classical [M1] or alternative activation [M2]). The latter are shaped by immune- and stromal cell-derived cytokines (e.g., IFN-γ, IL-4, IL-10, TGF-β), micro milieu factors (e.g., hypoxia, tonicity, amino acid availability), host cell-derived enzymes, secretory products and metabolites (e.g., heme oxygenase-1, arginase 1, indoleamine 2,3-dioxygenase, NOS2/NO, NOX2/ROS, lipids) as well as by parasite products (e.g., leishmanolysin/gp63, lipophosphoglycan). Exciting avenues of current research address the transcriptional, epigenetic and translational reprogramming of macrophages in a Leishmania species- and tissue context-dependent manner.
Collapse
Key Words
- (L)CL, (localized) cutaneous leishmaniasis
- AHR, aryl hydrocarbon receptor
- AMP, antimicrobial peptide
- Arg, arginase
- Arginase
- CAMP, cathelicidin-type antimicrobial peptide
- CR, complement receptor
- DC, dendritic cells
- DCL, diffuse cutaneous leishmaniasis
- HO-1, heme oxygenase 1
- Hypoxia
- IDO, indoleamine-2,3-dioxygenase
- IFN, interferon
- IFNAR, type I IFN (IFN-α/β) receptor
- IL, interleukin
- Interferon-α/β
- Interferon-γ
- JAK, Janus kinase
- LPG, lipophosphoglycan
- LRV1, Leishmania RNA virus 1
- Leishmaniasis
- Macrophages
- Metabolism
- NCX1, Na+/Ca2+ exchanger 1
- NFAT5, nuclear factor of activated T cells 5
- NK cell, natural killer cell
- NO, nitric oxide
- NOS2 (iNOS), type 2 (or inducible) nitric oxide synthase
- NOX2, NADPH oxidase 2 (gp91 or cytochrome b558 β-subunit of Phox)
- Nitric oxide
- OXPHOS, mitochondrial oxidative phosphorylation
- PKDL, post kala-azar dermal leishmaniasis
- Phagocyte NADPH oxidase
- Phox, phagocyte NADPH oxidase
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SOCS, suppressor of cytokine signaling
- STAT, signal transducer and activator of transcription
- TGF-β, transforming growth factor-beta
- TLR, toll-like receptor
- Th1 (Th2), type 1 (type2) T helper cell
- Tonicity
- VL, visceral leishmaniasis
- mTOR, mammalian/mechanistic target of rapamycin
Collapse
Affiliation(s)
- Christian Bogdan
- Mikrobiologisches Institut - klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
9
|
Aghaei M, Khanahmad H, Aghaei S, Hosseini SM, Farahmand M, Hejazi SH. Evaluation of transgenic Leishmania infantum expressing mLLO-BAX-SMAC in the apoptosis of the infected macrophages in vitro and in vivo. Parasite Immunol 2020; 42:e12726. [PMID: 32367588 DOI: 10.1111/pim.12726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/31/2020] [Accepted: 04/28/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Leishmaniasis is an important infectious disease that develops because of escaping parasite from the host immune system or preventing host macrophages apoptosis. Recently, the development of transgenic methods and the manipulation of the parasite genome has provided many advantages. So, in this study, the effect of the transgenic Leishmania infantum expressing mLLO-BAX-SMAC proteins was examined in accelerating host cell apoptosis. METHOD The entire coding sequence of designed codon-optimized mLLO-Bax-Smac was cloned in the pLexyNeo2 vector and integrated downstream of the 18srRNA locus of L infantum genome by homologous recombination. Next, the expression of mLLO-BAX-SMAC fusion protein was evaluated by the Western blotting technique and the pathogenesis of transgenic parasite was surveyed in vitro and in vivo. RESULTS The results of PCR and Western blot confirmed proper integration and expression of mLLO-Bax-Smac sequence into the 18srRNA locus of L infantum. Flow cytometry showed accelerating apoptosis of transgenic Leishmania-infected macrophages compared to wild-type parasite. Also, transgenic parasites were less virulent as a fewer parasitic burden was found in the spleen and liver of transgenic-infected mice compared to the control. CONCLUSION The data suggested that the transgenic L infantum expressing BAX-SMAC can be used as an experimental model for developing vaccination against leishmaniasis.
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
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sayed Mohsen Hosseini
- Department of Biostatistics & Epidemiology, School of Public Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahin Farahmand
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, 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
| |
Collapse
|
10
|
Tedla MG, Every AL, Scheerlinck JPY. Investigating immune responses to parasites using transgenesis. Parasit Vectors 2019; 12:303. [PMID: 31202271 PMCID: PMC6570953 DOI: 10.1186/s13071-019-3550-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/03/2019] [Indexed: 11/10/2022] Open
Abstract
Parasites comprise diverse and complex organisms, which substantially impact human and animal health. Most parasites have complex life-cycles, and by virtue of co-evolution have developed multifaceted, often life-cycle stage-specific relationships with the immune system of their hosts. The complexity in the biology of many parasites often limits our knowledge of parasite-specific immune responses, to in vitro studies only. The relatively recent development of methods to stably manipulate the genetic make-up of many parasites has allowed a better understanding of host-parasite interactions, particularly in vivo. In this regard, the use of transgenic parasites can facilitate the study of immunomodulatory mechanisms under in vivo conditions. Therefore, in this review, we specifically highlighted the current developments in the use of transgenic parasites to unravel the host's immune response to different life-cycle stages of some key parasite species such as Leishmania, Schistosoma, Toxoplasma, Plasmodium and Trypanosome and to some degree, the use of transgenic nematode parasites is also briefly discussed.
Collapse
Affiliation(s)
- Mebrahtu G. Tedla
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, VIC 3010 Australia
| | - Alison L. Every
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, VIC 3010 Australia
- Present Address: College of Science, Health and Engineering, La Trobe University, Melbourne, VIC 3086 Australia
| | - Jean-Pierre Y. Scheerlinck
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, VIC 3010 Australia
| |
Collapse
|
11
|
Martins TAF, Barbosa VS, Almeida GG, Antonelli LRDV, Tafuri WL, Mosser DM, Gonçalves R. Monocyte subpopulations as important biomarkers of resistence and susceptibility during experimental infection with Leishmania (Leishmania) major. Biomed Pharmacother 2018; 107:1530-1539. [PMID: 30257371 DOI: 10.1016/j.biopha.2018.08.154] [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: 07/13/2018] [Revised: 08/17/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022] Open
Abstract
Visceral Leishmaniasis is a chronic and lethal, parasitic disease. In the later infection stages, it is known that expressive hematological disorders can be observed, including changes in the frequency and phenotype of certain leukocytes. There is a lack of good prognostic indicators to characterize the on-goin clinical status of the patient. In this study, we have analyzed the frequency of monocyte subpopulations in mice infected with Leishmania major (L. major). Our results show a significant correlation between increased blood monocyte frequency and lesion development in both BALB/c and in the C57BL/6 mice infected with L. major. In BALB/c mice we observed a significant correlation between the frequency of GR1+ monocytes and lesion size. Furthermore, treatment of infected BALB/c mice with Anfotericin B, to resolve lesions, resulted in a lower frequency of GR1+ monocytes compared to untreated infected BALB/c mice. C57BL/6 infected mice, which normally resolve infections, show decreased numbers of monocytes during the healing phase of infection. The results indicate that disease severity can be predicted by analyzing monocyte frequency. Thus, we propose that the frequency of monocytes, can be used to define the severity of the disease as well as the success of the treatment in experimental leishmaniasis.
Collapse
Affiliation(s)
- Tassiane Assiria Fontes Martins
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vitor Silva Barbosa
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gregório Guilherme Almeida
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Wagner Luiz Tafuri
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - David M Mosser
- Department of Cell Biology and Molecular Genetics and Maryland Pathogen Research Institute and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA
| | - Ricardo Gonçalves
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
12
|
Azami M, Ranjkesh Adermanabadi V, Khanahmad H, Mohaghegh MA, Zaherinejad E, Aghaei M, Jalali A, Hejazi SH. Immunology and Genetic of Leishmania infantum: The Role of Endonuclease G in the Apoptosis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2018; 23:36. [PMID: 29887904 PMCID: PMC5961285 DOI: 10.4103/jrms.jrms_705_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/13/2018] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
Leishmania infantum is the causative agent of infantile visceral leishmaniasis (VL) in the Mediterranean region. Despite developing protective responses, the disease progresses due to many of factors. These include the action of suppressive cytokines, exhaustion of specific T cells, loss of lymphoid tissue, and defective humoral response. Genetic changes that occur inside the genome of alienated or parasite cells, along with immune responses, play an important role in controlling or progressing the disease. Proapoptotic proteins such as Smac/DIABLO, EndoG, AIF (apoptosis-inducing factor), and cytochrome C are effective in apoptosis. EndoG is a mitochondrion-specific nuclease that translocates to the nucleus during apoptosis. Once released from mitochondria, endoG cleaves chromatin DNA into nucleosomal fragments independently of caspases. Therefore, endoG represents a caspase-independent apoptotic pathway initiated from the mitochondria. A comprehensive understanding of the immune and genetic events that occur during VL is very important for designing immunotherapy strategies and developing effective vaccines for disease prevention. In this review which explained the immunological responses and also the important factors that can contribute to parasite apoptosis and are used in subsequent studies as a target for the preparation of drugs or recombinant vaccines against parasites are briefly reviewed.
Collapse
Affiliation(s)
- Mehdi Azami
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hossein Khanahmad
- Department of Molecular Biology and Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Ali Mohaghegh
- Department of Laboratory Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ebtesam Zaherinejad
- School of Pharmacy, Department of Health Sciences, Curtin University of Technology, Bentley Campus, Australia
| | - Maryam Aghaei
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Akram Jalali
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Caffeic acid combined with autoclaved Leishmania major boosted the protection of infected BALB/c mice by enhancing IgG2 production, IFN-γ/TGF-β and iNO synthase/arginase1 ratios, and the death of infected phagocytes. Inflammopharmacology 2017; 26:621-634. [DOI: 10.1007/s10787-017-0399-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/12/2017] [Indexed: 12/19/2022]
|
14
|
Nandi A, Bishayi B. CCR-2 neutralization augments murine fresh BMC activation by Staphylococcus aureus via two distinct mechanisms: at the level of ROS production and cytokine response. Innate Immun 2017; 23:345-372. [PMID: 28409543 DOI: 10.1177/1753425917697806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CCR-2 signaling regulates recruitment of monocytes from the bone marrow into the bloodstream and then to sites of infection. We sought to determine whether CCL-2/CCR-2 signaling is involved in the killing of Staphylococcus aureus by murine bone marrow cells (BMCs). The intermittent link of reactive oxygen species (ROS)-NF-κB/p38-MAPK-mediated CCL-2 production in CCR-2 signaling prompted us to determine whether neutralization of CCR-2 augments the response of murine fresh BMCs (FBMCs) after S. aureus infection. It was observed that anti-CCR-2 Ab-treated FBMCs released fewer ROS on encountering S. aureus infection than CCR-2 non-neutralized FBMCs, also correlating with reduced killing of S. aureus in CCR-2 neutralized FBMCs. Staphylococcal catalase and SOD were also found to play a role in protecting S. aureus from the ROS-mediated killing of FBMC. S. aureus infection of CCR-2 intact FBMCs pre-treated with either NF-κB or p-38-MAPK blocker induced less CCL-2, suggesting that NF-κB or p-38-MAPK is required for CCL-2 production by FBMCs. Moreover, blocking of CCR-2 along with NF-κB or p-38-MAPK resulted in elevated CCL-2 production and reduced CCR-2 expression. Inhibition of CCR-2 impairs the response of murine BMCs to S. aureus infection by attenuation ROS production and modulating the cytokine response.
Collapse
Affiliation(s)
- Ajeya Nandi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, West Bengal, India
| |
Collapse
|
15
|
Christensen SM, Dillon LAL, Carvalho LP, Passos S, Novais FO, Hughitt VK, Beiting DP, Carvalho EM, Scott P, El-Sayed NM, Mosser DM. Meta-transcriptome Profiling of the Human-Leishmania braziliensis Cutaneous Lesion. PLoS Negl Trop Dis 2016; 10:e0004992. [PMID: 27631090 PMCID: PMC5025153 DOI: 10.1371/journal.pntd.0004992] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 08/19/2016] [Indexed: 11/24/2022] Open
Abstract
Host and parasite gene expression in skin biopsies from Leishmania braziliensis-infected patients were simultaneously analyzed using high throughput RNA-sequencing. Biopsies were taken from 8 patients with early cutaneous leishmaniasis and 17 patients with late cutaneous leishmaniasis. Although parasite DNA was found in all patient lesions at the time of biopsy, the patients could be stratified into two groups: one lacking detectable parasite transcripts (PTNeg) in lesions, and another in which parasite transcripts were readily detected (PTPos). These groups exhibited substantial differences in host responses to infection. PTPos biopsies contained an unexpected increase in B lymphocyte-specific and immunoglobulin transcripts in the lesions, and an upregulation of immune inhibitory molecules. Biopsies without detectable parasite transcripts showed decreased evidence for B cell activation, but increased expression of antimicrobial genes and genes encoding skin barrier functions. The composition and abundance of L. braziliensis transcripts in PTPos lesions were surprisingly conserved among all six patients, with minimal meaningful differences between lesions from patients with early and late cutaneous leishmaniasis. The most abundant parasite transcripts expressed in lesions were distinct from transcripts expressed in vitro in human macrophage cultures infected with L. amazonensis or L. major. Therefore in vitro gene expression in macrophage monolayers may not be a strong predictor of gene expression in lesions. Some of the most highly expressed in vivo transcripts encoded amastin-like proteins, hypothetical genes, putative parasite virulence factors, as well as histones and tubulin. In summary, RNA sequencing allowed us to simultaneously analyze human and L. braziliensis transcriptomes in lesions of infected patients, and identify unexpected differences in host immune responses which correlated with active transcription of parasite genes. Leishmania spp are intracellular protozoan parasites that replicate primarily within host tissue macrophages. In this paper we simultaneously query host and parasite gene expression in human cutaneous L. braziliensis lesions. We observe an unexpectedly prominent role for B cells and immunoglobulins in lesions in which actively transcribing parasites reside. We also observe that parasite gene expression is surprisingly conserved among L. braziliensis lesions, and the genes that are expressed in lesions are not those that have been previously associated with parasite growth in vitro. This analysis of parasite and host gene expression in lesions may lead to the identification of new parasite virulence factors and may identify host responses that promote parasite persistence in lesions.
Collapse
Affiliation(s)
- Stephen M. Christensen
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Laura A. L. Dillon
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | | | - Sara Passos
- Universidade Federal da Bahia Salvador, Bahia, Brazil
| | - Fernanda O. Novais
- Department of Pathobiology, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - V. Keith Hughitt
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - David M. Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Costa DL, Lima-Júnior DS, Nascimento MS, Sacramento LA, Almeida RP, Carregaro V, Silva JS. CCR2 signaling contributes to the differentiation of protective inflammatory dendritic cells in Leishmania braziliensis infection. J Leukoc Biol 2016; 100:423-32. [PMID: 26884611 DOI: 10.1189/jlb.4a0715-288r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 01/26/2016] [Indexed: 12/11/2022] Open
Abstract
In vertebrate hosts, Leishmania braziliensis parasites infect mainly mononuclear phagocytic system cells, which when activated by T helper cell type 1 cytokines produce nitric oxide and kill the pathogens. Chemokine (C-C motif) receptor 2 is a chemokine receptor that binds primarily chemokine (C-C motif) ligand 2 and has an important role in the recruitment of monocytic phagocytes. Although it has been reported that Leishmania braziliensis infection induces CCR2 expression in the lesions, the role of CCR2 during Leishmania braziliensis infection remains unknown. Here, we showed that CCR2 has a role in mediating protection against Leishmania braziliensis infection in mice. The absence of CCR2 resulted in increased susceptibility to infection and was associated with low amounts of Ly6C(+) inflammatory dendritic cells in the lesions, which we found to be the major sources of tumor necrosis factor production and induced nitric oxide synthase expression in C57BL/6 mice lesions. Consequently, CCR2(-/-) mice showed decreased tumor necrosis factor production and induced nitric oxide synthase expression, resulting in impaired parasite elimination. We also demonstrated that CCR2 has a role in directly mediating the differentiation of monocytes into inflammatory dendritic cells at the infection sites, contributing to the accumulation of inflammatory dendritic cells in Leishmania braziliensis lesions and subsequent control of parasite replication. Therefore, these data provide new information on the role of chemokines during the immune response to infections and identify a potential target for therapeutic interventions in cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Diego L Costa
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Djalma S Lima-Júnior
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Manuela S Nascimento
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Laís A Sacramento
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - Roque P Almeida
- Department of Internal Medicine and Pathology, Center for Biological and Health Sciences, Federal University of Sergipe, Aracajú, SE, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; and
| |
Collapse
|
17
|
Passos S, Carvalho LP, Costa RS, Campos TM, Novais FO, Magalhães A, Machado PRL, Beiting D, Mosser D, Carvalho EM, Scott P. Intermediate monocytes contribute to pathologic immune response in Leishmania braziliensis infections. J Infect Dis 2014; 211:274-82. [PMID: 25139016 DOI: 10.1093/infdis/jiu439] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ulcer development in patients with cutaneous leishmaniasis (CL) caused by Leishmania braziliensis is associated with high levels of tumor necrosis factor (TNF). We found that early after infection, before ulcer development, the frequency of CD16(+) (both intermediate [CD14(+)CD16(+)] and nonclassical [CD14(dim)CD16(+)]) monocytes was increased in the peripheral blood of patients with L. braziliensis, compared with uninfected controls. These results suggest that CD16(+) monocytes might promote disease. Also, we found that intermediate monocytes expressed CCR2 and that increased levels of CCL2 protein were present in lesions from patients, suggesting that intermediate monocytes are more likely than nonclassical monocytes to migrate to the lesion site. Finally, we found that the intermediate monocytes produced TNF. Our results show that intermediate monocytes are increased in frequency soon after infection; express CCR2, which would promote their migration into the lesions; and, owing to their production of TNF, can enhance the inflammatory response.
Collapse
Affiliation(s)
- Sara Passos
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Lucas P Carvalho
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos Instituto de Ciências da Saúde, Universidade Federal da Bahia Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Rúbia S Costa
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos
| | - Taís M Campos
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos
| | - Fernanda O Novais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia
| | | | - Paulo R L Machado
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Daniel Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia
| | - David Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park
| | - Edgar M Carvalho
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
18
|
Redpath SA, Fonseca NM, Perona-Wright G. Protection and pathology during parasite infection: IL-10 strikes the balance. Parasite Immunol 2014; 36:233-52. [PMID: 24666543 DOI: 10.1111/pim.12113] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/18/2014] [Indexed: 12/16/2022]
Abstract
The host response to infection requires an immune response to be strong enough to control the pathogen but also restrained, to minimize immune-mediated pathology. The conflicting pressures of immune activation and immune suppression are particularly apparent in parasite infections, where co-evolution of host and pathogen has selected many different compromises between protection and pathology. Cytokine signals are critical determinants of both protective immunity and immunopathology, and, in this review, we focus on the regulatory cytokine IL-10 and its role in protozoan and helminth infections. We discuss the sources and targets of IL-10 during parasite infection, the signals that initiate and reinforce its action, and its impact on the invading parasite, on the host tissue, and on coincident immune responses.
Collapse
Affiliation(s)
- S A Redpath
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
19
|
Kling JC, Mack M, Körner H. The absence of CCR7 results in dysregulated monocyte migration and immunosuppression facilitating chronic cutaneous leishmaniasis. PLoS One 2013; 8:e79098. [PMID: 24205367 PMCID: PMC3813618 DOI: 10.1371/journal.pone.0079098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/18/2013] [Indexed: 12/22/2022] Open
Abstract
The protozoan parasite Leishmania major causes cutaneous lesions to develop at the site of infection, which are resolved with a strong Th1 immune response in resistant hosts, such as C57BL/6 mice. In contrast, the lesions ulcerate in susceptible hosts which display a Th2 response, such as BALB/c mice. The migration of cells in the immune response to L. major is regulated by chemokines and their receptors. The chemokine receptor CCR7 is expressed on activated DCs and naïve T cells, allowing them to migrate to the correct micro-anatomical positions within secondary lymphoid organs. While there have been many studies on the function of CCR7 during homeostasis or using model antigens, there are very few studies on the role of CCR7 during infection. In this study, we show that B6.CCR7-/- mice were unable to resolve the lesion and developed a chronic disease. The composition of the local infiltrate at the lesion was significantly skewed toward neutrophils while the proportion of CCR2+ monocytes was reduced. Furthermore, a greater percentage of CCR2+ monocytes expressed CCR7 in the footpad than in the lymph node or spleen of B6.WT mice. We also found an increased percentage of regulatory T cells in the draining lymph node of B6.CCR7-/- mice throughout infection. Additionally, the cytokine milieu of the lymph node showed a Th2 bias, rather than the resistant Th1 phenotype. This data shows that CCR7 is required for a protective immune response to intracellular L. major infection.
Collapse
Affiliation(s)
| | - Matthias Mack
- Innere Medizin II, Nephrologie/Forschung, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Heinrich Körner
- Menzies Research Institute, Hobart, Tasmania, Australia
- * E-mail:
| |
Collapse
|
20
|
Allers K, Fehr M, Conrad K, Epple HJ, Schürmann D, Geelhaar-Karsch A, Schinnerling K, Moos V, Schneider T. Macrophages accumulate in the gut mucosa of untreated HIV-infected patients. J Infect Dis 2013; 209:739-48. [PMID: 24133185 DOI: 10.1093/infdis/jit547] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mucosal macrophages are involved in the maintenance of epithelial barrier integrity and the elimination of invading pathogens. Although an intestinal barrier defect and microbial translocation are hallmarks of human immunodeficiency virus (HIV) infection, recent data on gut mucosal macrophages in HIV infection are sparse. METHODS Treatment-naive and treated HIV-infected patients and healthy controls were studied for frequencies and functional parameters of blood monocytes and macrophages in duodenal mucosa. RESULTS We found mucosal enrichment of macrophages in untreated HIV infection associated with reduced monocyte counts in blood and increased monocyte expression of the gut-homing molecule integrin β7. Increased CCR2 density on integrin β7-expressing monocytes and mucosal secretion of CCL2 suggest that CCR2/CCL2-chemotaxis is involved in enhanced trafficking of blood monocytes to the gut. Secretion of macrophage-related proinflammatory molecules interleukin 1β, CCL5, CXCL9, and CXCL10 was increased in the gut mucosa of untreated patients. Moreover, mucosal macrophages of untreated patients showed reduced phagocytic activity. CONCLUSIONS These data suggest a role for gut mucosal macrophages in HIV immune pathogenesis: infiltrated macrophages in the intestinal mucosa may promote local inflammation and tissue injury, whereas their low phagocytic activity prevents the efficient elimination of luminal antigens that cross the damaged intestinal barrier.
Collapse
Affiliation(s)
- Kristina Allers
- Department of Gastroenterology, Infectious Diseases, and Rheumatology, Medical Clinic I, Campus Benjamin Franklin
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Kling JC, Körner H. Different regulatory mechanisms in protozoan parasitic infections. Int J Parasitol 2013; 43:417-25. [PMID: 23470812 DOI: 10.1016/j.ijpara.2013.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 02/07/2023]
Abstract
The immune response to the protozoan pathogens, Leishmania spp., Trypanosoma spp. and Plasmodium spp., has been studied extensively with particular focus on regulation of the immune response by immunological mechanisms. More specifically, in diseases caused by parasites, immunosuppression frequently prevents immunopathology that can injure the host. However, this allows a small number of parasites to evade the immune response and remain in the host after a clinical cure. The consequences can be chronic infections, which establish a zoonotic or anthroponotic reservoir. This review will highlight some of the identified regulatory mechanisms of the immune system that govern immune responses to parasitic diseases, in particular leishmaniasis, trypanosomiasis and malaria, and discuss implications for the development of efficient vaccines against these diseases.
Collapse
Affiliation(s)
- Jessica C Kling
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | | |
Collapse
|
22
|
Bai J, Song H, Cai C, Zhang M, Xu S, Tan J. The association of monocyte chemotactic protein-1 and CC chemokine receptor 2 gene variants with chronic obstructive pulmonary disease. DNA Cell Biol 2012; 31:1058-63. [PMID: 22276574 DOI: 10.1089/dna.2011.1520] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chemokines are potent proinflammatory cytokines that are implicated in numerous inflammatory diseases. Monocyte chemoattractant protein-1 (MCP-1) and its receptor CC chemokine receptor-2 (CCR2) play a major role in the recruitment of inflammatory cells to the lungs of patients with chronic obstructive pulmonary disease (COPD). We investigated a possible association between polymorphisms in MCP-1 and CCR2 genes (MCP-1 -2518 A/G and CCR2 190G/A or V64I) and the development of COPD. Genotypes were determined by polymerase chain reaction-restriction fragment length polymorphism in 386 COPD cases and 398 age-matched healthy controls. Frequency of MCP-1 2518GG genotype for cases and controls was 0.396 and 0.324, respectively; individuals who had the GG genotype had a 1.59-fold increased risk of COPD (p=0.036). Frequency of CCR2 190AA (64I/64I) genotype for cases and controls was 0.285 and 0.21, respectively; subjects carrying the 64I/64I genotype had a 2.04-fold increased risk of COPD compared with the wild-type genotype (p=0.001). When analyzing the allele combination of these two polymorphisms, the combinations MCP-1-A/CCR2-A and MCP-1-G/CCR2-A were detected in significantly higher numbers in COPD cases than in healthy controls (odds ratio [OR]=1.50, 95% confidence interval [CI]: 1.04-2.17, p=0.032; and OR=1.89, 95% CI: 1.38-2.60, p=7.38×10(-5)). These data suggest that MCP-1 -2518 A/G and CCR2 190G/A polymorphisms are new risk factors for COPD.
Collapse
Affiliation(s)
- Jianwen Bai
- Emergency Center, Shanghai East Hospital, Shanghai, China
| | | | | | | | | | | |
Collapse
|
23
|
Pereira WF, Ribeiro-Gomes FL, Guillermo LVC, Vellozo NS, Montalvão F, DosReis GA, Lopes MF. Myeloid-derived suppressor cells help protective immunity to Leishmania major
infection despite suppressed T cell responses. J Leukoc Biol 2011; 90:1191-7. [DOI: 10.1189/jlb.1110608] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
24
|
Abstract
Intracellular parasitism has arisen only a few times during the long ancestry of protozoan parasites including in diverse groups such as microsporidians, kinetoplastids, and apicomplexans. Strategies used to gain entry differ widely from injection (e.g. microsporidians), active penetration of the host cell (e.g. Toxoplasma), recruitment of lysosomes to a plasma membrane wound (e.g. Trypanosoma cruzi), to host cell-mediated phagocytosis (e.g. Leishmania). The resulting range of intracellular niches is equally diverse ranging from cytosolic (e.g. T. cruzi) to residing within a non-fusigenic vacuole (e.g. Toxoplasma, Encephalitozoon) or a modified phagolysosome (e.g. Leishmania). These lifestyle choices influence access to nutrients, interaction with host cell signaling pathways, and detection by pathogen recognition systems. As such, intracellular life requires a repertoire of adaptations to assure entry-exit from the cell, as well as to thwart innate immune mechanisms and prevent clearance. Elucidating these pathways at the cellular and molecular level may identify key steps that can be targeted to reduce parasite survival or augment immunologic responses and thereby prevent disease.
Collapse
Affiliation(s)
- L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63100, USA.
| |
Collapse
|
25
|
Goncalves R, Zhang X, Cohen H, Debrabant A, Mosser DM. Platelet activation attracts a subpopulation of effector monocytes to sites of Leishmania major infection. ACTA ACUST UNITED AC 2011; 208:1253-65. [PMID: 21606505 PMCID: PMC3173254 DOI: 10.1084/jem.20101751] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Leishmania infection triggers the recruitment of Gr1+ monocytes to the site of infection via platelet-derived PDGF and subsequent CCL2 production. Leishmania species trigger a brisk inflammatory response and efficiently induce cell-mediated immunity. We examined the mechanisms whereby leukocytes were recruited into lesions after Leishmania major infection of mice. We found that a subpopulation of effector monocytes expressing the granulocyte marker GR1 (Ly6C) is rapidly recruited into lesions, and these monocytes efficiently kill L. major parasites. The recruitment of this subpopulation of monocytes depends on the chemokine receptor CCR2 and the activation of platelets. Activated platelets secrete platelet-derived growth factor, which induces the rapid release of CCL2 from leukocytes and mesenchymal cells. This work points to a new role for platelets in host defense involving the selective recruitment of a subpopulation of effector monocytes from the blood to efficiently kill this intracellular parasite.
Collapse
Affiliation(s)
- Ricardo Goncalves
- Department of Cell Biology and Molecular Genetics and the Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20782, USA
| | | | | | | | | |
Collapse
|
26
|
Chemokines and chemokine receptors coordinate the inflammatory immune response in human cutaneous leishmaniasis. Hum Immunol 2010; 71:1220-7. [DOI: 10.1016/j.humimm.2010.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 08/09/2010] [Accepted: 09/09/2010] [Indexed: 10/19/2022]
|
27
|
Hutchings CJ, Koglin M, Marshall FH. Therapeutic antibodies directed at G protein-coupled receptors. MAbs 2010; 2:594-606. [PMID: 20864805 DOI: 10.4161/mabs.2.6.13420] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are one of the most important classes of targets for small molecule drug discovery, but many current GPCRs of interest are proving intractable to small molecule discovery and may be better approached with bio-therapeutics. GPCRs are implicated in a wide variety of diseases where antibody therapeutics are currently used. These include inflammatory diseases such as rheumatoid arthritis and Crohn disease, as well as metabolic disease and cancer. Raising antibodies to GPCRs has been difficult due to problems in obtaining suitable antigen because GPCRs are often expressed at low levels in cells and are very unstable when purified. A number of new developments in over-expressing receptors, as well as formulating stable pure protein, are contributing to the growing interest in targeting GPCRs with antibodies. This review discusses the opportunities for targeting GPCRs with antibodies using these approaches and describes the therapeutic antibodies that are currently in clinical development.
Collapse
|
28
|
Reis MLC, Ferreira VM, Zhang X, Gonçalves R, Vieira LQ, Tafuri WL, Mosser DM, Tafuri WL. Murine immune response induced by Leishmania major during the implantation of paraffin tablets. Virchows Arch 2010; 457:609-18. [PMID: 20857143 PMCID: PMC3057163 DOI: 10.1007/s00428-010-0974-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 07/16/2010] [Accepted: 09/07/2010] [Indexed: 11/27/2022]
Abstract
We carried out a model of chronic inflammation using a subcutaneous paraffin tablet in mice experimentally infected with Leishmania major. It was previously reported that the parasite load following paraffin implantation occurred at a peak of 21 days in both BALB/c and C57BL/6 mice. At the present study, we have investigated what cytokines and chemokines are directly related to the parasite load in C57BL/6 mice. All mice were divided in four groups: mice implanted with paraffin tablets; mice experimentally infected with L. major; mice implanted with paraffin tablets and experimentally infected with L. major; and mice submitted only to the surgery were used for the Real-Time Polymerase Chain Reaction (RT-PCR) controls. Fragments of skin tissue and the tissue surrounding the paraffin tablets (inflammatory capsule) were collected for histopathology and RT-PCR studies. By 21 days, a diffuse chronic inflammatory reaction was mainly observed in the deep dermis where macrophages parasitized with Leishmania amastigotes were also found. RT-PCR analysis has shown that BALB/c mice showed strong IL-4 and IL-10 mRNA expression than controls with very little expression of IFN-γ. In contrast, both IFN-γ and IL-10 mRNA was found in higher levels in C57BL/6 animals. Moreover, in C57BL/6 mice the expression of chemokines mRNA of CCL3/MIP-1α was more highly expressed than CCL2/MCP-1. We conclude that the Th1 immune response C57BL/6 did not change to a Th2 response, even though C57BL/6 animals presented higher parasitism than BALB/c mice 21 days after infection and paraffin implantation.
Collapse
Affiliation(s)
- Maria Letícia Costa Reis
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Av Antonio Carlos, Campus Pampulha, Belo Horizonte 6627 MG, Brazil
| | - Vanessa Martins Ferreira
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Av Antonio Carlos, Campus Pampulha, Belo Horizonte 6627 MG, Brazil
| | - Xia Zhang
- Department of Cell Biology and Molecular Genetics, Univeristy of Maryland, Microbiology Bldg, College Park, MD 20742, USA
| | - Ricardo Gonçalves
- Instituto de Ciências Exatas e Biológicas, Núcleo de Pesquisas em Ciências Biológicas (NUPEB), Universidade Federal de Ouro Preto, Morro do Cruzeiro, Bauxita, Ouro Preto, MG, Brazil
| | - Leda Quércia Vieira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Av Antonio Carlos, Campus Pampulha, Belo Horizonte 6627 MG, Brazil
| | - Washington Luiz Tafuri
- Departamento de Anatomia Patológica e Medicina Legal, Universidade Federal de Minas Gerais, Faculdade de Medicina, Av Alfredo Balena 90, Belo Horizonte, MG, Brazil
| | - David M. Mosser
- Department of Cell Biology and Molecular Genetics, Univeristy of Maryland, Microbiology Bldg, College Park, MD 20742, USA
| | - Wagner Luiz Tafuri
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Av Antonio Carlos, Campus Pampulha, Belo Horizonte 6627 MG, Brazil
| |
Collapse
|
29
|
LÓPEZ R, CUCA LE, DELGADO G. Antileishmanial and immunomodulatory activity ofXylopia discreta. Parasite Immunol 2009; 31:623-30. [DOI: 10.1111/j.1365-3024.2009.01134.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Abstract
Monocyte chemoattractant protein-1 (MCP-1) is a major chemoattractant for monocytes and memory T cells by means of their binding to its specific cell-surface receptor, CC-chemokine receptor-2 (CCR2). CCR2 belongs to the G-protein-coupled seven-transmembrane receptor superfamily. The evidence in favor of CCR2 and MCP-1 having dominant roles in monocyte chemotaxis and chronic inflammation was provided by CCR2 and MCP-1 knockout mice. It has been recognized that CCR2 antagonists are potential therapeutic agents in preventing, treating, or ameliorating a CCR2-mediated inflammatory syndrome or disease such as psoriasis, uveitis, rheumatoid arthritis, multiple sclerosis, asthma, obesity, and chronic obstructive pulmonary disease. This review summarizes recent developments in small-molecule CCR2 antagonists disclosed by patent applications published between 2005 and 2008 and related publications.
Collapse
Affiliation(s)
- Mingde Xia
- Drug Discovery, Johnson & Johnson Pharmaceutical Research and Development LLC, Cranbury, NJ 08512, USA.
| | | |
Collapse
|
31
|
Benevides L, Milanezi CM, Yamauchi LM, Benjamim CF, Silva JS, Silva NM. CCR2 receptor is essential to activate microbicidal mechanisms to control Toxoplasma gondii infection in the central nervous system. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:741-51. [PMID: 18688032 DOI: 10.2353/ajpath.2008.080129] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Chemokines comprise a structurally related family of cytokines that regulate leukocyte trafficking. Because infection with Toxoplasma gondii can induce an important inflammatory reaction that, if left uncontrolled, can lead to death, we investigated the role of the chemokine receptor CCR2 in T. gondii infection. We orally infected CCR2(-/-) mice with five ME-49 T. gondii cysts and monitored morbidity, survival, and immune response thereafter. The CCR2(-/-) mice displayed higher susceptibility to infection as all mice died on day 28 after infection. Despite similar Th1 responses, a more evident anti-inflammatory response was induced in the peripheral organs of CCR2(-/-) mice compared with wild-type C57BL/6 mice. Additionally, CCR2(-/-) mice presented greater parasitism and a milder inflammatory reaction in their peripheral organs with lesser CD4(+) and MAC-1(+) and greater CD8(+) cell migration. The parasite load decreased in these organs in CCR2(-/-) mice but remained uncontrolled in the central nervous system. Additionally, we observed down-regulated inducible nitric oxide synthase expression in peripheral organs from CCR2(-/-) mice that was associated with a small nitric oxide production by spleen macrophages. In conclusion, in the absence of CCR2, another mechanism is activated to control tissue parasitism in peripheral organs. Nevertheless, CCR2 is essential for the activation of microbicidal mediators that control T. gondii replication in the central nervous system.
Collapse
Affiliation(s)
- Luciana Benevides
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
32
|
Substituted dipiperidine alcohols as potent CCR2 antagonists. Bioorg Med Chem Lett 2008; 18:3562-4. [DOI: 10.1016/j.bmcl.2008.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 04/30/2008] [Accepted: 05/01/2008] [Indexed: 11/18/2022]
|
33
|
Abstract
Genetic manipulation of single-celled organisms such as the Leishmania parasite enables in depth analysis of the consequences of genotypic change on biological function. In probing the immune responses to infection, use of transgenic Leishmania has the potential to unravel both the contribution of the parasite to the infection process and the cellular interactions and mechanisms that characterize the innate and adaptive immune responses of the host. Here, we briefly review recent technical advances in parasite genetics and explore how these methods are being used to investigate parasite virulence factors, elucidate immune regulatory mechanisms and contribute to the development of novel therapeutics for the leishmaniases. Recent developments in imaging technology, such as bioluminescence and intravital imaging, combined with parasite transfection with fluorescent or enzyme-encoding marker genes, provides a rich opportunity for novel assessment of intimate, real-time host-parasite interactions at a previously unexplored level. Further advances in transgenic technology, such as the introduction of robust inducible gene cassettes for expression in intracellular parasite stages or the development of RNA interference methods for down-regulation of parasite gene expression in the host, will further advance our ability to probe host-parasite interactions and unravel disease-promoting mechanisms in the leishmaniases.
Collapse
Affiliation(s)
- L Beattie
- Immunology and Infection Unit, Department of Biology, University of York, Hull York Medical School, Heslington, York, UK
| | - K J Evans
- Immunology and Infection Unit, Department of Biology, University of York, Hull York Medical School, Heslington, York, UK
| | - P M Kaye
- Immunology and Infection Unit, Department of Biology, University of York, Hull York Medical School, Heslington, York, UK
| | - D F Smith
- Immunology and Infection Unit, Department of Biology, University of York, Hull York Medical School, Heslington, York, UK
| |
Collapse
|
34
|
Abstract
Circulating blood monocytes supply peripheral tissues with macrophage and dendritic cell (DC) precursors and, in the setting of infection, also contribute directly to immune defense against microbial pathogens. In humans and mice, monocytes are divided into two major subsets that either specifically traffic into inflamed tissues or, in the absence of overt inflammation, constitutively maintain tissue macrophage/DC populations. Inflammatory monocytes respond rapidly to microbial stimuli by secreting cytokines and antimicrobial factors, express the CCR2 chemokine receptor, and traffic to sites of microbial infection in response to monocyte chemoattractant protein (MCP)-1 (CCL2) secretion. In murine models, CCR2-mediated monocyte recruitment is essential for defense against Listeria monocytogenes, Mycobacterium tuberculosis, Toxoplasma gondii, and Cryptococcus neoformans infection, implicating inflammatory monocytes in defense against bacterial, protozoal, and fungal pathogens. Recent studies indicate that inflammatory monocyte recruitment to sites of infection is complex, involving CCR2-mediated emigration of monocytes from the bone marrow into the bloodstream, followed by trafficking into infected tissues. The in vivo mechanisms that promote chemokine secretion, monocyte differentiation and trafficking, and finally monocyte-mediated microbial killing remain active and important areas of investigation.
Collapse
Affiliation(s)
- Natalya V. Serbina
- Infectious Diseases Service, Department of Medicine, Immunology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| | - Ting Jia
- Infectious Diseases Service, Department of Medicine, Immunology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| | - Tobias M. Hohl
- Infectious Diseases Service, Department of Medicine, Immunology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| | - Eric G. Pamer
- Infectious Diseases Service, Department of Medicine, Immunology Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| |
Collapse
|
35
|
Xia M, Hou C, DeMong DE, Pollack SR, Pan M, Brackley JA, Jain N, Gerchak C, Singer M, Malaviya R, Matheis M, Olini G, Cavender D, Wachter M. Synthesis, structure-activity relationship and in vivo antiinflammatory efficacy of substituted dipiperidines as CCR2 antagonists. J Med Chem 2007; 50:5561-3. [PMID: 17929797 DOI: 10.1021/jm070902b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A series of substituted dipiperidine compounds have been synthesized and identified as selective CCR2 antagonists. Combining the most favorable substituents led to the discovery of remarkably potent CCR2 antagonists displaying IC50 values in the nanomolar range. Compound 7a had outstanding selectivity over CCR1, CCR3, CCR4, CCR5, CCR6, CCR7, and CCR8 and showed excellent efficacy in adjuvant-induced arthritis model, collagen-induced arthritis model, and allergic asthma model.
Collapse
Affiliation(s)
- Mingde Xia
- Drug Discovery, Johnson & Johnson Pharmaceutical Research and Development, L.L.C., 8 Clarke Drive, Cranbury, New Jersey 08512, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Castellani ML, Bhattacharya K, Tagen M, Kempuraj D, Perrella A, De Lutiis M, Boucher W, Conti P, Theoharides TC, Cerulli G, Salini V, Neri G. Anti-chemokine therapy for inflammatory diseases. Int J Immunopathol Pharmacol 2007; 20:447-453. [PMID: 17880758 DOI: 10.1177/039463200702000303] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chemokines are inflammatory proteins acting via G-protein coupled chemokine receptors that trigger different signaling pathways. Monocyte chemoattractant protein-1 (CCL2/MCP-1) and regulated on activation, normal T expressed and secreted (CCL5/RANTES) are the two major members of the CC chemokine beta subfamily. The roles of RANTES and MCP-1 are emerging in regulating the recruitment of inflammatory cells into tissue during inflammation. The inhibition of MCP-1 and RANTES with corresponding antibodies or other inhibitors may provide benefits in different clinical scenarios including cancer, inflammation, CNS disorders, parasitic disease, autoimmune and heart diseases. RANTES and MCP-1 may represent targets for diagnostic procedures and therapeutic intervention, and may be useful as a prognostic factor in the above diseases.
Collapse
Affiliation(s)
- M L Castellani
- Department of Internal Medicine and Science of Ageing, University of Chieti, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol 2007; 82:244-52. [PMID: 17475785 DOI: 10.1189/jlb.0307191] [Citation(s) in RCA: 239] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The term "monocyte" implies a single, homogenous population of cells with uniform physiology. Recent evidence from a number of laboratories indicates that it is likely that blood monocytes may consist of several subpopulations of cells, which differ in size, nuclear morphology, granularity, and functionality. The aim of this review is to give a summary of the new findings in the emerging field of monocyte heterogeneity. We provide a short description of the differentiation patterns of blood monocyte subpopulations, with an emphasis on how these subpopulations can be influenced by infection. We provide a comparison among the main monocyte subpopulations in humans, mice, and rats and illustrate some of the common features of these cells and some of the important interspecies distinctions. We will also discuss the bone marrow precursors of these cells and the differentiation patterns of these subsets in different tissues in response to infection. Most of the data about monocyte trafficking during infection are necessarily derived from murine models, and comparisons between mouse and man must be made with caution. However, these models may provide interesting springboards to permit us to speculate about the topic of monocyte heterogeneity in humans.
Collapse
Affiliation(s)
- Dalit Strauss-Ayali
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, USA
| | | | | |
Collapse
|
38
|
Field AE, Wagage S, Conrad SM, Mosser DM. Reduced pathology following infection with transgenic Leishmania major expressing murine CD40 ligand. Infect Immun 2007; 75:3140-9. [PMID: 17403867 PMCID: PMC1932871 DOI: 10.1128/iai.00160-07] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leishmanization is the inoculation of live Leishmania into the host to vaccinate against subsequent infections. This approach has been largely discontinued due to safety concerns. We have previously shown that combining CD40 ligand (CD40L) with Leishmania antigen preferentially induces a type 1 immune response and provides some protection to vaccinated mice (G. Chen, P. A. Darrah, and D. M. Mosser, Infect. Immun. 69:3255-3263, 2001). In the present study, we developed transgenic L. major organisms which express and secrete the extracellular portion of CD40L (L. major CD40LE). We hypothesized that these organisms would be less virulent but more immunogenic than wild-type organisms and therefore be more effective at leishmanization. Transgenic parasites expressing CD40L mRNA and protein were developed. BALB/c mice infected with these parasites developed significantly smaller lesions containing fewer parasites than animals infected with wild-type organisms. Infection of resistant C57BL/6 mice with low doses of transgenic parasites induced a significant amount of protection against subsequent high-dose infection with wild-type organisms. These results demonstrate that transgenic organisms expressing CD40L are less virulent than wild-type organisms while retaining full immunogenicity.
Collapse
Affiliation(s)
- Ann E Field
- University of Maryland, 1103 Microbiology Building (Building 231), College Park, MD 20742, USA
| | | | | | | |
Collapse
|