1
|
Aldayel AM, Bosaeed M, Almansour S, Alharbi NK, Alenazi M, Aljami HA, Aldibasi O, Aljouie A, Xu H, Cui Z. IgM has a better relative distribution in inflammation sites and tumor tissues than IgG. J Nanobiotechnology 2025; 23:253. [PMID: 40156022 PMCID: PMC11954317 DOI: 10.1186/s12951-025-03213-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 02/10/2025] [Indexed: 04/01/2025] Open
Abstract
Immunoglobulins (Igs) play a crucial role in host's defense and in developing therapies against inflammatory diseases and cancer. Herein, we first studied the relative distribution of IgM and IgG in mouse models with acute or chronic inflammation. We found that IgM showed a more selective distribution towards inflammation sites than IgG. Similarly, in a tumor-bearing mouse model, IgM showed a higher tumor-to-blood or -to healthy organs ratio than IgG. We hypothesized that the difference in the sizes between IgM and IgG may have contributed to the differences in their relative distribution, which was supported by using an IgG nanoparticle system that was similar to IgM in size. To confirm the findings in clinics, we investigated IgM and IgG levels in the blood and bronchoalveolar lavage fluid (BALF) of patients diagnosed with fungal pneumonia and showed that the relative distribution of IgM was significantly higher than IgG in the BALF samples as compared to that in serum. Such an understanding of our immune system at the nano-level may help us develop more effective biotechnological interventions against inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Abdulaziz M Aldayel
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, 78712, USA.
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia.
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia.
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia.
| | - Mohammad Bosaeed
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Sarah Almansour
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Naif Khalaf Alharbi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia
| | - Mohammed Alenazi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Haya A Aljami
- Infectious Diseases Research Department, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Omar Aldibasi
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Abdulrhman Aljouie
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, 11426, Riyadh, Saudi Arabia
- College of Pharmacy & College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), 11426, Riyadh, Saudi Arabia
| | - Haiyue Xu
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Zhengrong Cui
- College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
2
|
de Oliveira HD, Batista CN, Lima MN, Lima AC, Dos Passos BABR, Freitas RJRX, Silva JD, Xisto DG, Rangel-Ferreira MV, Pelajo M, Rocco PRM, Ribeiro-Gomes FL, de Castro Faria-Neto HC, Maron-Gutierrez T. Acetylsalicylic acid and dihydroartemisinin combined therapy on experimental malaria-associated acute lung injury: analysis of lung function and the inflammatory process. Malar J 2024; 23:285. [PMID: 39300444 DOI: 10.1186/s12936-024-05017-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/16/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Severe malaria can cause respiratory symptoms, which may lead to malaria-acute lung injury (MA-ALI) due to inflammation and damage to the blood-gas barrier. Patients with severe malaria also often present thrombocytopenia, and the use of acetylsalicylic acid (ASA), a commonly used non-steroidal anti-inflammatory drug with immunomodulatory and antiplatelet effects, may pose a risk in regions where malaria is endemic. Thus, this study aimed to investigate the systemic impact of ASA and dihydroartemisinin (DHA) on ALI induced in mice by Plasmodium berghei NK65 (PbNK65). METHODS C57BL/6 mice were randomly divided into control (C) and PbNK65 infected groups and were inoculated with uninfected or 104 infected erythrocytes, respectively. Then, the animals were treated with DHA (3 mg/kg) or vehicle (DMSO) at the 8-day post-infection (dpi) for 7 days and with ASA (100 mg/kg, single dose), and analyses were performed at 9 or 15 dpi. Lung mechanics were performed, and lungs were collected for oedema evaluation and histological analyses. RESULTS PbNK65 infection led to lung oedema, as well as increased lung static elastance (Est, L), resistive (ΔP1, L) and viscoelastic (ΔP2, L) pressures, percentage of mononuclear cells, inflammatory infiltrate, hemorrhage, alveolar oedema, and alveolar thickening septum at 9 dpi. Mice that received DHA or DHA + ASA had an increase in Est, L, and CD36 expression on inflammatory monocytes and higher protein content on bronchoalveolar fluid (BALF). However, only the DHA-treated group presented a percentage of inflammatory monocytes similar to the control group and a decrease in ΔP1, L and ΔP2, L compared to Pb + DMSO. Also, combined treatment with DHA + ASA led to an impairment in diffuse alveolar damage score and lung function at 9 dpi. CONCLUSIONS Therapy with ASA maintained lung morpho-functional impairment triggered by PbNK65 infection, leading to a large influx of inflammatory monocytes to the lung tissue. Based on its deleterious effects in experimental MA-ALI, ASA administration or its treatment maintenance might be carefully reconsidered and further investigated in human malaria cases.
Collapse
Affiliation(s)
- Helena D'Anunciação de Oliveira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Camila Nunes Batista
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Maiara Nascimento Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Ana Carolina Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil
| | | | - Rodrigo Jose Rocha Xavier Freitas
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Johnatas Dutra Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora Gonçalves Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Marcelo Pelajo
- Laboratory of Pathology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Hugo Caire de Castro Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Av. Brasil, 4036 - Bloco 2. Manguinhos, Rio de Janeiro, RJ, 21040-361, Brazil.
| |
Collapse
|
3
|
Zhang M, Wang QR, Hou X, Wang Q, Yang X, Zhou T, Liu X, Wu L, Wang J, Jin X, Liu Z, Huang B. Blockage of mechanosensitive Piezo1 channel alleviates the severity of experimental malaria-associated acute lung injury. Parasit Vectors 2024; 17:46. [PMID: 38303078 PMCID: PMC10832208 DOI: 10.1186/s13071-024-06144-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Malaria-associated acute lung injury (MA-ALI) is a well-recognized clinical complication of severe, complicated malaria that is partly driven by sequestrations of infected red blood cells (iRBCs) on lung postcapillary induced impaired blood flow. In earlier studies the mechanosensitive Piezo1 channel emerged as a regulator of mechanical stimuli, but the function and underlying mechanism of Piezo1 impacting MA-ALI severity via sensing the impaired pulmonary blood flow are still not fully elucidated. Thus, the present study aimed to explore the role of Piezo1 in the severity of murine MA-ALI. METHODS Here, we utilized a widely accepted murine model of MA-ALI using C57BL/6 mice with Plasmodium berghei ANKA infection and then added a Piezo1 inhibitor (GsMTx4) to the model. The iRBC-stimulated Raw264.7 macrophages in vitro were also targeted with GsMTx4 to further explore the potential mechanism. RESULTS Our data showed an elevation in the expression of Piezo1 and number of Piezo1+-CD68+ macrophages in lung tissues of the experimental MA-ALI mice. Compared to the infected control mice, the blockage of Piezo1 with GsMTx4 dramatically improved the survival rate but decreased body weight loss, peripheral blood parasitemia/lung parasite burden, experimental cerebral malaria incidence, total protein concentrations in bronchoalveolar lavage fluid, lung wet/dry weight ratio, vascular leakage, pathological damage, apoptosis and number of CD68+ and CD86+ macrophages in lung tissues. This was accompanied by a dramatic increase in the number of CD206+ macrophages (M2-like subtype), upregulation of anti-inflammatory cytokines (e.g. IL-4 and IL-10) and downregulation of pro-inflammatory cytokines (e.g. TNF-α and IL-1β). In addition, GsMTx4 treatment remarkably decreased pulmonary intracellular iron accumulation, protein level of 4-HNE (an activator of ferroptosis) and the number of CD68+-Piezo1+ and CD68+-4-HNE+ macrophages but significantly increased protein levels of GPX4 (an inhibitor of ferroptosis) in experimental MA-ALI mice. Similarly, in vitro study showed that the administration of GsMTx4 led to a remarkable elevation in the mRNA levels of CD206, IL-4, IL-10 and GPX-4 but to a substantial decline in CD86, TNF-α, IL-1β and 4-HNE in the iRBC-stimulated Raw264.7 cells. CONCLUSIONS Our findings indicated that blockage of Piezo1 with GsMTx4 alleviated the severity of experimental MA-ALI in mice partly by triggering pulmonary macrophage M2 polarization and subsequent anti-inflammatory responses but inhibited apoptosis and ferroptosis in lung tissue. Our data suggested that targeting Piezo1 in macrophages could be a promising therapeutic strategy for treating MA-ALI.
Collapse
Affiliation(s)
- Min Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Qian Ru Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xinpeng Hou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Qi Wang
- Guangzhou Chest Hospital, Guangzhou, 510095, People's Republic of China
| | - Xiaoyan Yang
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, 511400, People's Republic of China
| | - Tingting Zhou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xiaobo Liu
- School of Basic Medical Science, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Lirong Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Jie Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Zhenlong Liu
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.
| | - Bo Huang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
- School of Basic Medical Science, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
4
|
Carpinter BA, Renhe DC, Bellei JCB, Vieira CD, Rodolphi CM, Ferreira MVR, de Freitas CS, Neto AFDS, Coelho EAF, Mietto BDS, Gomes FLR, Rocha VN, Scopel KKG. DHA-rich fish oil plays a protective role against experimental cerebral malaria by controlling inflammatory and mechanical events from infection. J Nutr Biochem 2024; 123:109492. [PMID: 37866427 DOI: 10.1016/j.jnutbio.2023.109492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/17/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Every year, thousands of children, particularly those under 5 years old, die because of cerebral malaria (CM). Following conventional treatment, approximately 25% of surviving individuals have lifelong severe neurocognitive sequelae. Therefore, improved conventional therapies or effective alternative therapies that prevent the severe infection are crucial. Omega-3 (Ω-3) polyunsaturated fatty acids (PUFAs) are known to have antioxidative and anti-inflammatory effects and protect against diverse neurological disorders, including Alzheimer's and Parkinson's diseases. However, little is known regarding the effects of Ω-3 PUFAs against parasitic infections. In this study, C57BL/6 mice received supplemental treatment of a fish oil rich in the Ω-3 PUFA, docosahexaenoic acid (DHA), which was started 15 days prior to infection with Plasmodium berghei ANKA and was maintained until the end of the study. Animals treated with the highest doses of DHA, 3.0 and 6.0 g/kg body weight, had 60 and 80% chance of survival, respectively, while all nontreated mice died by the 7th day postinfection due to CM. Furthermore, the parasite load during the critical period for CM development (5th to 11th day postinfection) was controlled in treated mice. However, after this period all animals developed high levels of parasitemia until the 20th day of infection. DHA treatment also effectively reduced blood-brain barrier (BBB) damage and brain edema and completely prevented brain hemorrhage and vascular occlusion. A strong anti-inflammatory profile was observed in the brains of DHA-treated mice, as well as, an increased number of neutrophil and reduced number of CD8+ T leukocytes in the spleen. Thus, this is the first study to demonstrate that the prophylactic use of DHA-rich fish oil exerts protective effects against experimental CM, reducing the mechanical and immunological events caused by the P. berghei ANKA infection.
Collapse
Affiliation(s)
- Bárbara Albuquerque Carpinter
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela Chaves Renhe
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Jéssica Correa Bezerra Bellei
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina David Vieira
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Cinthia Magalhães Rodolphi
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Camila Simões de Freitas
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno de Siqueira Mietto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Vinicius Novaes Rocha
- Department of Veterinary Medicine, Research Centre of Pathology and Veterinary Histology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| |
Collapse
|
5
|
Zafar I, Taniguchi T, Baghdadi HB, Kondoh D, Rizk MA, Galon EM, Ji S, El-Sayed SAES, Do T, Li H, Amer MM, Zhuowei M, Yihong M, Zhou J, Inoue N, Xuan X. Babesia microti alleviates disease manifestations caused by Plasmodium berghei ANKA in murine co-infection model of complicated malaria. Front Cell Infect Microbiol 2023; 13:1226088. [PMID: 37492527 PMCID: PMC10364126 DOI: 10.3389/fcimb.2023.1226088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Malaria remains one of the most significant health issues worldwide, accounting for 2.6% of the total global disease burden, and efforts to eliminate this threat continue. The key focus is to develop an efficient and long-term immunity to this disease via vaccination or therapeutic approach, and innovative strategies would enable us to achieve this target. Previously, using a mouse co-infection disease model, cross-protection was illustrated between Babesia microti and Plasmodium chabaudi. Hence, this study was planned to elucidate the impact of acute B. microti Peabody mjr and Plasmodium berghei ANKA co-infection on the consequence of complicated malaria in the C57BL/6J mouse model of malaria. Furthermore, immune response and pathological features were analyzed, and the course of the disease was compared among experimental groups. Our study established that acute B. microti infection activated immunity which was otherwise suppressed by P. berghei. The immunosuppressive tissue microenvironment was counteracted as evidenced by the enhanced immune cell population in co-infected mice, in contrast to P. berghei-infected control mice. Parasite sequestration in the brain, liver, lung, and spleen of co-infected mice was significantly decreased and tissue injury was ameliorated. Meanwhile, the serum levels of IFN-γ, TNF-α, and IL-12p70 were reduced while the secretion of IL-10 was promoted in co-infected mice. Eventually, co-infected mice showed an extended rate of survival. Hereby, the principal cytokines associated with the severity of malaria by P. berghei infection were TNF-α, IFN-γ, and IL-12p70. Moreover, it was evident from our flow cytometry results that innate immunity is crucial and macrophages are at the frontline of immunity against P. berghei infection. Our study recommended further investigations to shed light on the effects of babesiosis in suppressing malaria with the goal of developing Babesia-based therapy against malaria.
Collapse
Affiliation(s)
- Iqra Zafar
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Livestock and Dairy Development Department, Veterinary Research Institute, Lahore, Punjab, Pakistan
| | - Tomoyo Taniguchi
- Department of Immunology and Parasitology, Graduate School of Medicine University of the Ryukyus, Nishihara Cho, Japan
| | - Hanadi B. Baghdadi
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Center (BASRC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Daisuke Kondoh
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Mohamed Abdo Rizk
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Eloiza May Galon
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Indang, Cavite, Philippines
| | - Shengwei Ji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Shimaa Abd El-Salam El-Sayed
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Thom Do
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Hang Li
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Moaz M. Amer
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ma Zhuowei
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ma Yihong
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Jinlin Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Noboru Inoue
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| |
Collapse
|
6
|
Martin C, Clift S, Leisewitz A. Lung pathology of natural Babesia rossi infection in dogs. J S Afr Vet Assoc 2023; 94:59-69. [PMID: 37358318 DOI: 10.36303/jsava.523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
A proportion of Babesia rossi infections in dogs are classified as complicated and one of the most lethal complications is acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Most dogs that die succumb within 24 hours of presentation. The pulmonary pathology caused by B. rossi in dogs has not been described. The aim of this study was to provide a thorough macroscopic, histological and immunohistochemical description of the lung changes seen in dogs naturally infected with B. rossi that succumbed to the infection. Death was invariably accompanied by alveolar oedema. Histopathology showed acute interstitial pneumonia characterised by alveolar oedema and haemorrhages, with increased numbers of mononuclear leucocytes in alveolar walls and lumens. Intra-alveolar polymerised fibrin aggregates were observed in just over half the infected cases. Immunohistochemistry showed increased numbers of MAC387- and CD204-reactive monocyte-macrophages in alveolar walls and lumens, and increased CD3-reactive T-lymphocytes in alveolar walls, compared with controls. These histological features overlap to some extent (but far from perfectly) with the histological pattern of lung injury referred to as the exudative stage of diffuse alveolar damage (DAD) as is quite commonly reported in ALI/ARDS.
Collapse
Affiliation(s)
- C Martin
- Idexx Laboratories (Pty) Ltd, South Africa
| | - S Clift
- Section of Pathology, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, South Africa
| | - A Leisewitz
- Department of Clinical Sciences, Bailey Small Animal Teaching Hospital, Auburn University College of Veterinary Medicine, United States of America and Section of Small Animal Medicine, Companion Animal Clinical Sciences, Faculty of Veterinary Science, University of Pretoria, South Africa
| |
Collapse
|
7
|
He Z, Yu C, Pan Z, Li X, Zhang X, Huang Q, Liao X, Hu J, Zeng F, Ru L, Yu W, Xu Q, Song J, Liang J. Erythrocyte membrane with CLIPPKF as biomimetic nanodecoy traps merozoites and attaches to infected red blood cells to prevent Plasmodium infection. J Nanobiotechnology 2023; 21:15. [PMID: 36647056 PMCID: PMC9841648 DOI: 10.1186/s12951-022-01709-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 11/14/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Malaria remains a serious threat to global public health. With poor efficacies of vaccines and the emergence of drug resistance, novel strategies to control malaria are urgently needed. RESULTS We developed erythrocyte membrane-camouflaged nanoparticles loaded with artemether based on the growth characteristics of Plasmodium. The nanoparticles could capture the merozoites to inhibit them from repeatedly infecting normal erythrocytes, owing to the interactions between merozoites and heparin-like molecules on the erythrocyte membrane. Modification with a phosphatidylserine-targeting peptide (CLIPPKF) improved the drug accumulation in infected red blood cells (iRBCs) from the externalized phosphatidylserine induced by Plasmodium infection. In Plasmodium berghei ANKA strain (pbANKA)-infected C57BL/6 mice, the nanoparticles significantly attenuated Plasmodium-induced inflammation, apoptosis, and anemia. We observed reduced weight variation and prolonged survival time in pbANKA-challenged mice, and the nanoparticles showed good biocompatibility and negligible cytotoxicity. CONCLUSION Erythrocyte membrane-camouflaged nanoparticles loaded with artemether were shown to provide safe and effective protection against Plasmodium infection.
Collapse
Affiliation(s)
- Zhouqing He
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Chuyi Yu
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Ziyi Pan
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Xiaobo Li
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Xiangxiang Zhang
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Qijing Huang
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Xingcheng Liao
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Jiaoting Hu
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Feng Zeng
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Li Ru
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Wanlin Yu
- grid.413402.00000 0004 6068 0570Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120 China
| | - Qin Xu
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Jianping Song
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Jianming Liang
- grid.411866.c0000 0000 8848 7685Artemisinin Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China ,grid.8547.e0000 0001 0125 2443Key Laboratory of Smart Drug Delivery, School of Pharmacy, Ministry of Education, Fudan University, Shanghai, 201203 China
| |
Collapse
|
8
|
Olatunde AC, Cornwall DH, Roedel M, Lamb TJ. Mouse Models for Unravelling Immunology of Blood Stage Malaria. Vaccines (Basel) 2022; 10:1525. [PMID: 36146602 PMCID: PMC9501382 DOI: 10.3390/vaccines10091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria comprises a spectrum of disease syndromes and the immune system is a major participant in malarial disease. This is particularly true in relation to the immune responses elicited against blood stages of Plasmodium-parasites that are responsible for the pathogenesis of infection. Mouse models of malaria are commonly used to dissect the immune mechanisms underlying disease. While no single mouse model of Plasmodium infection completely recapitulates all the features of malaria in humans, collectively the existing models are invaluable for defining the events that lead to the immunopathogenesis of malaria. Here we review the different mouse models of Plasmodium infection that are available, and highlight some of the main contributions these models have made with regards to identifying immune mechanisms of parasite control and the immunopathogenesis of malaria.
Collapse
Affiliation(s)
| | | | | | - Tracey J. Lamb
- Department of Pathology, University of Utah, Emma Eccles Jones Medical Research Building, 15 N Medical Drive E, Room 1420A, Salt Lake City, UT 84112, USA
| |
Collapse
|
9
|
Babatunde KA, Adenuga OF. Neutrophils in malaria: A double-edged sword role. Front Immunol 2022; 13:922377. [PMID: 35967409 PMCID: PMC9367684 DOI: 10.3389/fimmu.2022.922377] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human peripheral blood. They form the first line of defense against invading foreign pathogens and might play a crucial role in malaria. According to World Health Organization (WHO), malaria is a globally significant disease caused by protozoan parasites from the Plasmodium genus, and it's responsible for 627,000 deaths in 2020. Neutrophils participate in the defense response against the malaria parasite via phagocytosis and reactive oxygen species (ROS) production. Neutrophils might also be involved in the pathogenesis of malaria by the release of toxic granules and the release of neutrophil extracellular traps (NETs). Intriguingly, malaria parasites inhibit the anti-microbial function of neutrophils, thus making malaria patients more susceptible to secondary opportunistic Salmonella infections. In this review, we will provide a summary of the role of neutrophils during malaria infection, some contradicting mouse model neutrophil data and neutrophil-related mechanisms involved in malaria patients' susceptibility to bacterial infection.
Collapse
Affiliation(s)
- Kehinde Adebayo Babatunde
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | | |
Collapse
|
10
|
Nguee SYT, Júnior JWBD, Epiphanio S, Rénia L, Claser C. Experimental Models to Study the Pathogenesis of Malaria-Associated Acute Respiratory Distress Syndrome. Front Cell Infect Microbiol 2022; 12:899581. [PMID: 35677654 PMCID: PMC9168995 DOI: 10.3389/fcimb.2022.899581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Malaria-associated acute respiratory distress syndrome (MA-ARDS) is increasingly gaining recognition as a severe malaria complication because of poor prognostic outcomes, high lethality rate, and limited therapeutic interventions. Unfortunately, invasive clinical studies are challenging to conduct and yields insufficient mechanistic insights. These limitations have led to the development of suitable MA-ARDS experimental mouse models. In patients and mice, MA-ARDS is characterized by edematous lung, along with marked infiltration of inflammatory cells and damage of the alveolar-capillary barriers. Although, the pathogenic pathways have yet to be fully understood, the use of different experimental mouse models is fundamental in the identification of mediators of pulmonary vascular damage. In this review, we discuss the current knowledge on endothelial activation, leukocyte recruitment, leukocyte induced-endothelial dysfunction, and other important findings, to better understand the pathogenesis pathways leading to endothelial pulmonary barrier lesions and increased vascular permeability. We also discuss how the advances in imaging techniques can contribute to a better understanding of the lung lesions induced during MA-ARDS, and how it could aid to monitor MA-ARDS severity.
Collapse
Affiliation(s)
- Samantha Yee Teng Nguee
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Sabrina Epiphanio
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Carla Claser
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- *Correspondence: Carla Claser,
| |
Collapse
|
11
|
Imai T, Ngo-Thanh H, Suzue K, Shimo A, Nakamura A, Horiuchi Y, Hisaeda H, Murakami T. Live Vaccination with Blood-Stage Plasmodium yoelii 17XNL Prevents the Development of Experimental Cerebral Malaria. Vaccines (Basel) 2022; 10:vaccines10050762. [PMID: 35632518 PMCID: PMC9145751 DOI: 10.3390/vaccines10050762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
In our work, we aim to develop a malaria vaccine with cross-strain (-species) protection. C57BL/6 mice infected with the P. berghei ANKA strain (PbA) develop experimental cerebral malaria (ECM). In contrast, ECM development is inhibited in infected mice depleted of T cells. The clinical applications of immune-cell depletion are limited due to the benefits of host defense against infectious diseases. Therefore, in the present study we attempted to develop a new method for preventing ECM without immune cell depletion. We demonstrated that mice inoculated with a heterologous live-vaccine of P. yoelii 17XNL were able to prevent both ECM and lung pathology and survived longer than control mice when challenged with PbA. Live vaccination protected blood–organ barriers from PbA infection. Meanwhile, live vaccination conferred sterile protection against homologous challenge with the P. yoelii 17XL virulent strain for the long-term. Analysis of the immune response induced by live vaccination showed that cross-reactive antibodies against PbA antigens were generated. IL-10, which has an immunosuppressive effect, was strongly induced in mice challenged with PbA, unlike the pro-inflammatory cytokine IFNγ. These results suggest that the protective effect of heterologous live vaccination against ECM development results from IL-10-mediated host protection.
Collapse
Affiliation(s)
- Takashi Imai
- Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (H.N.-T.); (K.S.)
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
- Correspondence: ; Tel.: +81-49-276-1166
| | - Ha Ngo-Thanh
- Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (H.N.-T.); (K.S.)
- National Hospital for Tropical Disease, 78 Giai Phong, Dong Da, Hanoi 10000, Vietnam
| | - Kazutomo Suzue
- Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (H.N.-T.); (K.S.)
| | - Aoi Shimo
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| | - Akihiro Nakamura
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| | - Yutaka Horiuchi
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo 162-0052, Japan;
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| |
Collapse
|
12
|
Possemiers H, Pham TT, Coens M, Pollenus E, Knoops S, Noppen S, Vandermosten L, D’haese S, Dillemans L, Prenen F, Schols D, Franke-Fayard B, Van den Steen PE. Skeleton binding protein-1-mediated parasite sequestration inhibits spontaneous resolution of malaria-associated acute respiratory distress syndrome. PLoS Pathog 2021; 17:e1010114. [PMID: 34843584 PMCID: PMC8659713 DOI: 10.1371/journal.ppat.1010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/09/2021] [Accepted: 11/15/2021] [Indexed: 11/18/2022] Open
Abstract
Malaria is a hazardous disease caused by Plasmodium parasites and often results in lethal complications, including malaria-associated acute respiratory distress syndrome (MA-ARDS). Parasite sequestration in the microvasculature is often observed, but its role in malaria pathogenesis and complications is still incompletely understood. We used skeleton binding protein-1 (SBP-1) KO parasites to study the role of sequestration in experimental MA-ARDS. The sequestration-deficiency of these SBP-1 KO parasites was confirmed with bioluminescence imaging and by measuring parasite accumulation in the lungs with RT-qPCR. The SBP-1 KO parasites induced similar lung pathology in the early stage of experimental MA-ARDS compared to wildtype (WT) parasites. Strikingly, the lung pathology resolved subsequently in more than 60% of the SBP-1 KO infected mice, resulting in prolonged survival despite the continuous presence of the parasite. This spontaneous disease resolution was associated with decreased inflammatory cytokine expression measured by RT-qPCR and lower expression of cytotoxic markers in pathogenic CD8+ T cells in the lungs of SBP-1 KO infected mice. These data suggest that SBP-1-mediated parasite sequestration and subsequent high parasite load are not essential for the development of experimental MA-ARDS but inhibit the resolution of the disease. Malaria is still a severe global disease with more than 200 million cases and 400 000 deaths each year. Plasmodium falciparum is the species responsible for most malaria deaths globally. The propensity of these parasites to sequester in peripheral vascular beds is assumed to play an important role in disease severity and mortality. Although sequestration has been observed in lungs of malaria patients, its role in the pathogenesis of MA-ARDS, a severe lung complication in malaria, was previously unknown. Therefore, we used sequestration-deficient SBP-1 KO Plasmodium berghei NK65 parasites to study the role of sequestration in experimental MA-ARDS. We observed that MA-ARDS developed similarly in WT and SBP-1 KO infected mice, but the majority of SBP-1 KO-infected mice were able to resolve the lung pathology despite the continuous presence of the parasite. This coincided with a prolonged survival, a decrease in inflammatory cytokine expression and lower expression of cytotoxicity markers in pathogenic CD8+ T cells. These results give important new insights in the role of parasite sequestration in malaria pathology.
Collapse
Affiliation(s)
- Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Thao-Thy Pham
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
- Currently at Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Belgium
| | - Marion Coens
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Sam Noppen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Sigrid D’haese
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
- Currently at Neuro-Aging & Viro-Immunotherapy (NAVI), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Luna Dillemans
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
| | | | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical research, KU Leuven, Belgium
- * E-mail:
| |
Collapse
|
13
|
PET Imaging of Translocator Protein as a Marker of Malaria-Associated Lung Inflammation. Infect Immun 2021; 89:e0002421. [PMID: 34251290 DOI: 10.1128/iai.00024-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose. Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a severe complication of malaria despite effective anti-malarial treatment. Currently, non-invasive imaging procedures such as chest X-rays are used to assess oedema in established MA-ARDS but earlier detection methods are needed to reduce morbidity and mortality. The early stages of MA-ARDS are characterized by the infiltration of leukocytes, in particular monocyte/macrophages, thus monitoring of immune infiltrates may provide a useful indicator of early pathology. Procedures. Plasmodium berghei ANKA-infected C57BL/6 mice, a rodent malaria model of MA-ARDS, were longitudinally imaged using the TSPO imaging agent [18F]FEPPA as a marker of macrophage accumulation during the development of pathology and response to combined artesunate and chloroquine diphosphate therapy (ART+CQ). [18F]FEPPA uptake was compared to blood parasitemia levels and pulmonary immune cell infiltrates using flow cytometry. Results. Infected animals showed rapid increases lung retention of [18F]FEPPA, correlating well with increases in blood parasitemia and pulmonary accumulation of interstitial inflammatory macrophages and MHC II+ alveolar macrophages. Treatment with ART+CQ therapy abrogated this increase in parasitemia and significantly reduced both lung uptake of [18F]FEPPA and macrophage infiltrates. Conclusions. Retention of [18F]FEPPA in the lungs is well correlated with changes in blood parasitemia and lung associated macrophages during disease progression and in response to ART+CQ therapy. With further development TSPO biomarkers may have the potential to be able to accurately assess early onset of MA-ARDS.
Collapse
|
14
|
Platelet α-granules contribute to organ-specific pathologies in a mouse model of severe malaria. Blood Adv 2021; 4:1-8. [PMID: 31891656 DOI: 10.1182/bloodadvances.2019000773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022] Open
Abstract
Key PointsNbeal2 deficiency leads to significantly reduced lung and brain pathology and enhanced survival in a mouse model of malaria. Both antibody-dependent and antibody-independent platelet depletion in mice recapitulate the findings observed in Nbeal2−/− mice.
Collapse
|
15
|
Mejia P, Treviño-Villarreal JH, De Niz M, Meibalan E, Longchamp A, Reynolds JS, Turnbull LB, Opoka RO, Roussilhon C, Spielmann T, Ozaki CK, Heussler VT, Seydel KB, Taylor TE, John CC, Milner DA, Marti M, Mitchell JR. Adipose tissue parasite sequestration drives leptin production in mice and correlates with human cerebral malaria. SCIENCE ADVANCES 2021; 7:7/13/eabe2484. [PMID: 33762334 PMCID: PMC7990332 DOI: 10.1126/sciadv.abe2484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/04/2021] [Indexed: 05/03/2023]
Abstract
Circulating levels of the adipokine leptin are linked to neuropathology in experimental cerebral malaria (ECM), but its source and regulation mechanism remain unknown. Here, we show that sequestration of infected red blood cells (iRBCs) in white adipose tissue (WAT) microvasculature increased local vascular permeability and leptin production. Mice infected with parasite strains that fail to sequester in WAT displayed reduced leptin production and protection from ECM. WAT sequestration and leptin induction were lost in CD36KO mice; however, ECM susceptibility revealed sexual dimorphism. Adipocyte leptin was regulated by the mechanistic target of rapamycin complex 1 (mTORC1) and blocked by rapamycin. In humans, although Plasmodium falciparum infection did not increase circulating leptin levels, iRBC sequestration, tissue leptin production, and mTORC1 activity were positively correlated with CM in pediatric postmortem WAT. These data identify WAT sequestration as a trigger for leptin production with potential implications for pathogenesis of malaria infection, prognosis, and treatment.
Collapse
Affiliation(s)
- Pedro Mejia
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | | | - Mariana De Niz
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Elamaran Meibalan
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Justin S Reynolds
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Lindsey B Turnbull
- Department of Pediatric Infectious Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Robert O Opoka
- Department of Pediatrics and Child Health, Makerere University, Kampala, Uganda
| | | | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - C Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Karl B Seydel
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, USA
| | - Terrie E Taylor
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, USA
| | - Chandy C John
- Department of Pediatric Infectious Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Danny A Milner
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- American Society for Clinical Pathology, Chicago, IL, USA
| | - Matthias Marti
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
16
|
Abstract
Respiratory distress (RD) is a complication of severe malaria associated with a particularly high risk for death in African children infected with the parasite Plasmodium falciparum. The pathophysiology underlying RD remains poorly understood, and the condition is managed supportively. Respiratory distress in severe malaria is associated with high mortality, but its pathogenesis remains unclear. The malaria pigment hemozoin (HZ) is abundant in target organs of severe malaria, including the lungs, and is known to be a potent innate immune activator of phagocytes. We hypothesized that HZ might also stimulate lung epithelial activation and thereby potentiate lung inflammation. We show here that airway epithelium stimulated with HZ undergoes global transcriptional reprogramming and changes in cell surface protein expression that comprise an epithelial activation phenotype. Proinflammatory signaling is induced, and key cytoadherence molecules are upregulated, including several associated with severe malaria, such as CD36 and ICAM1. Epithelial and extracellular matrix remodeling pathways are transformed, including induction of key metalloproteases and modulation of epithelial junctions. The overall program induced by HZ serves to promote inflammation and neutrophil transmigration, and is recapitulated in a murine model of HZ-induced acute pneumonitis. Together, our data demonstrate a direct role for hemozoin in stimulating epithelial activation that could potentiate lung inflammation in malaria.
Collapse
|
17
|
Abstract
Parasitic diseases, such as sleeping sickness, Chagas disease and malaria, remain a major cause of morbidity and mortality worldwide, but particularly in tropical, developing countries. Controlling these diseases requires a better understanding of host-parasite interactions, including a deep appreciation of parasite distribution in the host. The preferred accumulation of parasites in some tissues of the host has been known for many years, but recent technical advances have allowed a more systematic analysis and quantifications of such tissue tropisms. The functional consequences of tissue tropism remain poorly studied, although it has been associated with important aspects of disease, including transmission enhancement, treatment failure, relapse and clinical outcome. Here, we discuss current knowledge of tissue tropism in Trypanosoma infections in mammals, describe potential mechanisms of tissue entry, comparatively discuss relevant findings from other parasitology fields where tissue tropism has been extensively investigated, and reflect on new questions raised by recent discoveries and their potential impact on clinical treatment and disease control strategies.
Collapse
Affiliation(s)
- Sara Silva Pereira
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa , Lisbon , Portugal
| | - Sandra Trindade
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa , Lisbon , Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa , Lisbon , Portugal
| | - Luisa M Figueiredo
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa , Lisbon , Portugal
| |
Collapse
|
18
|
Endothelial Protein C Receptor Could Contribute to Experimental Malaria-Associated Acute Respiratory Distress Syndrome. J Immunol Res 2019; 2019:3105817. [PMID: 31871954 PMCID: PMC6913256 DOI: 10.1155/2019/3105817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/26/2019] [Accepted: 10/16/2019] [Indexed: 12/27/2022] Open
Abstract
The severity of Plasmodium falciparum malaria is associated with parasite cytoadherence, but there is limited knowledge about the effect of parasite cytoadherence in malaria-associated acute respiratory distress syndrome (ARDS). Our objective was to evaluate the cytoadherence of infected red blood cells (iRBCs) in a murine model of ARDS and to appraise a potential function of endothelial protein C receptor (EPCR) in ARDS pathogenesis. DBA/2 mice infected with P. berghei ANKA were classified as ARDS- or hyperparasitemia- (HP-) developing mice according to respiratory parameters and parasitemia. Lungs, blood, and bronchoalveolar lavage were collected for gene expression or protein analyses. Primary cultures of microvascular lung endothelial cells from DBA/2 mice were analyzed for iRBC interactions. Lungs from ARDS-developing mice showed evidence of iRBC accumulation along with an increase in EPCR and TNF concentrations. Furthermore, TNF increased iRBC adherence in vitro. Dexamethasone-treated infected mice showed low levels of TNF and EPCR mRNA expression and, finally, decreased vascular permeability, thus protecting mice from ARDS. In conclusion, we identified that increased iRBC cytoadherence in the lungs underlies malaria-associated ARDS in DBA/2-infected mice and that inflammation increased cytoadherence capacity, suggesting a participation of EPCR and a conceivable target for drug development.
Collapse
|
19
|
Claser C, Nguee SYT, Balachander A, Wu Howland S, Becht E, Gunasegaran B, Hartimath SV, Lee AWQ, Theng Theng Ho J, Bing Ong C, Newell EW, Goggi J, Guan Ng L, Renia L. Lung endothelial cell antigen cross-presentation to CD8 +T cells drives malaria-associated lung injury. Nat Commun 2019; 10:4241. [PMID: 31534124 PMCID: PMC6751193 DOI: 10.1038/s41467-019-12017-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/15/2019] [Indexed: 01/08/2023] Open
Abstract
Malaria-associated acute respiratory distress syndrome (ARDS) and acute lung injury (ALI) are life-threatening manifestations of severe malaria infections. The pathogenic mechanisms that lead to respiratory complications, such as vascular leakage, remain unclear. Here, we confirm that depleting CD8+T cells with anti-CD8β antibodies in C57BL/6 mice infected with P. berghei ANKA (PbA) prevent pulmonary vascular leakage. When we transfer activated parasite-specific CD8+T cells into PbA-infected TCRβ-/- mice (devoid of all T-cell populations), pulmonary vascular leakage recapitulates. Additionally, we demonstrate that PbA-infected erythrocyte accumulation leads to lung endothelial cell cross-presentation of parasite antigen to CD8+T cells in an IFNγ-dependent manner. In conclusion, pulmonary vascular damage in ALI is a consequence of IFNγ-activated lung endothelial cells capturing, processing, and cross-presenting malaria parasite antigen to specific CD8+T cells induced during infection. The mechanistic understanding of the immunopathogenesis in malaria-associated ARDS and ALI provide the basis for development of adjunct treatments.
Collapse
Affiliation(s)
- Carla Claser
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore.
| | - Samantha Yee Teng Nguee
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2 Blk MD4, Level 3, Singapore, 117545, Singapore
| | - Akhila Balachander
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Shanshan Wu Howland
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Etienne Becht
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Bavani Gunasegaran
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Siddesh V Hartimath
- Isotopic Molecular Imaging Laboratory, Singapore Bioimaging Consortium (SBIC), A*STAR, 11 Biopolis Way, #02-02 Helios, Singapore, 138667, Singapore
| | - Audrey W Q Lee
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Jacqueline Theng Theng Ho
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Chee Bing Ong
- Histolopathology/Advanced Molecular Pathology Lab, Institute of Molecular and Cell Biology (IMCB), A*STAR, 61 Biopolis Drive, Level 6 Proteos Building, Singapore, 138673, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Julian Goggi
- Isotopic Molecular Imaging Laboratory, Singapore Bioimaging Consortium (SBIC), A*STAR, 11 Biopolis Way, #02-02 Helios, Singapore, 138667, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore
| | - Laurent Renia
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Level 3 & 4 Immunos Building, Singapore, 138648, Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2 Blk MD4, Level 3, Singapore, 117545, Singapore.
| |
Collapse
|
20
|
Kraisin S, Verhenne S, Pham TT, Martinod K, Tersteeg C, Vandeputte N, Deckmyn H, Vanhoorelbeke K, Van den Steen PE, De Meyer SF. von Willebrand factor in experimental malaria-associated acute respiratory distress syndrome. J Thromb Haemost 2019; 17:1372-1383. [PMID: 31099973 PMCID: PMC9906160 DOI: 10.1111/jth.14485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/02/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a lethal complication of severe malaria, characterized by marked pulmonary inflammation. Patient studies have suggested a link between von Willebrand factor (VWF) and malaria severity. OBJECTIVES To investigate the role of VWF in the pathogenesis of experimental MA-ARDS. METHODS Plasmodium berghei NK65-E (PbNK65) parasites were injected in Vwf+/+ and Vwf-/- mice. Pathological parameters were assessed following infection. RESULTS In accordance with patients with severe malaria, plasma VWF levels were increased and ADAMTS13 activity levels were reduced in experimental MA-ARDS. ADAMTS13- and plasmin-independent reductions of high molecular weight VWF multimers were observed at the end stage of disease. Thrombocytopenia was VWF-independent because it was observed in both Vwf+/+ and Vwf-/- mice. Interestingly, Vwf-/- mice had a shorter survival time compared with Vwf+/+ controls following PbNK65 infection. Lung edema could not explain this shortened survival because alveolar protein levels in Vwf-/- mice were approximately two times lower than in Vwf+/+ controls. Parasite load, on the other hand, was significantly increased in Vwf-/- mice compared with Vwf+/+ mice in both peripheral blood and lung tissue. In addition, anemia was only observed in PbNK65-infected Vwf-/- mice. Of note, Vwf-/- mice presented with two times more reticulocytes, a preferential target of the parasites. CONCLUSIONS This study suggests that parasite load together with malarial anemia, rather than alveolar leakage, might contribute to shortened survival in PbNK65-infected Vwf-/- mice. VWF deficiency is associated with early reticulocytosis following PbNK65 infection, which potentially explains the increase in parasite load.
Collapse
Affiliation(s)
- Sirima Kraisin
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Sebastien Verhenne
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Thao-Thy Pham
- Laboratory of Immunoparasitology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Nele Vandeputte
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Hans Deckmyn
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| |
Collapse
|
21
|
Dromparis P, Aboelnazar NS, Wagner S, Himmat S, White CW, Hatami S, Luc JGY, Rotich S, Freed DH, Nagendran J, Mengel M, Adam BA. Ex vivo perfusion induces a time- and perfusate-dependent molecular repair response in explanted porcine lungs. Am J Transplant 2019; 19:1024-1036. [PMID: 30230229 DOI: 10.1111/ajt.15123] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 01/25/2023]
Abstract
Ex vivo lung perfusion (EVLP) shows promise in ameliorating pretransplant acute lung injury (ALI) and expanding the donor organ pool, but the mechanisms of ex vivo repair remain poorly understood. We aimed to assess the utility of gene expression for characterizing ALI during EVLP. One hundred sixty-nine porcine lung samples were collected in vivo (n = 25), after 0 (n = 11) and 12 (n = 11) hours of cold static preservation (CSP), and after 0 (n = 57), 6 (n = 8), and 12 (n = 57) hours of EVLP, utilizing various ventilation and perfusate strategies. The expression of 53 previously described ALI-related genes was measured and correlated with function and histology. Twenty-eight genes were significantly upregulated and 6 genes downregulated after 12 hours of EVLP. Aggregate gene sets demonstrated differential expression with EVLP (P < .001) but not CSP. Upregulated 28-gene set expression peaked after 6 hours of EVLP, whereas downregulated 6-gene set expression continued to decline after 12 hours. Cellular perfusates demonstrated a greater reduction in downregulated 6-gene set expression vs acellular perfusate (P < .038). Gene set expression correlated with relevant functional and histologic parameters, including P/F ratio (P < .001) and interstitial inflammation (P < .005). Further studies with posttransplant results are warranted to evaluate the clinical significance of this novel molecular approach for assessing organ quality during EVLP.
Collapse
Affiliation(s)
- Peter Dromparis
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Nader S Aboelnazar
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Siegfried Wagner
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Sayed Himmat
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Christopher W White
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Sanaz Hatami
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Jessica G Y Luc
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Silas Rotich
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Jayan Nagendran
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Benjamin A Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
22
|
Abstract
Neutrophils are abundant in the circulation and are one of the immune system's first lines of defense against infection. There has been substantial work carried out investigating the role of neutrophils in malaria and it is clear that during infection neutrophils are activated and are capable of clearing malaria parasites by a number of mechanisms. This review focuses on neutrophil responses to human malarias, summarizing evidence which helps us understand where neutrophils are, what they are doing, how they interact with parasites as well as their potential role in vaccine mediated immunity. We also outline future research priorities for these, the most abundant of leukocytes.
Collapse
Affiliation(s)
- Elizabeth H Aitken
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Agersew Alemu
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Chin VK, Asyran AMY, Zakaria ZA, Abdullah WO, Chong PP, Nordin N, Ibraheem ZO, Majid RA, Basir R. TREM-1 modulation produces positive outcome on the histopathology and cytokines release profile of Plasmodium berghei-infected mice. J Parasit Dis 2018; 43:139-153. [PMID: 30956457 DOI: 10.1007/s12639-018-1070-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/03/2018] [Indexed: 11/25/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 1 (TREM-1) is a potential molecular therapeutic target for various inflammatory diseases. Despite that, the role of TREM-1 during malaria pathogenesis remains obscure with present literature suggesting a link between TREM-1 with severe malaria development. Therefore, this study aims to investigate the role of TREM-1 and TREM-1 related drugs during severe malaria infection in Plasmodium berghei-infected mice model. Our findings revealed that TREM-1 concentration was significantly increased throughout the infection periods and TREM-1 was positively correlated with malaria parasitemia development. This suggests a positive involvement of TREM-1 in severe malaria development. Meanwhile, blocking of TREM-1 activation using rmTREM-1/Fc and TREM-1 clearance by mTREM-1/Ab had significantly reduced malaria parasitemia and suppressed the production of pro- inflammatory cytokines (TNF-α, IL-6 and IFN-γ) and anti-inflammatory cytokine (IL-10). Furthermore, histopathological analysis of TREM-1 related drug treatments, in particular rmTREM-1/Fc showed significant improvements in the histological conditions of major organs (kidneys, spleen, lungs, liver and brain) of Plasmodium berghei-infected mice. This study showed that modulation of TREM-1 released during malaria infection produces a positive outcome on malaria infection through inhibition of pro-inflammatory cytokines secretion and alleviation of histopathological conditions of affected organs. Nevertheless, further investigation on its optimal dosage and dose dependant study should be carried out to maximise its full potential as immunomodulatory or as an adjuvant in line with current antimalarial agents.
Collapse
Affiliation(s)
- Voon Kin Chin
- 2School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, No 1, Jalan Taylor's, 47500 Subang Jaya, Selangor Malaysia
| | - Afiq Mohd Yusof Asyran
- 1Pharmacology Unit, Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Zainul Amiruddin Zakaria
- 4Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Wan Omar Abdullah
- 5Department of Medical Sciences, Faculty of Medicine and Health Sciences, Universiti Sains Islam Malaysia, 55100 Pandan Indah, Kuala Lumpur Malaysia
| | - Pei Pei Chong
- 2School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, No 1, Jalan Taylor's, 47500 Subang Jaya, Selangor Malaysia
| | - Norshariza Nordin
- 4Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Zaid Osamah Ibraheem
- 1Pharmacology Unit, Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Roslaini Abdul Majid
- 3Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Rusliza Basir
- 1Pharmacology Unit, Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| |
Collapse
|
24
|
Niewold P, Cohen A, van Vreden C, Getts DR, Grau GE, King NJC. Experimental severe malaria is resolved by targeting newly-identified monocyte subsets using immune-modifying particles combined with artesunate. Commun Biol 2018; 1:227. [PMID: 30564748 PMCID: PMC6292940 DOI: 10.1038/s42003-018-0216-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/31/2018] [Indexed: 12/29/2022] Open
Abstract
Current treatment of severe malaria and associated cerebral malaria (CM) and respiratory distress syndromes are directed primarily at the parasite. Targeting the parasite has only partial efficacy in advanced infection, as neurological damage and respiratory distress are due to accumulation of host blood cells in the brain microvasculature and lung interstitium. Here, computational analysis identifies Ly6Clo monocytes as a major component of the immune infiltrate in both organs in a preclinical mouse model. Specifically targeting Ly6Clo monocyte precursors, identified by adoptive transfer, with immune-modifying particles (IMP) prevents experimental CM (ECM) in 50% of Plasmodium berghei ANKA-infected mice in early treatment protocols. Furthermore, treatment at onset of clinical ECM with 2 doses of a novel combination of IMP and anti-malarial drug artesunate results in 88% survival. This combination confers protection against ECM and mortality in late stage severe experimental malaria and provides a viable advance on current treatment regimens.
Collapse
Affiliation(s)
- Paula Niewold
- 1Viral Immunopathology, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Amy Cohen
- 2Vascular Immunology Unit, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Caryn van Vreden
- 3Sydney Cytometry, The University of Sydney and The Centenary Institute, Camperdown, NSW 2050 Australia
| | - Daniel R Getts
- 4Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA.,TcR2, Therapeutics, 100 Binney Street, Suite 710, Cambridge, MA 02142 USA
| | - Georges E Grau
- 2Vascular Immunology Unit, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Nicholas J C King
- 1Viral Immunopathology, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia.,3Sydney Cytometry, The University of Sydney and The Centenary Institute, Camperdown, NSW 2050 Australia
| |
Collapse
|
25
|
Pereira MLM, Marinho CRF, Epiphanio S. Could Heme Oxygenase-1 Be a New Target for Therapeutic Intervention in Malaria-Associated Acute Lung Injury/Acute Respiratory Distress Syndrome? Front Cell Infect Microbiol 2018; 8:161. [PMID: 29868517 PMCID: PMC5964746 DOI: 10.3389/fcimb.2018.00161] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/26/2018] [Indexed: 01/17/2023] Open
Abstract
Malaria is a serious disease and was responsible for 429,000 deaths in 2015. Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is one of the main clinical complications of severe malaria; it is characterized by a high mortality rate and can even occur after antimalarial treatment when parasitemia is not detected. Rodent models of ALI/ARDS show similar clinical signs as in humans when the rodents are infected with murine Plasmodium. In these models, it was shown that the induction of the enzyme heme oxygenase 1 (HO-1) is protective against severe malaria complications, including cerebral malaria and ALI/ARDS. Increased lung endothelial permeability and upregulation of VEGF and other pro-inflammatory cytokines were found to be associated with malaria-associated ALI/ARDS (MA-ALI/ARDS), and both were reduced after HO-1 induction. Additionally, mice were protected against MA-ALI/ARDS after treatment with carbon monoxide- releasing molecules or with carbon monoxide, which is also released by the HO-1 activity. However, high HO-1 levels in inflammatory cells were associated with the respiratory burst of neutrophils and with an intensification of inflammation during episodes of severe malaria in humans. Here, we review the main aspects of HO-1 in malaria and ALI/ARDS, presenting the dual role of HO-1 and possibilities for therapeutic intervention by modulating this important enzyme.
Collapse
Affiliation(s)
- Marcelo L M Pereira
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Claudio R F Marinho
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Vandermosten L, Pham TT, Possemiers H, Knoops S, Van Herck E, Deckers J, Franke-Fayard B, Lamb TJ, Janse CJ, Opdenakker G, Van den Steen PE. Experimental malaria-associated acute respiratory distress syndrome is dependent on the parasite-host combination and coincides with normocyte invasion. Malar J 2018; 17:102. [PMID: 29506544 PMCID: PMC5839036 DOI: 10.1186/s12936-018-2251-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
Background Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a complication of malaria with a lethality rate of up to 80% despite anti-malarial treatment. It is characterized by a vast infiltration of leukocytes, microhaemorrhages and vasogenic oedema in the lungs. Previously, a mouse model for MA-ARDS was developed by infection of C57BL/6 mice with the Edinburgh line NK65-E of Plasmodium berghei. Results Here, both host and parasite factors were demonstrated to play crucial roles in the development and severity of lung pathology. In particular, the genetic constitution of the host was an important determinant in the development of MA-ARDS. Both male and female C57BL/6, but not BALB/c, mice developed MA-ARDS when infected with P. berghei NK65-E. However, the New York line of P. berghei NK65 (NK65-NY) did not induce demonstrable MA-ARDS, despite its accumulation in the lungs and fat tissue to a similar or even higher extent as P. berghei NK65-E. These two commonly used lines of P. berghei differ in their red blood cell preference. P. berghei NK65-NY showed a stronger predilection for reticulocytes than P. berghei NK65-E and this appeared to be associated with a lower pathogenicity in the lungs. The pulmonary pathology in the C57BL/6/P. berghei NK65-E model was more pronounced than in the model with infection of DBA/2 mice with P. berghei strain ANKA. The transient lung pathology in DBA/2 mice infected with P. berghei ANKA coincided with the infection phase in which parasites mainly infected normocytes. This phase was followed by a less pathogenic phase in which P. berghei ANKA mainly infected reticulocytes. Conclusions The propensity of mice to develop MA-ARDS during P. berghei infection depends on both host and parasite factors and appears to correlate with RBC preference. These data provide insights in induction of MA-ARDS and may guide the choice of different mouse-parasite combinations to study lung pathology.
Collapse
Affiliation(s)
- Leen Vandermosten
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Thao-Thy Pham
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Hendrik Possemiers
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Evelien Van Herck
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Julie Deckers
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium.,Laboratory of Immunoregulation, VIB Center for Inflammation Research, Department of Internal Medicine, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Tracey J Lamb
- Department of Pathology, University of Utah, 15 N Medical Drive E, Salt Lake City, UT, 84112, USA
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Philippe E Van den Steen
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium.
| |
Collapse
|
27
|
A single rapamycin dose protects against late-stage experimental cerebral malaria via modulation of host immunity, endothelial activation and parasite sequestration. Malar J 2017; 16:455. [PMID: 29121917 PMCID: PMC5679345 DOI: 10.1186/s12936-017-2092-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/27/2017] [Indexed: 01/05/2023] Open
Abstract
Background Maladaptive immune responses during cerebral malaria (CM) result in high mortality despite opportune anti-malarial chemotherapy. Rapamycin, an FDA-approved immunomodulator, protects against experimental cerebral malaria (ECM) in mice through effects on the host. However, the potential for reduced adaptive immunity with chronic use, combined with an incomplete understanding of mechanisms underlying protection, limit translational potential as an adjunctive therapy in CM. Results The results presented herein demonstrate that a single dose of rapamycin, provided as late as day 4 or 5 post-infection, protected mice from ECM neuropathology and death through modulation of distinct host responses to infection. Rapamycin prevented parasite cytoadherence in peripheral organs, including white adipose tissue, via reduction of CD36 expression. Rapamycin also altered the splenic immune response by reducing the number of activated T cells with migratory phenotype, while increasing local cytotoxic T cell activation. Finally, rapamycin reduced brain endothelial ICAM-1 expression concomitant with reduced brain pathology. Together, these changes potentially contributed to increased parasite elimination while reducing CD8 T cell migration to the brain. Conclusions Rapamycin exerts pleotropic effects on host immunity, vascular activation and parasite sequestration that rescue mice from ECM, and thus support the potential clinical use of rapamycin as an adjunctive therapy in CM.
Collapse
|
28
|
Totino PR, Lopes SC. Insights into the Cytoadherence Phenomenon of Plasmodium vivax: The Putative Role of Phosphatidylserine. Front Immunol 2017; 8:1148. [PMID: 28979260 PMCID: PMC5611623 DOI: 10.3389/fimmu.2017.01148] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022] Open
Abstract
Plasmodium vivax is the most geographically widespread and the dominant human malaria parasite in most countries outside of sub-Saharan Africa and, although it was classically recognized to cause benign infection, severe cases and deaths caused by P. vivax have remarkably been reported. In contrast to Plasmodium falciparum, which well-known ability to bind to endothelium and placental tissue and form rosettes is related to severity of the disease, it has been a dogma that P. vivax is unable to undergo cytoadherent phenomena. However, some studies have demonstrated that red blood cells (RBCs) infected by P. vivax can cytoadhere to host cells, while the molecules participating in this host–parasite interaction are still a matter of speculation. In the present overview, we address the evidences currently supporting the adhesive profile of P. vivax and, additionally, discuss the putative role of phosphatidylserine—a cell membrane phospholipid with cytoadhesive properties that has been detected on the surface of Plasmodium-parasitized RBCs.
Collapse
Affiliation(s)
- Paulo Renato Totino
- Laboratory of Malaria Research, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
29
|
Craig JM, Scott AL. Antecedent Nippostrongylus infection alters the lung immune response to Plasmodium berghei. Parasite Immunol 2017; 39. [PMID: 28475238 DOI: 10.1111/pim.12441] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 04/25/2017] [Indexed: 12/13/2022]
Abstract
In endemic regions, it is not uncommon for patients to be co-infected with soil-transmitted helminths and malaria. Although both malaria and many helminth species use the lungs as a site of development, little attention has been paid to the impact that pulmonary immunity induced by one parasite has on the lung response to the other. To model the consequences of a prior hookworm exposure on the development of immunity to malaria in the lungs, mice were infected with Nippostrongylus brasiliensis and 2 weeks later challenged with Plasmodium berghei. We found that a pre-existing hookworm-induced type 2 immune environment had a measurable but modest impact on the nature of the malaria-driven type 1 cytokine response in the lungs that was associated with a transient effect on parasite development and no significant changes in morbidity and mortality after malaria infection. However, prior hookworm infection did have a lasting effect on lung macrophages, where the malaria-induced M1-like response was blunted by previous M2 polarization. These results demonstrate that, although helminth parasites confer robust changes to the immunological status of the pulmonary microenvironment, lung immunity is plastic and capable of rapidly adapting to consecutive heterologous infections.
Collapse
Affiliation(s)
- J M Craig
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - A L Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
30
|
Cunningham DA, Lin JW, Brugat T, Jarra W, Tumwine I, Kushinga G, Ramesar J, Franke-Fayard B, Langhorne J. ICAM-1 is a key receptor mediating cytoadherence and pathology in the Plasmodium chabaudi malaria model. Malar J 2017; 16:185. [PMID: 28468674 PMCID: PMC5415785 DOI: 10.1186/s12936-017-1834-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/25/2017] [Indexed: 02/07/2023] Open
Abstract
Background Parasite cytoadherence within the microvasculature of tissues and organs of infected individuals is implicated in the pathogenesis of several malaria syndromes. Multiple host receptors may mediate sequestration. The identity of the host receptor(s), or the parasite ligand(s) responsible for sequestration of Plasmodium species other than Plasmodium falciparum is largely unknown. The rodent malaria parasites may be useful to model interactions of parasite species, which lack the var genes with their respective hosts, as other multigene families are shared between the species. The role of the endothelial receptors ICAM-1 and CD36 in cytoadherence and in the development of pathology was investigated in a Plasmodium chabaudi infection in C57BL/6 mice lacking these receptors. The schizont membrane-associated cytoadherence (SMAC) protein of Plasmodium berghei has been shown to exhibit reduced CD36-associated cytoadherence in P. berghei ANKA-infected mice. Methods Parasite tissue sequestration and the development of acute stage pathology in P. chabaudi infections of mice lacking CD36 or ICAM-1, their respective wild type controls, and in infections with mutant P. chabaudi parasites lacking the smac gene were compared. Peripheral blood parasitaemia, red blood cell numbers and weight change were monitored throughout the courses of infection. Imaging of bioluminescent parasites in isolated tissues (spleen, lungs, liver, kidney and gut) was used to measure tissue parasite load. Results This study shows that neither the lack of CD36 nor the deletion of the smac gene from P. chabaudi significantly impacted on acute-stage pathology or parasite sequestration. By contrast, in the absence of ICAM-1, infected animals experience less anaemia and weight loss, reduced parasite accumulation in both spleen and liver and higher peripheral blood parasitaemia during acute stage malaria. The reduction in parasite tissue sequestration in infections of ICAM-1 null mice is maintained after mosquito transmission. Conclusions These results indicate that ICAM-1-mediated cytoadherence is important in the P. chabaudi model of malaria and suggest that for rodent malarias, as for P. falciparum, there may be multiple host and parasite molecules involved in sequestration. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1834-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Jing-Wen Lin
- The Francis Crick Institute, London, NW1 1AT, UK
| | | | | | | | | | - Jai Ramesar
- Leiden Malaria Research Group, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | | |
Collapse
|
31
|
Thylur RP, Wu X, Gowda NM, Punnath K, Neelgund SE, Febbraio M, Gowda DC. CD36 receptor regulates malaria-induced immune responses primarily at early blood stage infection contributing to parasitemia control and resistance to mortality. J Biol Chem 2017; 292:9394-9408. [PMID: 28416609 DOI: 10.1074/jbc.m117.781294] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/12/2017] [Indexed: 12/31/2022] Open
Abstract
In malaria, CD36 plays several roles, including mediating parasite sequestration to host organs, phagocytic clearance of parasites, and regulation of immunity. Although the functions of CD36 in parasite sequestration and phagocytosis have been clearly defined, less is known about its role in malaria immunity. Here, to understand the function of CD36 in malaria immunity, we studied parasite growth, innate and adaptive immune responses, and host survival in WT and Cd36-/- mice infected with a non-lethal strain of Plasmodium yoelii Compared with Cd36-/- mice, WT mice had lower parasitemias and were resistant to death. At early but not at later stages of infection, WT mice had higher circulatory proinflammatory cytokines and lower anti-inflammatory cytokines than Cd36-/- mice. WT mice showed higher frequencies of proinflammatory cytokine-producing and lower frequencies of anti-inflammatory cytokine-producing dendritic cells (DCs) and natural killer cells than Cd36-/- mice. Cytokines produced by co-cultures of DCs from infected mice and ovalbumin-specific, MHC class II-restricted α/β (OT-II) T cells reflected CD36-dependent DC function. WT mice also showed increased Th1 and reduced Th2 responses compared with Cd36-/- mice, mainly at early stages of infection. Furthermore, in infected WT mice, macrophages and neutrophils expressed higher levels of phagocytic receptors and showed enhanced phagocytosis of parasite-infected erythrocytes than those in Cd36-/- mice in an IFN-γ-dependent manner. However, there were no differences in malaria-induced humoral responses between WT and Cd36-/- mice. Overall, the results show that CD36 plays a significant role in controlling parasite burden by contributing to proinflammatory cytokine responses by DCs and natural killer cells, Th1 development, phagocytic receptor expression, and phagocytic activity.
Collapse
Affiliation(s)
- Ramesh P Thylur
- From the Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Xianzhu Wu
- From the Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Nagaraj M Gowda
- From the Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Kishore Punnath
- From the Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Shivayogeeshwara E Neelgund
- From the Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Maria Febbraio
- the Department of Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - D Channe Gowda
- From the Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| |
Collapse
|
32
|
Roussilhon C, Bang G, Bastaert F, Solhonne B, Garcia-Verdugo I, Peronet R, Druilhe P, Sakuntabhai A, Mecheri S, Sallenave JM. The antimicrobial molecule trappin-2/elafin has anti-parasitic properties and is protective in vivo in a murine model of cerebral malaria. Sci Rep 2017; 7:42243. [PMID: 28181563 PMCID: PMC5299836 DOI: 10.1038/srep42243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/05/2017] [Indexed: 11/18/2022] Open
Abstract
According to the WHO, and despite reduction in mortality rates, there were an estimated 438 000 malaria deaths in 2015. Therefore new antimalarials capable of limiting organ damage are still required. We show that systemic and lung adenovirus (Ad)-mediated over-expression of trappin-2 (T-2) an antibacterial molecule with anti-inflammatory activity, increased mice survival following infection with the cerebral malaria-inducing Plasmodium berghei ANKA (PbANKA) strain. Systemically, T-2 reduced PbANKA sequestration in spleen, lung, liver and brain, associated with a decrease in pro-inflammatory cytokines (eg TNF-α in spleen and lung) and an increase in IL-10 production in the lung. Similarly, local lung instillation of Ad-T-2 resulted in a reduced organ parasite sequestration and a shift towards an anti-inflammatory/repair response, potentially implicating monocytes in the protective phenotype. Relatedly, we demonstrated in vitro that human monocytes incubated with Plasmodium falciparum-infected red blood cells (Pf-iRBCs) and IgGs from hyper-immune African human sera produced T-2 and that the latter colocalized with merozoites and inhibited Pf multiplication. This array of data argues for the first time for the potential therapeutic usefulness of this host defense peptide in human malaria patients, with the aim to limit acute lung injury and respiratory distress syndrom often observed during malaria episodes.
Collapse
Affiliation(s)
- Christian Roussilhon
- Unité de génétique fonctionnelle des maladies infectieuses and CNRS Unité de recherche associée 3012; Paris, 75015, France
| | - Gilles Bang
- Unité de génétique fonctionnelle des maladies infectieuses and CNRS Unité de recherche associée 3012; Paris, 75015, France
| | - Fabien Bastaert
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| | - Brigitte Solhonne
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| | - Ignacio Garcia-Verdugo
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| | - Roger Peronet
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- INSERM U1201, Paris F-75015, France
| | | | - Anavaj Sakuntabhai
- Unité de génétique fonctionnelle des maladies infectieuses and CNRS Unité de recherche associée 3012; Paris, 75015, France
| | - Salaheddine Mecheri
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- INSERM U1201, Paris F-75015, France
| | - Jean-Michel Sallenave
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| |
Collapse
|
33
|
Zin NM, Baba MS, Zainal-Abidin AH, Latip J, Mazlan NW, Edrada-Ebel R. Gancidin W, a potential low-toxicity antimalarial agent isolated from an endophytic Streptomyces SUK10. Drug Des Devel Ther 2017; 11:351-363. [PMID: 28223778 PMCID: PMC5308589 DOI: 10.2147/dddt.s121283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Endophytic Streptomyces strains are potential sources for novel bioactive molecules. In this study, the diketopiperazine gancidin W (GW) was isolated from the endophytic actinobacterial genus Streptomyces, SUK10, obtained from the bark of Shorea ovalis tree, and it was tested in vivo against Plasmodium berghei PZZ1/100. GW exhibited an inhibition rate of nearly 80% at 6.25 and 3.125 μg kg-1 body weight on day four using the 4-day suppression test method on male ICR strain mice. Comparing GW at both concentrations with quinine hydrochloride and normal saline as positive and negative controls, respectively, 50% of the mice treated with 3.125 μg kg-1 body weight managed to survive for more than 11 months after infection, which almost reached the life span of normal mice. Biochemical tests of selected enzymes and proteins in blood samples of mice treated with GW were also within normal levels; in addition, no abnormalities or injuries were found on internal vital organs. These findings indicated that this isolated bioactive compound from Streptomyces SUK10 exhibits very low toxicity and is a good candidate for potential use as an antimalarial agent in an animal model.
Collapse
Affiliation(s)
- Noraziah Mohamad Zin
- Programme of Biomedical Science, School of Diagnostic and Applied Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur
| | - Mohd Shukri Baba
- Department of Biomedical Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan
| | | | - Jalifah Latip
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi
| | - Noor Wini Mazlan
- Analytical and Environmental Chemistry, School of Marine and Environmental Sciences, Universiti Malaysia Terengganu, Kuala Terengganu, Malaysia
| | - RuAngelie Edrada-Ebel
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
34
|
Sercundes MK, Ortolan LS, Debone D, Soeiro-Pereira PV, Gomes E, Aitken EH, Neto AC, Russo M, D' Império Lima MR, Alvarez JM, Portugal S, Marinho CRF, Epiphanio S. Targeting Neutrophils to Prevent Malaria-Associated Acute Lung Injury/Acute Respiratory Distress Syndrome in Mice. PLoS Pathog 2016; 12:e1006054. [PMID: 27926944 PMCID: PMC5142790 DOI: 10.1371/journal.ppat.1006054] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/10/2016] [Indexed: 12/28/2022] Open
Abstract
Malaria remains one of the greatest burdens to global health, causing nearly 500,000 deaths in 2014. When manifesting in the lungs, severe malaria causes acute lung injury/acute respiratory distress syndrome (ALI/ARDS). We have previously shown that a proportion of DBA/2 mice infected with Plasmodium berghei ANKA (PbA) develop ALI/ARDS and that these mice recapitulate various aspects of the human syndrome, such as pulmonary edema, hemorrhaging, pleural effusion and hypoxemia. Herein, we investigated the role of neutrophils in the pathogenesis of malaria-associated ALI/ARDS. Mice developing ALI/ARDS showed greater neutrophil accumulation in the lungs compared with mice that did not develop pulmonary complications. In addition, mice with ALI/ARDS produced more neutrophil-attracting chemokines, myeloperoxidase and reactive oxygen species. We also observed that the parasites Plasmodium falciparum and PbA induced the formation of neutrophil extracellular traps (NETs) ex vivo, which were associated with inflammation and tissue injury. The depletion of neutrophils, treatment with AMD3100 (a CXCR4 antagonist), Pulmozyme (human recombinant DNase) or Sivelestat (inhibitor of neutrophil elastase) decreased the development of malaria-associated ALI/ARDS and significantly increased mouse survival. This study implicates neutrophils and NETs in the genesis of experimentally induced malaria-associated ALI/ARDS and proposes a new therapeutic approach to improve the prognosis of severe malaria.
Collapse
Affiliation(s)
- Michelle K. Sercundes
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Luana S. Ortolan
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Daniela Debone
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Eliane Gomes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Elizabeth H. Aitken
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Antonio Condino Neto
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Momtchilo Russo
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Maria R. D' Império Lima
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - José M. Alvarez
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Silvia Portugal
- Center of Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Claudio R. F. Marinho
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Sabrina Epiphanio
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
35
|
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: 2.9] [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
|
36
|
Liu J, Huang S, Su XZ, Song J, Lu F. Blockage of Galectin-receptor Interactions by α-lactose Exacerbates Plasmodium berghei-induced Pulmonary Immunopathology. Sci Rep 2016; 6:32024. [PMID: 27554340 PMCID: PMC4995515 DOI: 10.1038/srep32024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022] Open
Abstract
Malaria-associated acute lung injury (ALI) is a frequent complication of severe malaria that is often caused by "excessive" immune responses. To better understand the mechanism of ALI in malaria infection, here we investigated the roles of galectin (Gal)-1, 3, 8, 9 and the receptors of Gal-9 (Tim-3, CD44, CD137, and PDI) in malaria-induced ALI. We injected alpha (α)-lactose into mice-infected with Plasmodium berghei ANKA (PbANKA) to block galectins and found significantly elevated total proteins in bronchoalveolar lavage fluid, higher parasitemia and tissue parasite burden, and increased numbers of CD68(+) alveolar macrophages as well as apoptotic cells in the lungs after blockage. Additionally, mRNA levels of Gal-9, Tim-3, CD44, CD137, and PDI were significantly increased in the lungs at day 5 after infection, and the levels of CD137, IFN-α, IFN-β, IFN-γ, IL-4, and IL-10 in the lungs were also increased after α-lactose treatment. Similarly, the levels of Gal-9, Tim-3, IFN-α, IFN-β, IFN-γ, and IL-10 were all significantly increased in murine peritoneal macrophages co-cultured with PbANKA-infected red blood cells in vitro; but only IFN-α and IFN-β were significantly increased after α-lactose treatment. Our data indicate that Gal-9 interaction with its multiple receptors play an important role in murine malaria-associated ALI.
Collapse
Affiliation(s)
- Jinfeng Liu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China
| | - Shiguang Huang
- School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States of America.,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Jianping Song
- Institute of Science and Technology, Guangzhou University of Chinese Medicine, 436 Chentai Road, Baiyun District, Guangzhou 510445, Guangdong, China
| | - Fangli Lu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China
| |
Collapse
|
37
|
de Azevedo-Quintanilha IG, Vieira-de-Abreu A, Ferreira AC, Nascimento DO, Siqueira AM, Campbell RA, Teixeira Ferreira TP, Gutierrez TM, Ribeiro GM, E Silva PMR, Carvalho AR, Bozza PT, Zimmerman GA, Castro-Faria-Neto HC. Integrin αDβ2 (CD11d/CD18) mediates experimental malaria-associated acute respiratory distress syndrome (MA-ARDS). Malar J 2016; 15:393. [PMID: 27473068 PMCID: PMC4967320 DOI: 10.1186/s12936-016-1447-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 07/20/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a potentially lethal complication of clinical malaria. Acute lung injury in MA-ARDS shares features with ARDS triggered by other causes, including alveolar inflammation and increased alveolar-capillary permeability, leading to leak of protein-rich pulmonary oedema fluid. Mechanisms and physiologic alterations in MA-ARDS can be examined in murine models of this syndrome. Integrin αDβ2 is a member of the leukocyte, or β2 (CD18), sub-family of integrins, and emerging observations indicate that it has important activities in leukocyte adhesion, accumulation and signalling. The goal was to perform analysis of the lungs of mice wild type C57Bl/6 (a D (+/+) ) and Knockout C57Bl/6 (a D (-/-) ) with malaria-associated acute lung injury to better determine the relevancy of the murine models and investigate the mechanism of disease. METHODS C57BL/6 wild type (a D (+/+) ) and deficient for CD11d sub-unit (a D (-/-) ) mice were monitored after infection with 10(5) Plasmodium berghei ANKA. CD11d subunit expression RNA was measured by real-time polymerase chain reaction, vascular barrier integrity by Evans blue dye (EBD) exclusion and cytokines by ELISA. Protein and leukocytes were measured in bronchoalveolar lavage fluid (BALF) samples. Tissue cellularity was measured by the point-counting technique, F4/80 and VCAM-1 expression by immunohistochemistry. Respiratory function was analysed by non-invasive BUXCO and mechanical ventilation. RESULTS Alveolar inflammation, vascular and interstitial accumulation of monocytes and macrophages, and disrupted alveolar-capillary barrier function with exudation of protein-rich pulmonary oedema fluid were present in P. berghei-infected wild type mice and were improved in αDβ2-deficient animals. Key pro-inflammatory cytokines were also decreased in lung tissue from α D (-/-) mice, providing a mechanistic explanation for reduced alveolar-capillary inflammation and leak. CONCLUSIONS The results indicate that αDβ2 is an important inflammatory effector molecule in P. berghei-induced MA-ARDS, and that leukocyte integrins regulate critical inflammatory and pathophysiologic events in this model of complicated malaria. Genetic deletion of integrin subunit αD in mice, leading to deficiency of integrin αDβ2, alters lung inflammation and acute lung injury in a mouse model of MA-ARDS caused by P. berghei.
Collapse
Affiliation(s)
- Isaclaudia G de Azevedo-Quintanilha
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil.
| | - Adriana Vieira-de-Abreu
- Program in Molecular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.
| | - André Costa Ferreira
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil
| | - Daniele O Nascimento
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil
| | - Alessandra M Siqueira
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil
| | - Robert A Campbell
- Program in Molecular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Tatiana P Teixeira Ferreira
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Rio de Janeiro, Brazil
| | - Tatiana M Gutierrez
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil
| | - Gabriel M Ribeiro
- Laboratório de Engenharia Pulmonar no Programa de Engenharia Biomédica, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia-COPPE/Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia M R E Silva
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Rio de Janeiro, Brazil
| | - Alysson R Carvalho
- Laboratório de Fisiologia da Respiração, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia T Bozza
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil
| | - Guy A Zimmerman
- Program in Molecular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Hugo C Castro-Faria-Neto
- Laboratório de Immunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Pavilhão Ozório de Almeida, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil.,Programa de Produtividade Científica, Universidade Estácio de Sá, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
38
|
Brugat T. Editorial: CD36: Russian roulette of host and parasites during malaria infection. J Leukoc Biol 2016; 99:643-5. [PMID: 27130887 DOI: 10.1189/jlb.4ce1115-494r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/23/2015] [Indexed: 11/24/2022] Open
Affiliation(s)
- Thibaut Brugat
- Mill Hill Laboratory, Francis Crick Institute, London, United Kingdom http://www.twitter.com/@TheCrick
| |
Collapse
|
39
|
Barker KR, Conroy AL, Hawkes M, Murphy H, Pandey P, Kain KC. Biomarkers of hypoxia, endothelial and circulatory dysfunction among climbers in Nepal with AMS and HAPE: a prospective case-control study. J Travel Med 2016; 23:taw005. [PMID: 26984355 PMCID: PMC5731443 DOI: 10.1093/jtm/taw005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mechanisms underlying acute mountain sickness (AMS) and high-altitude pulmonary edema (HAPE) are not fully understood. We hypothesized that regulators of endothelial function, circulatory homeostasis, hypoxia and cell stress contribute to the pathobiology of AMS and HAPE. METHODS We conducted a prospective case-control study of climbers developing altitude illness who were evacuated to the CIWEC clinic in Kathmandu, compared to healthy acclimatized climbers. ELISA was used to measure plasma biomarkers of the above pathways. RESULTS Of the 175 participants, there were 71 cases of HAPE, 54 cases of AMS and 50 acclimatized controls (ACs). Markers of endothelial function were associated with HAPE: circulating levels of endothelin-1 (ET-1) were significantly elevated and levels of sKDR (soluble kinase domain receptor) were significantly decreased in cases of HAPE compared to AC or AMS. ET-1 levels were associated with disease severity as indicated by oxygen saturation. Angiopoietin-like 4 (Angptl4) and resistin, a marker of cell stress, were associated with AMS and HAPE irrespective of severity. Corin and angiotensin converting enzyme, regulators of volume homeostasis, were significantly decreased in HAPE compared to AC. CONCLUSION Our findings indicate that regulators of endothelial function, vascular tone and cell stress are altered in altitude illness and may mechanistically contribute to the pathobiology of HAPE.
Collapse
Affiliation(s)
- Kevin R Barker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Andrea L Conroy
- Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Michael Hawkes
- Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada, Division of Infectious Diseases, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada and
| | - Holly Murphy
- CIWEC Hospital and Travel Medicine Center, Kathmandu, Nepal
| | - Prativa Pandey
- CIWEC Hospital and Travel Medicine Center, Kathmandu, Nepal
| | - Kevin C Kain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada, The Tropical Disease Unit, Department of Medicine, University of Toronto, Toronto, ON, Canada,
| |
Collapse
|
40
|
Lagassé HAD, Anidi IU, Craig JM, Limjunyawong N, Poupore AK, Mitzner W, Scott AL. Recruited monocytes modulate malaria-induced lung injury through CD36-mediated clearance of sequestered infected erythrocytes. J Leukoc Biol 2015; 99:659-71. [PMID: 26516185 DOI: 10.1189/jlb.4hi0315-130rrr] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 10/11/2015] [Indexed: 12/28/2022] Open
Abstract
Pulmonary complications occur in a significant percentage of adults and children during the course of severe malaria. The cellular and molecular innate immune mechanisms that limit the extent of pulmonary inflammation and preserve lung function during severe Plasmodium infections remain unclear. In particular, the contributions to pulmonary complications by parasitized erythrocyte sequestration and subsequent clearance from the lung microvasculature by immune cells have not been clearly defined. We used the Plasmodium berghei ANKA-C57BL/6 mouse model of severe malaria to investigate the mechanisms governing the nature and extent of malaria-associated lung injury. We have demonstrated that sequestration of infected erythrocytes on postcapillary endothelial surfaces results in acute lung injury and the rapid recruitment of CCR2(+)CD11b(+)Ly6C(hi) monocytes from the circulation. These recruited cells remain in the lungs as monocyte-derived macrophages and are instrumental in the phagocytic clearance of adherent Plasmodium berghei-infected erythrocytes. In contrast, alveolar macrophages do not play a significant role in the clearance of malaria-infected cells. Furthermore, the results obtained from Ccr2(-/-), Cd36(-/-), and CD36 bone marrow chimeric mice showed that sequestration in the absence of CD36-mediated phagocytic clearance by monocytes leads to exaggerated lung pathologic features. In summary, our data indicate that the intensity of malaria-induced lung pathologic features is proportional to the steady-state levels of Plasmodium-infected erythrocytes adhering to the pulmonary vasculature. Moreover, the present work has defined a major role of recruited monocytes in clearing infected erythrocytes from the pulmonary interstitium, thus minimizing lung damage.
Collapse
Affiliation(s)
- H A Daniel Lagassé
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA, and
| | - Ifeanyi U Anidi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA, and
| | - John M Craig
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA, and
| | - Nathachit Limjunyawong
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Amy K Poupore
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA, and
| | - Wayne Mitzner
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alan L Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA, and
| |
Collapse
|
41
|
Ampawong S, Chaisri U, Viriyavejakul P, Prapansilp P, Grau GE, Turner GDH, Pongponratn E. A potential role for interleukin-33 and γ-epithelium sodium channel in the pathogenesis of human malaria associated lung injury. Malar J 2015; 14:389. [PMID: 26437894 PMCID: PMC4595310 DOI: 10.1186/s12936-015-0922-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/27/2015] [Indexed: 01/05/2023] Open
Abstract
Background The pathogenesis of pulmonary oedema (PE) in patients with severe malaria is still unclear. It has been hypothesized that lung injury depends, in addition to microvascular obstruction, on an increased pulmonary capillary pressure and altered alveolar-capillary membrane permeability, causing pulmonary fluid accumulation. Methods This study compared the histopathological features of lung injury in Southeast Asian patients (n = 43) who died from severe Plasmodium falciparum malaria, and correlated these with clinical history in groups with or without PE. To investigate the expression of mediators that may influence fluid accumulation in PE, immunohistochemistry and image analysis were performed on controls and sub-sets of patient with or without PE. Results The expression of leukocyte sub-set antigens, bronchial interleukin (IL)-33, γ-epithelium sodium channel (ENaC), aquaporin (AQP)-1 and -5, and control cytokeratin staining was quantified in the lung tissue of severe malaria patients. Bronchial IL-33 expression was significantly increased in severe malaria patients with PE. Malaria patients with shock showed significantly increased bronchial IL-33 compare to other clinical manifestations. Bronchial IL-33 levels were positively correlated with CD68+ monocyte and elastase + neutrophil, septal congestion and hyaline membrane formation. Moreover, the expression of both vascular smooth muscle cell (VSMC) and bronchial γ-ENaC significantly decreased in severe malaria patients with PE. Both VSMC and bronchial γ-ENaC were negatively correlated with the degree of parasitized erythrocyte sequestration, alveolar thickness, alveolar expansion score, septal congestion score, and malarial pigment score. In contrast AQP-1 and -5 and pan cytokeratin levels were similar between groups. Conclusions The results suggest that IL-33 may play a role in lung injury during severe malaria and lead to PE. Both VSMC and bronchial γ-ENaC downregulation may explain pulmonary fluid disturbances and participate in PE pathogenesis in severe malaria patients.
Collapse
Affiliation(s)
- Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Parnpen Viriyavejakul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Panote Prapansilp
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Georges E Grau
- Vascular Immunology Unit, Department of Pathology, Sydney Medical School, The University of Sydney, Parramatta Road, Camperdown, NSW, Australia.
| | - Gareth D H Turner
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Nuffield Department of Clinical Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK.
| | - Emsri Pongponratn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
42
|
Punsawad C, Viriyavejakul P, Setthapramote C, Palipoch S. Enhanced expression of Fas and FasL modulates apoptosis in the lungs of severe P. falciparum malaria patients with pulmonary edema. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:10002-10013. [PMID: 26617708 PMCID: PMC4637523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 08/25/2015] [Indexed: 06/05/2023]
Abstract
Apoptosis mediated by Fas/FasL has been implicated in pulmonary disorders. However, little is known about the relationship between Fas and FasL in the process of lung injury during malaria infection. Paraffin-embedded lung tissues from malaria patients were divided into two groups: those with pulmonary edema (PE) and those without pulmonary edema (non-PE). Normal lung tissues were used as the control group. Cellular expression of Fas, FasL, and the markers of apoptotic caspases, including cleaved caspase-3 and cleaved caspase-8 in the lung tissues were investigated by the immunohistochemistry (IHC) method. Semi-quantitative analysis of IHC staining revealed that cellular expression of Fas, FasL, cleaved caspase-8, and cleaved caspase-3 were significantly increased in the lungs of patients with PE compared with the lungs of patients with non-PE and control groups (all P < 0.05). In addition, significant positive correlations were obtained between Fas and apoptosis (rs = 0.937, P < 0.001) and FasL and apoptosis (rs = 0.808, P < 0.001). Significant positive correlations were found between Fas and FasL expression (rs = 0.827, P < 0.001) and between cleaved caspase-8 and cleaved caspase-3 expression (rs = 0.823, P < 0.001), which suggests that Fas-dependent initiator and effector caspases, including cleaved caspase-8 and caspase-3, are necessary for inducing apoptosis in the lungs of patients with severe P. falciparum malaria. The Fas/FasL system and downstream activation of caspases are important mediators of apoptosis and may be involved in the pathogenesis of pulmonary edema in severe P. falciparum malaria patients. The proper regulation of the Fas/FasL pathway can be a potential treatment for pulmonary complications in falciparum malaria patients.
Collapse
Affiliation(s)
- Chuchard Punsawad
- School of Medicine, Walailak University222 Thaiburi, Thasala District, Nakhon Si Thammarat 80161, Thailand
| | - Parnpen Viriyavejakul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University420/6 Rajvithi Road, Bangkok 10400, Thailand
| | - Chayanee Setthapramote
- Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University681 Samsen Road, Bangkok 10300, Thailand
| | - Sarawoot Palipoch
- School of Medicine, Walailak University222 Thaiburi, Thasala District, Nakhon Si Thammarat 80161, Thailand
| |
Collapse
|
43
|
Interleukin-18 Antagonism Improved Histopathological Conditions of Malaria Infection in Mice. IRANIAN JOURNAL OF PARASITOLOGY 2015; 10:389-401. [PMID: 26622294 PMCID: PMC4662739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND Interleukin 18 (IL-18) exerts pleiotropic roles in many inflammatory-related diseases including parasitic infection. Previous studies have demonstrated the promising therapeutic potential of modulating IL-18 bioactivity in various pathological conditions. However, its involvement during malaria infection has yet to be established. In this study, we demonstrated the effect of modulating IL-18 on the histopathological conditions of malaria infected mice. METHODS Plasmodium berghei ANKA infection in male ICR mice was used as a model for malaria infection. Modulation of IL-18 release was carried out by treatment of malarial mice with recombinant mouse IL-18 (rmIL-18) and recombinant mouse IL-18 Fc chimera (rmIL-18Fc) intravenously. Histopathological study and analysis were performed on major organs including brain, liver, spleen, lungs and kidney. RESULTS Treatment with rmIL-18Fc resulted in significant improvements on the histopathological conditions of the organs in malaria-infected mice. CONCLUSION IL-18 is an important mediator of malaria pathogenesis and targeting IL-18 could prove beneficial in malaria-infected host.
Collapse
|
44
|
Werner JL, Steele C. Innate receptors and cellular defense against pulmonary infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3842-50. [PMID: 25281754 PMCID: PMC4185409 DOI: 10.4049/jimmunol.1400978] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the United States, lung infections consistently rank in the top 10 leading causes of death, accounting for >50,000 deaths annually. Moreover, >140,000 deaths occur annually as a result of chronic lung diseases, some of which may be complicated by an infectious process. The lung is constantly exposed to the environment and is susceptible to infectious complications caused by bacterial, viral, fungal, and parasitic pathogens. Indeed, we are continually faced with the threat of morbidity and mortality associated with annual influenza virus infections, new respiratory viruses (e.g., SARS-CoV), and lung infections caused by antibiotic-resistant "ESKAPE pathogens" (three of which target the lung). This review highlights innate immune receptors and cell types that function to protect against infectious challenges to the respiratory system yet also may be associated with exacerbations in chronic lung diseases.
Collapse
Affiliation(s)
- Jessica L Werner
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109; and
| | - Chad Steele
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
45
|
Predictive criteria to study the pathogenesis of malaria-associated ALI/ARDS in mice. Mediators Inflamm 2014; 2014:872464. [PMID: 25276057 PMCID: PMC4167651 DOI: 10.1155/2014/872464] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/16/2014] [Indexed: 12/14/2022] Open
Abstract
Malaria-associated acute lung injury/acute respiratory distress syndrome (ALI/ARDS) often results in morbidity and mortality. Murine models to study malaria-associated ALI/ARDS have been described; we still lack a method of distinguishing which mice will develop ALI/ARDS before death. This work aimed to characterize malaria-associated ALI/ARDS in a murine model and to demonstrate the first method to predict whether mice are suffering from ALI/ARDS before death. DBA/2 mice infected with Plasmodium berghei ANKA developing ALI/ARDS or hyperparasitemia (HP) were compared using histopathology, PaO2 measurement, pulmonary X-ray, breathing capacity, lung permeability, and serum vascular endothelial growth factor (VEGF) levels according to either the day of death or the suggested predictive criteria. We proposed a model to predict malaria-associated ALI/ARDS using breathing patterns (enhanced pause and frequency respiration) and parasitemia as predictive criteria from mice whose cause of death was known to retrospectively diagnose the sacrificed mice as likely to die of ALI/ARDS as early as 7 days after infection. Using this method, we showed increased VEGF levels and increased lung permeability in mice predicted to die of ALI/ARDS. This proposed method for accurately identifying mice suffering from ALI/ARDS before death will enable the use of this model to study the pathogenesis of this disease.
Collapse
|
46
|
Cabrera A, Neculai D, Kain KC. CD36 and malaria: friends or foes? A decade of data provides some answers. Trends Parasitol 2014; 30:436-44. [PMID: 25113859 DOI: 10.1016/j.pt.2014.07.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/16/2014] [Accepted: 07/17/2014] [Indexed: 02/07/2023]
Abstract
The past 10 years have generated new insights into the complex interaction between CD36 (cluster of differentiation 36) and malaria. These range from the crystallization of the CD36 homolog, LIMPII (lysosomal integral membrane protein II), permitting modeling of CD36 and its binding to diverse ligands, to cell biology-based studies of CD36 and large population genetic studies assessing the association of CD36 polymorphisms and malarial disease severity. Collectively these lines of evidence indicate that a receptor other than CD36 is associated with severity. CD36 plays an important role in innate immunity and in the phagocytic uptake of multiple pathogens including malaria. CD36 polymorphisms lack association with severity, and isolates that cause severe disease primarily bind to endothelial protein C receptor (EPCR) rather than to CD36.
Collapse
Affiliation(s)
- Ana Cabrera
- Sandra Ann Rotman (SAR) Laboratories, SAR Centre, Toronto General Hospital, University Health Network, Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dante Neculai
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kevin C Kain
- Sandra Ann Rotman (SAR) Laboratories, SAR Centre, Toronto General Hospital, University Health Network, Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
47
|
Aitken EH, Negri EM, Barboza R, Lima MRI, Álvarez JM, Marinho CRF, Caldini EG, Epiphanio S. Ultrastructure of the lung in a murine model of malaria-associated acute lung injury/acute respiratory distress syndrome. Malar J 2014; 13:230. [PMID: 24927627 PMCID: PMC4062769 DOI: 10.1186/1475-2875-13-230] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/07/2014] [Indexed: 02/06/2023] Open
Abstract
Background The mechanisms through which infection with Plasmodium spp. result in lung disease are largely unknown. Recently a number of mouse models have been developed to research malaria-associated lung injury but no detailed ultrastructure studies of the disease in its terminal stages in a murine model have yet been published. The goal was to perform an ultrastructural analysis of the lungs of mice that died with malaria-associated acute lung injury/acute respiratory distress syndrome to better determine the relevancy of the murine models and investigate the mechanism of disease. Methods DBA/2 mice were infected with Plasmodium berghei strain ANKA. Mice had their lungs removed immediately after death, processed using standard methods and viewed by transmission electron microscopy (TEM). Results Infected red blood cell:endothelium contact, swollen endothelium with distended cytoplasmic extensions and thickening of endothelium basement membrane were observed. Septa were thick and filled with congested capillaries and leukocytes and the alveolar spaces contained blood cells, oedema and cell debris. Conclusion Results show that the lung ultrastructure of P. berghei ANKA-infected mice has similar features to what has been described in post-mortem TEM studies of lungs from individuals infected with Plasmodium falciparum. These data support the use of murine models to study malaria-associated acute lung injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sabrina Epiphanio
- Department of Immunology, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
48
|
Deroost K, Opdenakker G, Van den Steen PE. MalarImDB: an open-access literature-based malaria immunology database. Trends Parasitol 2014; 30:309-16. [DOI: 10.1016/j.pt.2014.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/28/2014] [Accepted: 04/04/2014] [Indexed: 12/23/2022]
|
49
|
Zuzarte-Luis V, Mota MM, Vigário AM. Malaria infections: what and how can mice teach us. J Immunol Methods 2014; 410:113-22. [PMID: 24837740 DOI: 10.1016/j.jim.2014.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/24/2014] [Accepted: 05/01/2014] [Indexed: 01/07/2023]
Abstract
Malaria imposes a horrific public health burden - hundreds of millions of infections and millions of deaths - on large parts of the world. While this unacceptable health burden and its economic and social impact have made it a focal point of the international development agenda, it became consensual that malaria control or elimination will be difficult to attain prior to gain a better understanding of the complex interactions occurring between its main players: Plasmodium, the causative agent of disease, and its hosts. Practical and ethical limitations exist regarding the ability to carry out research with human subjects or with human samples. In this review, we highlight how rodent models of infection have contributed significantly during the past decades to a better understanding of the basic biology of the parasite, host response and pathogenesis.
Collapse
Affiliation(s)
- Vanessa Zuzarte-Luis
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Ana M Vigário
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; Unidade de Ciências Médicas, Centro de Competência de Ciências da Vida, Universidade da Madeira, Funchal, Portugal.
| |
Collapse
|
50
|
Brugat T, Cunningham D, Sodenkamp J, Coomes S, Wilson M, Spence PJ, Jarra W, Thompson J, Scudamore C, Langhorne J. Sequestration and histopathology in Plasmodium chabaudi malaria are influenced by the immune response in an organ-specific manner. Cell Microbiol 2014; 16:687-700. [PMID: 24003897 PMCID: PMC4234010 DOI: 10.1111/cmi.12212] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/26/2013] [Accepted: 08/26/2013] [Indexed: 11/28/2022]
Abstract
Infection with the malaria parasite, Plasmodium, is associated with a strong inflammatory response and parasite cytoadhesion (sequestration) in several organs. Here, we have carried out a systematic study of sequestration and histopathology during infection of C57Bl/6 mice with Plasmodium chabaudi AS and determined the influence of the immune response. This parasite sequesters predominantly in liver and lung, but not in the brain, kidney or gut. Histopathological changes occur in multiple organs during the acute infection, but are not restricted to the organs where sequestration takes place. Adaptive immunity, and signalling through the IFNγ receptor increased sequestration and histopathology in the liver, but not in the lung, suggesting that there are differences in the adhesion molecules and/or parasite ligands utilized and mechanisms of pathogenesis in these two organs. Exacerbation of pro-inflammatory responses during infection by deletion of the il10 gene resultsin the aggravation of damage to lung and kidney irrespective of the degree of sequestration. The immune response therefore affected both sequestration and histopathology in an organ-specific manner. P. chabaudi AS provides a good model to investigate the influence of the host response on the sequestration and specific organ pathology, which is applicable to human malaria.
Collapse
Affiliation(s)
- Thibaut Brugat
- Division of Parasitology, MRC National Institute for Medical Research, London, NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|