1
|
Chen KM, Lan KP, Lai SC. Heme oxygenase-1 modulates brain inflammation and apoptosis in mice with angiostrongyliasis. Parasitol Int 2021; 87:102528. [PMID: 34942361 DOI: 10.1016/j.parint.2021.102528] [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: 05/07/2020] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 10/19/2022]
Abstract
The rat nematode lungworm Angiostrongylus cantonensis undergoes obligatory intracerebral migration in its hosts and causes eosinophilic meningitis or meningoencephalitis. Heme oxygenase 1 (HO-1) has several cytoprotective properties such as anti-oxidative, anti-inflammatory, and anti-apoptotic effects. HO-1 in brain tissues was induced in A. cantonensis-infected group and showed positive modulation in cobalt protoporphyrin (CoPP)-treated groups. Assay methods for the therapeutic effect include western blot analysis, enzyme-linked immunosorbent assay, gelatin zymography, blood-brain barrier permeability evaluation and eosinophil count in cerebrospinal fluid. The combination of albendazole (ABZ) and CoPP significantly decreased pro-inflammatory cytokines, tumor necrosis factor-α, interleukin (IL)-1β, IL-5, and IL-33 but significantly increased anti-inflammatory cytokines IL-10 and transforming growth factor-β. In addition, worm recovery, matrix metalloproteinase-9, BBB permeability, and eosinophil counts were decreased in the ABZ and CoPP co-treated groups. Induction of HO-1 with CoPP strongly inhibited the protein levels of caspase-3 and increased the induction of annexin-V and B-cell leukemia 2. Thus, co-treatment with ABZ and CoPP prevented A. cantonensis-induced eosinophilic meningoencephalitis and its anti-apoptotic effect by promoting HO-1 signaling prior to BBB dysfunction. HO-1 induction might be a therapeutic modality for eosinophilic meningoencephalitis.
Collapse
Affiliation(s)
- Ke-Min Chen
- Department of Parasitology, Chung Shan Medical University, Taichung 402, Taiwan
| | - Kuang-Ping Lan
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Taiwan
| | - Shih-Chan Lai
- Department of Parasitology, Chung Shan Medical University, Taichung 402, Taiwan; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan.
| |
Collapse
|
2
|
Bichiou H, Rabhi S, Ben Hamda C, Bouabid C, Belghith M, Piquemal D, Trentin B, Rabhi I, Guizani-Tabbane L. Leishmania Parasites Differently Regulate Antioxidant Genes in Macrophages Derived From Resistant and Susceptible Mice. Front Cell Infect Microbiol 2021; 11:748738. [PMID: 34722338 PMCID: PMC8554229 DOI: 10.3389/fcimb.2021.748738] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
Macrophage-Leishmania interactions are central to parasite growth and disease outcome. Macrophages have developed various strategies to fight invaders, including oxidative burst. While some microorganisms seem to survive and even thrive in an oxidative environment, others are susceptible and get killed. To counter oxidative stress, macrophages switch the expressions of cytoprotective and detoxifying enzymes, which are downstream targets of the nuclear factor erythroid 2-related factor 2 (Nrf2), to enhance cell survival. We have explored the transcription of NRF2 and of its target genes and compared the effect of the parasite on their transcription in bone marrow-derived macrophages (BMdMs) from Leishmania-resistant and Leishmania-susceptible mice. While heme oxygenase 1 (HO-1) transcription is independent of the genetic background, the transcription of glutathione reductase (Gsr) and of cysteine/glutamate exchange transporter (Slc7a11), involved in glutathione accumulation, was differentially regulated in BMdMs from both mouse strains. We also show that, except for HO-1, known to favor the survival of the parasite, the transcription of the selected genes, including Gsr, CD36, and catalase (CAT), was actively repressed, if not at all time points at least at the later ones, by the parasite, especially in Balb/c BMdMs. Consistent with these results, we found that the silencing of NRF2 in this study increases the survival and multiplication of the parasite.
Collapse
Affiliation(s)
- Haifa Bichiou
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Sameh Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia
| | - Cherif Ben Hamda
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia
| | - Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Meriam Belghith
- Department of Immunology, Institut Pasteur de Tunis, University Tunis El-Manar, Tunis, Tunisia
| | | | | | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia.,Higher Institute of Biotechnology at Sidi-Thabet, Biotechpole Sidi-Thabet, University of Manouba, Sidi-Thabet, Tunisia
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institut Pasteur de Tunis, Tunis-Belvedere, Tunisia
| |
Collapse
|
3
|
Almeida MPO, Mota CM, Mineo TWP, Ferro EAV, Barbosa BF, Silva NM. Heme Oxygenase-1 Induction in Human BeWo Trophoblast Cells Decreases Toxoplasma gondii Proliferation in Association With the Upregulation of p38 MAPK Phosphorylation and IL-6 Production. Front Microbiol 2021; 12:659028. [PMID: 33912151 PMCID: PMC8071940 DOI: 10.3389/fmicb.2021.659028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/09/2021] [Indexed: 11/13/2022] Open
Abstract
Heme oxygenase-1 (HO-1) enzyme exerts beneficial effects at the maternal-fetal interface, especially in trophoblasts, being involved in survival and maturation of these cell phenotypes. Trophoblast cells play essential roles throughout pregnancy, being the gateway for pathogens vertically transmitted, such as Toxoplasma gondii. It was previously shown that HO-1 activity was involved in the control of T. gondii infection in vivo; however, its contribution in trophoblast cells during T. gondii infection, remain undefined. Thus, this study aimed to investigate the influence of HO-1 in T. gondii-infected BeWo and HTR-8/SVneo human trophoblast cells. For this purpose, trophoblast cells were infected and the HO-1 expression was evaluated. T. gondii-infected BeWo cells were treated with hemin or CoPPIX, as inducers of HO-1, or with bilirubin, an end-product of HO-1, and the parasitism was quantified. The involvement of p38 MAPK, a regulator of HO-1, and the cytokine production, were also evaluated. It was found that T. gondii decreased the HO-1 expression in BeWo but not in HTR-8/SVneo cells. When treated with the HO-1 inducers or bilirubin, BeWo cells reduced the parasite proliferation. T. gondii also decreased the p38 MAPK phosphorylation in BeWo cells; on the other hand, HO-1 induction sustained its activation. Finally, the IL-6 production was upregulated by HO-1 induction in T. gondii-infected cells, which was associated with the control of infection.
Collapse
Affiliation(s)
- Marcos Paulo Oliveira Almeida
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Caroline Martins Mota
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo," Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo," Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Bellisa Freitas Barbosa
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
4
|
Bichiou H, Bouabid C, Rabhi I, Guizani-Tabbane L. Transcription Factors Interplay Orchestrates the Immune-Metabolic Response of Leishmania Infected Macrophages. Front Cell Infect Microbiol 2021; 11:660415. [PMID: 33898331 PMCID: PMC8058464 DOI: 10.3389/fcimb.2021.660415] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Leishmaniasis is a group of heterogenous diseases considered as an important public health problem in several countries. This neglected disease is caused by over 20 parasite species of the protozoa belonging to the Leishmania genus and is spread by the bite of a female phlebotomine sandfly. Depending on the parasite specie and the immune status of the patient, leishmaniasis can present a wide spectrum of clinical manifestations. As an obligate intracellular parasite, Leishmania colonize phagocytic cells, mainly the macrophages that orchestrate the host immune response and determine the fate of the infection. Once inside macrophages, Leishmania triggers different signaling pathways that regulate the immune and metabolic response of the host cells. Various transcription factors regulate such immune-metabolic responses and the associated leishmanicidal and inflammatory reaction against the invading parasite. In this review, we will highlight the most important transcription factors involved in these responses, their interactions and their impact on the establishment and the progression of the immune response along with their effect on the physiopathology of the disease.
Collapse
Affiliation(s)
- Haifa Bichiou
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Biotechnology Department, Higher Institute of Biotechnology at Sidi-Thabet (ISBST), Biotechpole Sidi-Thabet- University of Manouba, Tunis, Tunisia
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
| |
Collapse
|
5
|
Cristina Borges Araujo E, Cariaco Y, Paulo Oliveira Almeida M, Patricia Pallete Briceño M, Neto de Sousa JE, Rezende Lima W, Maria Costa-Cruz J, Maria Silva N. Beneficial effects of Strongyloides venezuelensis antigen extract in acute experimental toxoplasmosis. Parasite Immunol 2020; 43:e12811. [PMID: 33247953 DOI: 10.1111/pim.12811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Toxoplasma gondii is a protozoan with worldwide distribution and triggers a strong Th1 immune response in infected susceptible hosts. On the contrary, most helminth infections are characterized by Th2 immune response and the use of helminth-derived antigens to regulate immune response in inflammatory disorders has been broadly investigated. OBJECTIVES The aim of this study was to investigate whether treatment with Strongyloides venezuelensis antigen extract (SvAg) would alter immune response against T gondii. METHODS C57BL/6 mice were orally infected with T gondii and treated with SvAg, and parasitological, histological and immunological parameters were investigated. RESULTS It was observed that SvAg treatment improved survival rates of T gondii-infected mice. At day 7 post-infection, the parasite load was lower in the lung and small intestine of infected SvAg-treated mice than untreated infected mice. Remarkably, SvAg-treated mice infected with T gondii presented reduced inflammatory lesions in the small intestine than infected untreated mice and decreased intestinal and systemic levels of IFN-γ, TNF-α and IL-6. In contrast, SvAg treatment increased T gondii-specific IgA serum levels in infected mice. CONCLUSIONS S venezuelensis antigen extract has anti-parasitic and anti-inflammatory properties during T gondii infection suggesting as a possible alternative to parasite and inflammation control.
Collapse
Affiliation(s)
- Ester Cristina Borges Araujo
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Yusmaris Cariaco
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Marcos Paulo Oliveira Almeida
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | | | - José Eduardo Neto de Sousa
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Wânia Rezende Lima
- Instituto de Biotecnologia, Universidade Federal de Catalão, Rua Terezinha Margon Vaz, s/n Residencial Barka II, Catalão, Brasil
| | - Julia Maria Costa-Cruz
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Neide Maria Silva
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| |
Collapse
|
6
|
Costa DL, Amaral EP, Andrade BB, Sher A. Modulation of Inflammation and Immune Responses by Heme Oxygenase-1: Implications for Infection with Intracellular Pathogens. Antioxidants (Basel) 2020; 9:antiox9121205. [PMID: 33266044 PMCID: PMC7761188 DOI: 10.3390/antiox9121205] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Its expression is induced in response to stress signals such as reactive oxygen species and inflammatory mediators with antioxidant, anti-inflammatory and immunosuppressive consequences for the host. Interestingly, several intracellular pathogens responsible for major human diseases have been shown to be powerful inducers of HO-1 expression in both host cells and in vivo. Studies have shown that this HO-1 response can be either host detrimental by impairing pathogen control or host beneficial by limiting infection induced inflammation and tissue pathology. These properties make HO-1 an attractive target for host-directed therapy (HDT) of the diseases in question, many of which have been difficult to control using conventional antibiotic approaches. Here we review the mechanisms by which HO-1 expression is induced and how the enzyme regulates inflammatory and immune responses during infection with a number of different intracellular bacterial and protozoan pathogens highlighting mechanistic commonalities and differences with the goal of identifying targets for disease intervention.
Collapse
Affiliation(s)
- Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil
- Correspondence: ; Tel.: +55-16-3315-3061
| | - Eduardo P. Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (E.P.A.); (A.S.)
| | - Bruno B. Andrade
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa;
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador 40210-320, Bahia, Brazil
- Curso de Medicina, Faculdade de Tecnologia e Ciências (UniFTC), Salvador 41741-590, Bahia, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate International Universities, Salvador 41770-235, Bahia, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador 40290-000, Bahia, Brazil
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (E.P.A.); (A.S.)
| |
Collapse
|
7
|
Silva RCMC, Travassos LH, Paiva CN, Bozza MT. Heme oxygenase-1 in protozoan infections: A tale of resistance and disease tolerance. PLoS Pathog 2020; 16:e1008599. [PMID: 32692767 PMCID: PMC7373268 DOI: 10.1371/journal.ppat.1008599] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. HO-1 activity deeply impacts host capacity to tolerate infection through reduction of tissue damage or affecting resistance, the ability of the host to control pathogen loads. In this Review, we will discuss the contribution of HO-1 in different and complex protozoan infections, such as malaria, leishmaniasis, Chagas disease, and toxoplasmosis. The complexity of these infections and the pleiotropic effects of HO-1 constitute an interesting area of study and an opportunity for drug development.
Collapse
Affiliation(s)
- Rafael C. M. C. Silva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Leonardo H. Travassos
- Laboratório de Imunoreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia N. Paiva
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
8
|
Oliveira MC, Coutinho LB, Almeida MPO, Briceño MP, Araujo ECB, Silva NM. The Availability of Iron Is Involved in the Murine Experimental Toxoplasma gondii Infection Outcome. Microorganisms 2020; 8:microorganisms8040560. [PMID: 32295126 PMCID: PMC7232304 DOI: 10.3390/microorganisms8040560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/07/2020] [Accepted: 04/11/2020] [Indexed: 02/07/2023] Open
Abstract
Iron is an important constituent of our environment, being necessary for both mammalian and pathogenic protozoa survival. Iron-containing proteins exert a wide range of biological processes such as biodegradation and biosynthesis, as well as immune function, fetal development, and physical and mental well-being. This work aimed to investigate the effect of iron deprivation in Toxoplasma gondii infection outcome. C57BL/6 mice were orally infected with T. gondii and treated with an iron chelator, deferoxamine, or supplemented with iron (ferrous sulfate), and the parasitism as well as immunological and histological parameters were analyzed. It was observed that the infection increased iron accumulation in the organs, as well as systemically, and deferoxamine treatment diminished the iron content in serum samples and intestine. The deferoxamine treatment decreased the parasitism and inflammatory alterations in the small intestine and lung. Additionally, they partially preserved the Paneth cells and decreased the intestinal dysbiosis. The ferrous sulfate supplementation, despite not significantly increasing the parasite load in the organs, increased the inflammatory alterations in the liver. Together, our results suggest that iron chelation, which is commonly used to treat iron overload, could be a promising medicine to control T. gondii proliferation, mainly in the small intestine, and consequently inflammation caused by infection.
Collapse
|
9
|
Luo XH, Liu JZ, Wang B, Men QL, Ju YQ, Yin FY, Zheng C, Li W. KLF14 potentiates oxidative adaptation via modulating HO-1 signaling in castrate-resistant prostate cancer. Endocr Relat Cancer 2019; 26:181-195. [PMID: 30400002 DOI: 10.1530/erc-18-0383] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 09/11/2018] [Indexed: 12/23/2022]
Abstract
Insights into the mechanisms by which key factors stimulate cell growth under androgen-depleted conditions is a premise to the development of effective treatments with clinically significant activity in patients with castration-resistant prostate cancer (CRPC). Herein, we report that, the expression of Krüppel-like factor 14 (KLF14), a master transcription factor in the regulation of lipid metabolism, was significantly induced in castration-insensitive PCa cells and tumor tissues from a mouse xenograft model of CRPC. KLF14 upregulation in PCa cells, which was stimulated upstream by oxidative stress, was dependent on multiple pathways including PI3K/AKT, p42/p44 MAPK, AMPK and PKC pathways. By means of ectopic overexpression and genetic inactivation, we further show that KLF14 promoted cell growth via positive regulation of the antioxidant response under androgen-depleted conditions. Mechanistically, KLF14 coupled to p300 and CBP to enhance the transcriptional activation of HMOX1, the gene encoding the antioxidative enzyme heme oxygenase-1 (HO-1) that is one of the most important mechanisms of cell adaptation to stress. Transient knockdown of HMOX1 is sufficient to overcome KLF14 overexpression-potentiated PCa cell growth under androgen-depleted conditions. From a pharmacological standpoint, in vivo administration of ZnPPIX (a specific inhibitor of HO-1) effectively attenuates castration-resistant progression in the mouse xenograft model, without changing KLF14 level. Together, these results provide comprehensive insight into the KLF14-dependent regulation of antioxidant response and subsequent pathogenesis of castration resistance and indicate that interventions targeting the KLF14/HO-1 adaptive mechanism should be further explored for CRPC treatment.
Collapse
Affiliation(s)
- Xiao-Hui Luo
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Jian-Zhou Liu
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Bo Wang
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Qun-Li Men
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Yu-Quan Ju
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Feng-Yan Yin
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Chao Zheng
- Department of Urology, Baoji Center Hospital, Baoji, Shaanxi Province, People's Republic of China
| | - Wei Li
- Department of Human Anatomy, Histology and Embryology, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
10
|
Xu L, Sang R, Yu Y, Li J, Ge B, Zhang X. The polysaccharide from Inonotus obliquus protects mice from Toxoplasma gondii-induced liver injury. Int J Biol Macromol 2018; 125:1-8. [PMID: 30445083 DOI: 10.1016/j.ijbiomac.2018.11.114] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 10/23/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
The study aimed to explore the protective effects and mechanism of Inonotus obliquus polysaccharide (IOP) on liver injury caused by Toxoplasma gondii (T. gondii) infection in mice. The results showed that treatment with IOP significantly decreased the liver coefficient, the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), malondialdehyde (MDA) and nitric oxide (NO), and increased the contents of antioxidant enzyme superoxide dismutase (SOD) and glutathione (GSH). IOP effectively decreased the expression of serum tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β), interferon-γ (IFN-γ) and interluekin-4 (IL-4) in T. gondii-infected mice. In agreement with these observations, IOP also alleviated hepatic pathological damages caused by T. gondii. Furthermore, we found that IOP down-regulated the levels of toll-like receptor 2 (TLR2) and toll-like receptor 4 (TLR4), phosphorylations of nuclear factor-κappaB (NF-κB) p65 and inhibitor kappaBα (IκBα), whereas up-regulated the expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). These findings suggest that IOP possesses hepatoprotective effects against T. gondii-induced liver injury in mice, and such protection is at least in part due to its anti-inflammatory effects through inhibiting the TLRs/NF-κB signaling axis and the activation of an antioxidant response by inducing the Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Lu Xu
- Department of Animal Medicine, Agricultural College, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Rui Sang
- Department of Animal Medicine, Agricultural College, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Yifan Yu
- Department of Animal Medicine, Agricultural College, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Jinxia Li
- Department of Animal Medicine, Agricultural College, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Bingjie Ge
- Department of Animal Medicine, Agricultural College, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China
| | - Xuemei Zhang
- Department of Animal Medicine, Agricultural College, Yanbian University, Gongyuan Street, Yanji, Jilin 133002, PR China.
| |
Collapse
|
11
|
Nascimento BB, Cartelle CT, Noviello MDL, Pinheiro BV, de Almeida Vitor RW, Souza DDG, de Vasconcelos Generoso S, Cardoso VN, Martins FDS, Nicoli JR, Arantes RME. Influence of indigenous microbiota on experimental toxoplasmosis in conventional and germ-free mice. Int J Exp Pathol 2017; 98:191-202. [PMID: 28895246 DOI: 10.1111/iep.12236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/04/2017] [Indexed: 01/12/2023] Open
Abstract
Toxoplasmosis represents one of the most common zoonoses worldwide. Its agent, Toxoplasma gondii, causes a severe innate pro-inflammatory response. The indigenous intestinal microbiota promotes host animal homoeostasis and may protect the host against pathogens. Germ-free (GF) animals provide an important tool for the study of interactions between host and microbiota. In this study, we assessed the role of indigenous microorganisms in disease development utilizing a murine toxoplasmosis model, which includes conventional (CV) and GF NIH Swiss mice. CV and GF mice orally inoculated with T. gondii had similar survival curves. However, disease developed differently in the two animal groups. In CV mice, intestinal permeability increased and levels of intestinal pro-inflammatory cytokines were altered. In GF animals, there were discrete epithelial degenerative changes and mucosal oedema, but the liver and lungs displayed significant lesions. We conclude that, despite similar survival curves, CV animals succumb to an exaggerated inflammatory response, whereas GF mice fail to produce an adequate systemic response.
Collapse
Affiliation(s)
- Bruna B Nascimento
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Christiane T Cartelle
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria de L Noviello
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Breno V Pinheiro
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo W de Almeida Vitor
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle da G Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Simone de Vasconcelos Generoso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert N Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviano Dos S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacques R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rosa M E Arantes
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
12
|
Carasi P, Rodríguez E, da Costa V, Frigerio S, Brossard N, Noya V, Robello C, Anegón I, Freire T. Heme-Oxygenase-1 Expression Contributes to the Immunoregulation Induced by Fasciola hepatica and Promotes Infection. Front Immunol 2017; 8:883. [PMID: 28798750 PMCID: PMC5526848 DOI: 10.3389/fimmu.2017.00883] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/11/2017] [Indexed: 02/04/2023] Open
Abstract
Fasciola hepatica, also known as the liver fluke, is a trematode that infects livestock and humans causing fasciolosis, a zoonotic disease of increasing importance due to its worldwide distribution and high economic losses. This parasite immunoregulates the host immune system by inducing a strong Th2 and regulatory T immune response by immunomodulating dendritic cell (DC) maturation and alternative activation of macrophages. In this paper, we show that F. hepatica infection in mice induces the upregulation of heme-oxygenase-1 (HO-1), the rate-limiting enzyme in the catabolism of free heme that regulates the host inflammatory response. We show and characterize two different populations of antigen presenting cells that express HO-1 during infection in the peritoneum of infected animals. Cells that expressed high levels of HO-1 expressed intermediate levels of F4/80 but high expression of CD11c, CD38, TGFβ, and IL-10 suggesting that they correspond to regulatory DCs. On the other hand, cells expressing intermediate levels of HO-1 expressed high levels of F4/80, CD68, Ly6C, and FIZZ-1, indicating that they might correspond to alternatively activated macrophages. Furthermore, the pharmacological induction of HO-1 with the synthetic metalloporphyrin CoPP promoted F. hepatica infection increasing the clinical signs associated with the disease. In contrast, treatment with the HO-1 inhibitor SnPP protected mice from parasite infection, indicating that HO-1 plays an essential role during F. hepatica infection. Finally, HO-1 expression during F. hepatica infection was associated with TGFβ and IL-10 levels in liver and peritoneum, suggesting that HO-1 controls the expression of these immunoregulatory cytokines during infection favoring parasite survival in the host. These results contribute to the elucidation of the immunoregulatory mechanisms induced by F. hepatica in the host and provide alternative checkpoints to control fasciolosis.
Collapse
Affiliation(s)
- Paula Carasi
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| | - Ernesto Rodríguez
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| | - Valeria da Costa
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| | - Sofía Frigerio
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| | - Natalie Brossard
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| | - Verónica Noya
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| | - Carlos Robello
- Departamento de Bioquimica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Ignacio Anegón
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Teresa Freire
- Laboratorio de Inmunomodulación y Desarrollo de Vacunas, Facultad de Medicina, Departamento de Inmunobiología, Universidad de República, Montevideo, Uruguay
| |
Collapse
|
13
|
Association of Heme Oxygenase 1 with Lung Protection in Malaria-Associated ALI/ARDS. Mediators Inflamm 2016; 2016:4158698. [PMID: 27974865 PMCID: PMC5126464 DOI: 10.1155/2016/4158698] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/10/2016] [Accepted: 10/18/2016] [Indexed: 12/25/2022] Open
Abstract
Malaria is a serious disease, caused by the parasite of the genus Plasmodium, which was responsible for 440,000 deaths in 2015. Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is one of the main clinical complications in severe malaria. The murine model DBA/2 reproduces the clinical signs of ALI/ARDS in humans, when infected with Plasmodium berghei ANKA. High levels of HO-1 were reported in cases of severe malaria. Our data indicated that the HO-1 mRNA and protein expression are increased in mice that develop malaria-associated ALI/ARDS (MA-ALI/ARDS). Additionally, the hemin, a HO-1 inducing drug, prevented mice from developing MA-ALI/ARDS when administered prior to the development of MA-ALI/ARDS in this model. Also, hemin treatment showed an amelioration of respiratory parameters in mice, high VEGF levels in the sera, and a decrease in vascular permeability in the lung, which are signs of ALI/ARDS. Therefore, the induction of HO-1 before the development of MA-ALI/ARDS could be protective. However, the increased expression of HO-1 on the onset of MA-ALI/ARDS development may represent an effort to revert the phenotype of this syndrome by the host. We therefore confirm that HO-1 inducing drugs could be used for prevention of MA-ALI/ARDS in humans.
Collapse
|
14
|
Scharn CR, Collins AC, Nair VR, Stamm CE, Marciano DK, Graviss EA, Shiloh MU. Heme Oxygenase-1 Regulates Inflammation and Mycobacterial Survival in Human Macrophages during Mycobacterium tuberculosis Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4641-9. [PMID: 27183573 PMCID: PMC4875857 DOI: 10.4049/jimmunol.1500434] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/22/2016] [Indexed: 12/17/2022]
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis, is responsible for 1.5 million deaths annually. We previously showed that M. tuberculosis infection in mice induces expression of the CO-producing enzyme heme oxygenase (HO1) and that CO is sensed by M. tuberculosis to initiate a dormancy program. Further, mice deficient in HO1 succumb to M. tuberculosis infection more readily than do wild-type mice. Although mouse macrophages control intracellular M. tuberculosis infection through several mechanisms, such as NO synthase, the respiratory burst, acidification, and autophagy, how human macrophages control M. tuberculosis infection remains less well understood. In this article, we show that M. tuberculosis induces and colocalizes with HO1 in both mouse and human tuberculosis lesions in vivo, and that M. tuberculosis induces and colocalizes with HO1 during primary human macrophage infection in vitro. Surprisingly, we find that chemical inhibition of HO1 both reduces inflammatory cytokine production by human macrophages and restricts intracellular growth of mycobacteria. Thus, induction of HO1 by M. tuberculosis infection may be a mycobacterial virulence mechanism to enhance inflammation and bacterial growth.
Collapse
Affiliation(s)
- Caitlyn R Scharn
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Angela C Collins
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Vidhya R Nair
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Chelsea E Stamm
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Denise K Marciano
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX 77030; and
| | - Michael U Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
15
|
Macrophage-activating lipopeptide-2 requires Mal and PI3K for efficient induction of heme oxygenase-1. PLoS One 2014; 9:e103433. [PMID: 25077631 PMCID: PMC4117634 DOI: 10.1371/journal.pone.0103433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/29/2014] [Indexed: 11/19/2022] Open
Abstract
AIMS This study is to investigate the mechanisms by which macrophage-activating lipopeptide-2 (MALP-2) induces heme oxygenase (HO)-1, a cytoprotective enzyme that catalyzes the degradation of heme, in human monocytes. METHODS Human monocytic THP-1 cells were cultured for transient transfection with plasmids and stimulation with MALP-2 for indicative time intervals. After incubation with MALP-2, cells were collected and disrupted, before being tested for promoter activity using luciferase assay. For analysis of proteins, immunoreactive bands were detected using an enhanced chemiluminescence Western blotting system, and the band intensity was measured by densitometryic analysis. For the detection of co-immunoprecipitation, SDS-PAGE was performed and the membranes were probed using respective antibodies. To investigate the cellular localization of NF-E2-related factor 2 (Nrf2), cells underwent immunofluorescence staining and confocal microscopy, and were analyzed using electrophoretic mobility shift assay. RESULTS MALP-2-induced HO-1 expression and promoter activity were abrogated by transfection with dominant negative (DN) plasmids of TLR2 and TLR6, or their neutralizing antibodies. However, inhibition of MyD88 or transfection with the DN-MyD88 was insufficient to attenuate HO-1 expression. In contrast, mutation or silencing of MyD88 adapter-like (Mal) by DN-Mal or siRNA almost completely blocked HO-1 induction. Btk, c-Src and PI3K were also involved in MALP-2-induced HO-1 expression, as revealed by specific inhibitors LFM-A13, PP1 and LY294002, or by transfection with siRNA of c-Src. MALP-2-induced activation of PI3K was attenuated by transfection with DN mutant of Mal, and by pretreatment with LFM-A13 or PP1. Furthermore, MALP-2 stimulated the translocation of Nrf2 from the cytosol to the nucleus and Nrf2 binding to the ARE site in the HO-1 promoter, which could also be inhibited by pretreatment with a PI3K inhibitor, LY294002. CONCLUSIONS These results indicated that MALP-2 required TLR2/6, Btk, Mal and c-Src to activate PI3K, which in turn initiated the activation of Nrf2 for efficient HO-1 induction.
Collapse
|