1
|
Frömmel N, El Baraka I, Walz B, Hellmich B. [Atraumatic splenic rupture in Legionella pneumonia]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2025; 66:546-549. [PMID: 39715833 DOI: 10.1007/s00108-024-01829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 12/25/2024]
Abstract
A 48-year-old male patient was admitted to hospital with Legionella pneumonia. He developed a splenic rupture 1 day after admission, which was surgically treated. Retrospectively, the splenomegaly could already be seen by sonography on admission. An alternative etiology of the splenic rupture was excluded. The case highlights the importance of early detection of splenomegaly in patients with legionellosis. Through an early interdisciplinary presentation this life-threatening complication can be recognized in a timely manner and safely treated.
Collapse
Affiliation(s)
- Niklas Frömmel
- Klinik für Radiologie, medius Klinik Kirchheim, Eugenstraße 3, 73230, Kirchheim unter Teck, Deutschland.
| | - Ibtissame El Baraka
- Klinik für Radiologie, medius Klinik Kirchheim, Eugenstraße 3, 73230, Kirchheim unter Teck, Deutschland
| | - Bastian Walz
- Klinik für Innere Medizin, Rheumatologie, Pneumologie, Nephrologie und Diabetologie, medius Klinik Kirchheim, Kirchheim unter Teck, Deutschland
| | - Bernhard Hellmich
- Klinik für Innere Medizin, Rheumatologie, Pneumologie, Nephrologie und Diabetologie, medius Klinik Kirchheim, Kirchheim unter Teck, Deutschland
| |
Collapse
|
2
|
Zorrinho-Almeida M, de-Carvalho J, Bernabeu M, Silva Pereira S. Leveraging microphysiological systems to expedite understanding of host-parasite interactions. PLoS Pathog 2025; 21:e1013088. [PMID: 40273176 PMCID: PMC12021206 DOI: 10.1371/journal.ppat.1013088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Abstract
Microphysiological systems (MPS) replicate the dynamic interactions between cells, tissues, and fluids. They have emerged as transformative tools for biology and have been increasingly applied to host-parasite interactions. Offering a better representation of cellular behavior compared with traditional in vitro models, MPS can facilitate the study of parasite tropism, immune evasion, and life cycle transitions across diverse parasitic diseases. Applications span multiple host tissues and pathogens, leveraging advanced bioengineering and microfabrication techniques to address long-standing knowledge gaps. Here, we review recent advances in MPS applied to parasitic diseases and identify persisting challenges and opportunities for investment. By refining these systems and integrating host multicellular models and parasites, MPS hold vast potential to revolutionize parasitology, enhancing our ability to combat parasitic diseases through deeper mechanistic understanding and targeted interventions.
Collapse
Affiliation(s)
- Maria Zorrinho-Almeida
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Oeiras, Portugal
| | | | | | - Sara Silva Pereira
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Oeiras, Portugal
| |
Collapse
|
3
|
Du MRM, Wang C, Law CW, Amann-Zalcenstein D, Anttila CJA, Ling L, Hickey PF, Sargeant CJ, Chen Y, Ioannidis LJ, Rajasekhar P, Yip RKH, Rogers KL, Hansen DS, Bowden R, Ritchie ME. Benchmarking spatial transcriptomics technologies with the multi-sample SpatialBenchVisium dataset. Genome Biol 2025; 26:77. [PMID: 40156041 PMCID: PMC11954323 DOI: 10.1186/s13059-025-03543-4] [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: 03/13/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Spatial transcriptomics allows gene expression to be measured within complex tissue contexts. Among the array of spatial capture technologies available is 10x Genomics' Visium platform, a popular method which enables transcriptome-wide profiling of tissue sections. Visium offers a range of sample handling and library construction methods which introduces a need for benchmarking to compare data quality and assess how well the technology can recover expected tissue features and biological signatures. RESULTS Here we present SpatialBenchVisium, a unique reference dataset generated from spleen tissue of mice responding to malaria infection spanning several tissue preparation protocols (both fresh frozen and FFPE, with either manual or CytAssist tissue placement). We note better quality control metrics in reference samples prepared using probe-based capture methods, particularly those processed with CytAssist, validating the improvement in data quality produced with the platform. Our analysis of replicate samples extends to explore spatially variable gene detection, the outcomes of clustering and cell deconvolution using matched single-cell RNA-sequencing data and publicly available reference data to identify cell types and tissue regions expected in the spleen. Multi-sample differential expression analysis recovered known gene signatures related to biological sex or gene knockout.
Collapse
Affiliation(s)
- Mei R M Du
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Changqing Wang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Charity W Law
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniela Amann-Zalcenstein
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Casey J A Anttila
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Ling Ling
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Peter F Hickey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Callum J Sargeant
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Yunshun Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Pradeep Rajasekhar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Raymond K H Yip
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Diana S Hansen
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Rory Bowden
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
4
|
Kalkal M, Das J. Differential B cell mediated immune response during Plasmodium yoelii infection in mice. Acta Trop 2025; 263:107533. [PMID: 39889895 DOI: 10.1016/j.actatropica.2025.107533] [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: 11/04/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
B lymphocytes are essential components of the humoral immune response and categorized into various subsets according to specific surface markers, functions, and developmental stages. Each subset of B cells plays a distinct role in the immune response, contributing to the overall effectiveness of the immune system. In this study, we investigated the modulation of different splenic subsets of B cells during Plasmodium yoelii infection. Balb/c mice infected with each Plasmodium yoelii XL and Plasmodium yoelii XNL parasite were used for phenotypic characterization of splenic B cell subsets through flow-cytometry. Our findings indicate that both lethal and non-lethal infections of Plasmodium yoelii result in significant alterations within the B cell compartment of the spleen in Balb/c mice during malaria infection. Notably, a differential expansion of immature B cell subsets T1 and T2 was noticed. A continuous reduction in frequency of both subsets (T1 and T2) during infection with lethal parasite while an increase in these subsets during the recovery from infection with non-lethal parasite was observed. Further, the frequencies of mature B cell subsets, follicular B cells and marginal zone B cells, were reduced during lethal infection which may be leading to susceptibility. Whereas non-lethal parasite infection resulted in increased frequency of follicular B cells in spleen which indicates towards establishment of germinal centre for generation of long-term immunity/resistance to infection. This differential expansion of splenic B cell subsets reflects the distinct characteristics of lethal and non-lethal parasite. Overall, these findings illustrate the potential role of B cells in resistance/susceptibility during malaria infection and further enhance our understanding of the B cell mediated immunological aspects of Plasmodium infection.
Collapse
Affiliation(s)
- Meenu Kalkal
- Immunology Division, ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India
| | - Jyoti Das
- Immunology Division, ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India.
| |
Collapse
|
5
|
Rodríguez Muñoz D. Immunophenotyping of Hematopoietic Cells in the Spleen in Hypothyroid Mice. Methods Mol Biol 2025; 2876:105-115. [PMID: 39579311 DOI: 10.1007/978-1-0716-4252-8_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] [Indexed: 11/25/2024]
Abstract
Hypothyroidism, which is characterized by insufficient production of thyroid hormones, and malaria, a mosquito-borne infectious disease caused by Plasmodium parasites, are significant global health challenges. Studying how these two conditions interact could provide valuable insights into their complex relationship and potential treatment options.To induce hypothyroidism in the research, scientists used drugs to block the production of thyroid hormones. Then, they infected mice with Plasmodium berghei ANKA to mimic cerebral malaria infection. The spleen is essential in the body's immune response to malaria. It is involved in both innate and adaptive immunity, iron recycling, and the removal of old red blood cells or damaged cells infected with Plasmodium. Monitoring disease progression in male mice is crucial for early detection, and techniques like flow cytometry can help identify specific immune system populations within the spleen that are relevant to the research.
Collapse
|
6
|
Murshed M, AL-Tamimi J, Mares MM, Hailan WAQ, Ibrahim KE, Al-Quraishy S. Pharmacological Effects of Biosynthesis Silver Nanoparticles Utilizing Calotropis procera Leaf Extracts on Plasmodium berghei-Infected Liver in Experiment Mice. Int J Nanomedicine 2024; 19:13717-13733. [PMID: 39726977 PMCID: PMC11669542 DOI: 10.2147/ijn.s490119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Malaria caused by Plasmodium spp. is the most hazardous disease in the world. It is regarded as a life-threatening hematological disorder caused by parasites transferred to humans by the bite of Anopheles mosquitoes. Purpose Calotropis procera leaf extract combined with biosynthesized silver nanoparticles (CPLEAgNPs) to evaluate its antiplasmodium and hepatoprotective effects against P. berghei-induced infection in experimental mice. Methods The animal groups were divided into four groups: the first non-infected group was orally administered distilled water daily 7 days. The second group received an oral dose of 50 mg/kg of CPLE AgNPs. The third group received intraperitoneal injections of 105 P. berghei. The fourth group received of 105 P. berghei with 50 mg/kg CPLE AgNPs. All mice were anesthetized with CO2 and dissected for sample collection. Results This study of C. procera leaves showed that they contain chemically active substances, as shown by the amounts of phenols, flavonoids, and tannins. The antioxidant activity of the samples was assessed using 1.1-diphenyl-2-picrylhydrazyl (DPPH) and 2.2'-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid (ABTS) assays. Treatment of infected mice with CPLE AgNPs for 7 days resulted in a significant decrease in parasitemia and a reduction in histopathological alterations in the liver. Furthermore, CPLE AgNPs mitigated oxidative damage caused by P. berghei infection in the liver. In addition, after receiving the medication, the liver levels of alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase decreased. In addition, CPLE AgNPs regulated the expression of liver cytokines, including IL-1β, and I-10. Discussion Based on these findings, the study proved that CPLE AgNPs have hepatoprotective and antiplasmodial properties.
Collapse
Affiliation(s)
- Mutee Murshed
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Jameel AL-Tamimi
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Mohammed M Mares
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Waleed A Q Hailan
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Khalid Elfaki Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| |
Collapse
|
7
|
Sissoko A, Othmene YB, Buffet P. Splenic filtration of red blood cells in physiology, malaria and sickle cell disease. Curr Opin Hematol 2024; 31:307-314. [PMID: 39259191 DOI: 10.1097/moh.0000000000000839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
PURPOSE OF REVIEW The human spleen clears the blood from circulating microorganisms and red blood cells (RBCs) displaying alterations. This review analyzes how generic mechanisms by which the spleen senses RBC, such pitting, trapping and erythrophagocytosis, impact the pathogenesis of two major spleen-related diseases, malaria and sickle cell disease (SCD). RECENT FINDINGS Scintigraphy, functional histology, comparison of circulating and splenic RBC, ex-vivo perfusion of human spleens and in-silico modeling enable relevant exploration of how the spleen retains and processes RBC in health and disease. Iterative cross-validations between medical observations, in-vitro experiments and in-silico modeling point to mechanical sensing of RBC as a central event in both conditions. Spleen congestion is a common pathogenic process explaining anemia and splenomegaly, the latter carrying a risk of severe complications such as acute splenic sequestration crisis and hypersplenism in SCD. Sickling of hemoglobin S-containing RBC may contribute to these complications without necessarily being the trigger. SUMMARY Ongoing progress in the exploration and understanding of spleen-related complications in malaria and SCD open the way to optimized prognosis evaluation and therapeutic applications.
Collapse
Affiliation(s)
| | | | - Pierre Buffet
- Université Paris Cité, Inserm, BIGR
- Assistance publique des hôpitaux de Paris
- Institut Pasteur, Université de Paris Cité, Paris, France
| |
Collapse
|
8
|
Mukherjee S, Ghosh P, Ghosh S, Sengupta A, Sarkar S, Chatterjee R, Saha A, Bawali S, Choudhury A, Daptary AH, Gangopadhyay A, Keswani T, Bhattacharyya A. Administration of rIL-33 Restores Altered mDC/pDC Ratio, MDSC Frequency, and Th-17/Treg Ratio during Experimental Cerebral Malaria. Pathogens 2024; 13:877. [PMID: 39452748 PMCID: PMC11509898 DOI: 10.3390/pathogens13100877] [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: 06/24/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
The onset of malaria causes the induction of various inflammatory markers in the host's body, which in turn affect the body's homeostasis and create several cerebral complications. Polarization of myeloid-derived suppressor cells (MDSCs) from the classically activated M1 to alternatively activated M2 phenotype increases the secretion of pro-inflammatory molecules. Treatment with recombinant IL-33 (rIL-33) not only alters this MDSC's polarization but also targets the glycolysis pathway of the metabolism in MDSCs, rendering them less immunosuppressive. Along with that, the Helper T-cells subset 17 (Th17)/T regulatory cells (Tregs) ratio is skewed towards Th17, which increases inflammation by producing more IL-17. However, treating with rIL-33 also helps to restore this ratio, which brings back homeostasis. During malaria infection, there is an upregulation of IL-12 production from dendritic cells along with a distorted myeloid dendritic cells (mDC)/plasmacytoid dendritic cells (pDC) ratio towards mDCs promoting inflammation. Administering rIL-33 will also subvert this IL-12 production and increase the population of pDC in the host's immune system during malaria infection, thus restoring mDC/pDC to homeostasis. Therefore, treatment with rIL-33 to reduce the pro-inflammatory signatures and maintenance of immune homeostasis along with the increase in survivability could be a potential therapeutic approach for cerebral malaria.
Collapse
Affiliation(s)
- Saikat Mukherjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Pronabesh Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Soubhik Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Anirban Sengupta
- Centre for Infectious Medicine, Department of Medicine Huddinge, Karolinksa Institutet, 14152 Stockholm, Sweden;
| | - Samrat Sarkar
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Rimbik Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Atreyee Saha
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Sriparna Bawali
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Abhishek Choudhury
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Altamas Hossain Daptary
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Anwesha Gangopadhyay
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| |
Collapse
|
9
|
Hoo R, Ruiz-Morales ER, Kelava I, Rawat M, Mazzeo CI, Tuck E, Sancho-Serra C, Chelaghma S, Predeus AV, Murray S, Fernandez-Antoran D, Waller RF, Álvarez-Errico D, Lee MCS, Vento-Tormo R. Acute response to pathogens in the early human placenta at single-cell resolution. Cell Syst 2024; 15:425-444.e9. [PMID: 38703772 DOI: 10.1016/j.cels.2024.04.002] [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: 05/19/2023] [Revised: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Mukul Rawat
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | | | | | | | - Sara Chelaghma
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - David Fernandez-Antoran
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Marcus C S Lee
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Feng X, Yu JL, Sun YF, Du CY, Shen Y, Zhang L, Kong WZ, Han S, Cheng Y. Plasmodium yoelii surface-related antigen (PySRA) modulates the host pro-inflammatory responses via binding to CD68 on macrophage membrane. Infect Immun 2024; 92:e0011324. [PMID: 38624215 PMCID: PMC11075460 DOI: 10.1128/iai.00113-24] [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: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/17/2024] Open
Abstract
Malaria, one of the major infectious diseases in the world, is caused by the Plasmodium parasite. Plasmodium antigens could modulate the inflammatory response by binding to macrophage membrane receptors. As an export protein on the infected erythrocyte membrane, Plasmodium surface-related antigen (SRA) participates in the erythrocyte invasion and regulates the immune response of the host. This study found that the F2 segment of P. yoelii SRA activated downstream MAPK and NF-κB signaling pathways by binding to CD68 on the surface of the macrophage membrane and regulating the inflammatory response. The anti-PySRA-F2 antibody can protect mice against P. yoelii, and the pro-inflammatory responses such as IL-1β, TNF-α, and IL-6 after infection with P. yoelii are attenuated. These findings will be helpful for understanding the involvement of the pathogenic mechanism of malaria with the exported protein SRA.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Antigens, CD/metabolism
- Antigens, CD/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Protozoan/immunology
- Antigens, Protozoan/metabolism
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Cell Membrane/metabolism
- Cell Membrane/immunology
- Inflammation/immunology
- Inflammation/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/parasitology
- Malaria/immunology
- Malaria/parasitology
- NF-kappa B/metabolism
- NF-kappa B/immunology
- Plasmodium yoelii/immunology
- Protein Binding
- Signal Transduction
Collapse
Affiliation(s)
- Xin Feng
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jia-Li Yu
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yi-Fan Sun
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Chen-Yan Du
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yao Shen
- Department of Food Quality and Safety, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Lu Zhang
- Department of General Practice, Rongxiang Street Community Health Service Center, Binhu District, Wuxi, China
| | - Wei-Zhong Kong
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Su Han
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yang Cheng
- Department of Public Health and Preventive Medicine, Laboratory of Pathogen Infection and Immunity, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Sharma I, Kataria P, Das J. Cerebral malaria pathogenesis: Dissecting the role of CD4 + and CD8 + T-cells as major effectors in disease pathology. Int Rev Immunol 2024; 43:309-325. [PMID: 38618863 DOI: 10.1080/08830185.2024.2336539] [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: 09/25/2023] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024]
Abstract
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum (P. falciparum) infection, with complex pathogenesis involving multiple factors, including the host's immunological response. T lymphocytes, specifically CD4+ T helper cells and CD8+ cytotoxic T cells, are crucial in controlling parasite growth and activating cells for parasite clearance via cytokine secretion. Contrary to this, reports also suggest the pathogenic nature of T lymphocytes as they are often involved in disease progression and severity. CD8+ cytotoxic T cells migrate to the host's brain vasculature, disrupting the blood-brain barrier and causing neurological manifestations. CD4+ T helper cells on the other hand play a variety of functions as they differentiate into different subtypes which may function as pro-inflammatory or anti-inflammatory. The excessive pro-inflammatory response in CM can lead to multi-organ failure, necessitating a check mechanism to maintain immune homeostasis. This is achieved by regulatory T cells and their characteristic cytokines, which counterbalance the pro-inflammatory immune response. Maintaining a critical balance between pro and anti-inflammatory responses is crucial for determining disease outcomes in CM. A slight change in this balance may contribute to a disease severity owing to an extreme inflammatory response or unrestricted parasite growth, a potential target for designing immunotherapeutic treatment approaches. The review briefly discusses the pathogenesis of CM and various mechanisms responsible for the disruption of the blood-brain barrier. It also highlights the role of different T cell subsets during infection and emphasizes the importance of balance between pro and anti-inflammatory T cells that ultimately decides the outcome of the disease.
Collapse
Affiliation(s)
- Indu Sharma
- Academy of Scientific and Innovative Research (AcSIR), Noida, India
- Division of Immunology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Poonam Kataria
- Academy of Scientific and Innovative Research (AcSIR), Noida, India
- Division of Immunology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Jyoti Das
- Academy of Scientific and Innovative Research (AcSIR), Noida, India
- Division of Immunology, National Institute of Malaria Research, Dwarka, New Delhi, India
| |
Collapse
|
12
|
Silva Pereira S, Brás D, Porqueddu T, Nascimento AM, De Niz M. Investigation of Trypanosoma-induced vascular damage sheds insights into Trypanosoma vivax sequestration. Cell Surf 2023; 10:100113. [PMID: 37954640 PMCID: PMC10632540 DOI: 10.1016/j.tcsw.2023.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/22/2023] [Accepted: 10/22/2023] [Indexed: 11/14/2023] Open
Abstract
Multiple blood-borne pathogens infecting mammals establish close interactions with the host vascular endothelium as part of their life cycles. In this work, we investigate differences in the interactions of three Trypanosoma species: T. brucei, T. congolense and T. vivax with the blood vasculature. Infection with these species results in vastly different pathologies, including different effects on vascular homeostasis, such as changes in vascular permeability and microhemorrhages. While all three species are extracellular parasites, T. congolense is strictly intravascular, while T. brucei is capable of surviving both extra- and intravascularly. Our knowledge regarding T. vivax tropism and its capacity of migration across the vascular endothelium is unknown. In this work, we show for the first time that T. vivax parasites sequester to the vascular endothelium of most organs, and that, like T. congolense, T. vivax Y486 is largely incapable of extravasation. Infection with this parasite species results in a unique effect on vascular endothelium receptors including general downregulation of ICAM1 and ESAM, and upregulation of VCAM1, CD36 and E-selectin. Our findings on the differences between the two sequestering species (T. congolense and T. vivax) and the non-sequestering, but extravasating, T. brucei raise important questions on the relevance of sequestration to the parasite's survival in the mammalian host, and the evolutionary relevance of both sequestration and extravasation.
Collapse
Affiliation(s)
- Sara Silva Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Daniela Brás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Teresa Porqueddu
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M. Nascimento
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Center for Advanced Microscopy and Nikon Imaging Center, Northwestern University, Chicago, USA
| |
Collapse
|
13
|
Adachi R, Tamura T. Plasmodium Infection-Cure Cycles Increase the Capacity of Phagocytosis in Conventional Dendritic Cells. Pathogens 2023; 12:1262. [PMID: 37887778 PMCID: PMC10609740 DOI: 10.3390/pathogens12101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Malaria stands as one of the most pervasive human infectious diseases globally and represents a prominent cause of mortality. Immunity against clinical malaria disease is achieved through multiple infection and treatment cycles, culminating in a substantial reduction in parasite burden. To investigate this phenomenon, we established a murine model involving repeated infection-cure cycles, whereby mice were infected with the lethal rodent malarial parasite Plasmodium berghei ANKA and subsequently treated with the anti-malarial drug pyrimethamine. Our earlier study revealed a significant decrease in the capacity of conventional dendritic cells (cDCs) to produce cytokines upon stimulation in infection-cured mice. In the present study, we aimed to further elucidate the modulation of cDC functionality during repeated infection-cure cycles by examining their phagocytic capacity. Administering fluorescent beads to mice resulted in no significant difference in the total number of bead-positive cells within the spleens of both uninfected and 3-cure (three cycles of infection-cure) mice. However, the proportion of the CD11c+F4/80- population within bead-positive cells was notably higher in 3-cure mice compared to uninfected mice. Subsequent in vitro analysis of bead phagocytosis by purified CD11c+cDCs revealed that the cDC2 subset from 3-cure mice exhibited significantly enhanced phagocytic capacity in comparison to their uninfected counterparts. These findings underscore the substantial impact of repeated infection-cure cycles on various facets of cDC function, potentially influencing the trajectory of immune responses against subsequent malaria infections.
Collapse
Affiliation(s)
| | - Takahiko Tamura
- School of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
14
|
Wideman SK, Frost JN, Richter FC, Naylor C, Lopes JM, Viveiros N, Teh MR, Preston AE, White N, Yusuf S, Draper SJ, Armitage AE, Duarte TL, Drakesmith H. Cellular iron governs the host response to malaria. PLoS Pathog 2023; 19:e1011679. [PMID: 37812650 PMCID: PMC10586691 DOI: 10.1371/journal.ppat.1011679] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/19/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
Malaria and iron deficiency are major global health problems with extensive epidemiological overlap. Iron deficiency-induced anaemia can protect the host from malaria by limiting parasite growth. On the other hand, iron deficiency can significantly disrupt immune cell function. However, the impact of host cell iron scarcity beyond anaemia remains elusive in malaria. To address this, we employed a transgenic mouse model carrying a mutation in the transferrin receptor (TfrcY20H/Y20H), which limits the ability of cells to internalise iron from plasma. At homeostasis TfrcY20H/Y20H mice appear healthy and are not anaemic. However, TfrcY20H/Y20H mice infected with Plasmodium chabaudi chabaudi AS showed significantly higher peak parasitaemia and body weight loss. We found that TfrcY20H/Y20H mice displayed a similar trajectory of malaria-induced anaemia as wild-type mice, and elevated circulating iron did not increase peak parasitaemia. Instead, P. chabaudi infected TfrcY20H/Y20H mice had an impaired innate and adaptive immune response, marked by decreased cell proliferation and cytokine production. Moreover, we demonstrated that these immune cell impairments were cell-intrinsic, as ex vivo iron supplementation fully recovered CD4+ T cell and B cell function. Despite the inhibited immune response and increased parasitaemia, TfrcY20H/Y20H mice displayed mitigated liver damage, characterised by decreased parasite sequestration in the liver and an attenuated hepatic immune response. Together, these results show that host cell iron scarcity inhibits the immune response but prevents excessive hepatic tissue damage during malaria infection. These divergent effects shed light on the role of iron in the complex balance between protection and pathology in malaria.
Collapse
Affiliation(s)
- Sarah K. Wideman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Joe N. Frost
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Felix C. Richter
- Kennedy Institute of Rheumatology, Roosevelt Drive, Oxford, United Kingdom
| | - Caitlin Naylor
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - José M. Lopes
- Faculty of Medicine (FMUP) and Institute of Molecular Pathology, Immunology (IPATIMUP), University of Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nicole Viveiros
- Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Megan R. Teh
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alexandra E. Preston
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Natasha White
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Shamsideen Yusuf
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, United Kingdom
| | - Andrew E. Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Tiago L. Duarte
- Faculty of Medicine (FMUP) and Institute of Molecular Pathology, Immunology (IPATIMUP), University of Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
15
|
Quaye IK, Aleksenko L, Paganotti GM, Peloewetse E, Haiyambo DH, Ntebela D, Oeuvray C, Greco B. Malaria Elimination in Africa: Rethinking Strategies for Plasmodium vivax and Lessons from Botswana. Trop Med Infect Dis 2023; 8:392. [PMID: 37624330 PMCID: PMC10458071 DOI: 10.3390/tropicalmed8080392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 08/26/2023] Open
Abstract
The global malaria community has picked up the theme of malaria elimination in more than 90% of the world's population in the next decade. Recent reports of Plasmodium vivax (P. vivax) in sub-Saharan Africa, including in Duffy-negative individuals, threaten the efforts aimed at achieving elimination. This is not only in view of strategies that are tailored only to P. falciparum elimination but also due to currently revealed biological characteristics of P. vivax concerning the relapse patterns of hypnozoites and conservation of large biomasses in cryptic sites in the bone marrow and spleen. A typical scenario was observed in Botswana between 2008 and 2018, which palpably projects how P. vivax could endanger malaria elimination efforts where the two parasites co-exist. The need for the global malaria community, national malaria programs (NMPs), funding agencies and relevant stakeholders to engage in a forum to discuss and recommend clear pathways for elimination of malaria, including P. vivax, in sub-Saharan Africa is warranted.
Collapse
Affiliation(s)
- Isaac K. Quaye
- Pan African Vivax and Ovale Network, Faculty of Engineering Computer and Allied Sciences, Regent University College of Science and Technology, #1 Regent Ave, McCarthy Hill, Mendskrom, Dansoman, Accra P.O. Box DS1636, Ghana
| | - Larysa Aleksenko
- Department of Health Sciences, School of Public Health, College of Health, Medicine and Life Sciences, Brunel University, Kingston Lane, Uxbridge, Middlesex, London UB8 3PH, UK;
| | - Giacomo M. Paganotti
- Botswana-University of Pennsylvania Partnership, Riverwalk, Gaborone P.O. Box 45498, Botswana;
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elias Peloewetse
- Department of Biological Sciences, Faculty of Sciences, University of Botswana, Gaborone Private Bag 00704, Botswana;
| | - Daniel H. Haiyambo
- Department of Human, Biological and Translational Medical Sciences, Faculty of Health Sciences and Veterinary Medicine, University of Namibia School of Medicine, Hage Geingob Campus, Windhoek Private Bag 13301, Namibia;
| | - Davies Ntebela
- National Malaria Program, Ministry of Health, Gaborone Private Bag 0038, Botswana;
| | - Claude Oeuvray
- Global Health Institute of Merck, Terre Bonne Building Z0, Route de Crassier 1, Eysin, 1266 Geneva, Switzerland; (C.O.); (B.G.)
| | - Beatrice Greco
- Global Health Institute of Merck, Terre Bonne Building Z0, Route de Crassier 1, Eysin, 1266 Geneva, Switzerland; (C.O.); (B.G.)
| | - the PAVON Consortium
- PAVON, Regent University College of Science and Technology, #1 Regent Avenue, McCarthy Hiil, Mendskrom, Dansoman, Accra P.O. Box DS1636, Ghana
| |
Collapse
|
16
|
Rupar MJ, Sasserath T, Smith E, Comiter B, Sriram N, Long CJ, McAleer CW, Hickman JJ. Development of a human malaria-on-a-chip disease model for drug efficacy and off-target toxicity evaluation. Sci Rep 2023; 13:10509. [PMID: 37380653 DOI: 10.1038/s41598-023-35694-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
A functional, multi-organ, serum-free system was developed for the culture of P. falciparum in an attempt to establish innovative platforms for therapeutic drug development. It contains 4 human organ constructs including hepatocytes, splenocytes, endothelial cells, as well as recirculating red blood cells which allow for infection with the parasite. Two strains of P. falciparum were used: the 3D7 strain, which is sensitive to chloroquine; and the W2 strain, which is resistant to chloroquine. The maintenance of functional cells was successfully demonstrated both in healthy and diseased conditions for 7 days in the recirculating microfluidic model. To demonstrate an effective platform for therapeutic development, systems infected with the 3D7 strain were treated with chloroquine which significantly decreased parasitemia, with recrudescence observed after 5 days. Conversely, when the W2 systems were dosed with chloroquine, parasitemia levels were moderately decreased when compared to the 3D7 model. The system also allows for the concurrent evaluation of off-target toxicity for the anti-malarial treatment in a dose dependent manner which indicates this model could be utilized for therapeutic index determination. The work described here establishes a new approach to the evaluation of anti-malarial therapeutics in a realistic human model with recirculating blood cells for 7 days.
Collapse
Affiliation(s)
- Michael J Rupar
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Trevor Sasserath
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Ethan Smith
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Brandon Comiter
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Narasimhan Sriram
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Christopher J Long
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | | | - James J Hickman
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA.
| |
Collapse
|
17
|
Li G, Qiang Y, Li H, Li X, Dao M, Karniadakis GE. In silico and in vitro study of the adhesion dynamics of erythrophagocytosis in sickle cell disease. Biophys J 2023; 122:2590-2604. [PMID: 37231647 PMCID: PMC10323029 DOI: 10.1016/j.bpj.2023.05.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/12/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
Erythrophagocytosis occurring in the spleen is a critical process for removing senescent and diseased red blood cells (RBCs) from the microcirculation. Although some progress has been made in understanding how the biological signaling pathways mediate the phagocytic processes, the role of the biophysical interaction between RBCs and macrophages, particularly under pathological conditions such as sickle cell disease, has not been adequately studied. Here, we combine computational simulations with microfluidic experiments to quantify RBC-macrophage adhesion dynamics under flow conditions comparable to those in the red pulp of the spleen. We also investigate the RBC-macrophage interaction under normoxic and hypoxic conditions. First, we calibrate key model parameters in the adhesion model using microfluidic experiments for normal and sickle RBCs under normoxia and hypoxia. We then study the adhesion dynamics between the RBC and the macrophage. Our simulation illustrates three typical adhesion states, each characterized by a distinct dynamic motion of the RBCs, namely firm adhesion, flipping adhesion, and no adhesion (either due to no contact with macrophages or detachment from the macrophages). We also track the number of bonds formed when RBCs and macrophages are in contact, as well as the contact area between the two interacting cells, providing mechanistic explanations for the three adhesion states observed in the simulations and microfluidic experiments. Furthermore, we quantify, for the first time to our knowledge, the adhesive forces between RBCs (normal and sickle) and macrophages under different oxygenated conditions. Our results show that the adhesive forces between normal cells and macrophages under normoxia are in the range of 33-58 pN and 53-92 pN for sickle cells under normoxia and 155-170 pN for sickle cells under hypoxia. Taken together, our microfluidic and simulation results improve our understanding of the biophysical interaction between RBCs and macrophages in sickle cell disease and provide a solid foundation for investigating the filtration function of the splenic macrophages under physiological and pathological conditions.
Collapse
Affiliation(s)
- Guansheng Li
- Division of Applied Mathematics, Brown University, Providence, Rhode Island
| | - Yuhao Qiang
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - He Li
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia.
| | - Xuejin Li
- Department of Engineering Mechanics and Center for X-Mechanics, Zhejiang University, Hangzhou, China
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | | |
Collapse
|
18
|
Dias LR, Caldas JP, Teixeira JPA, Gomes V, Babo A, Faustino AS, Baptista R, Valdoleiros SR, Santos L. Spontaneous splenic rupture due to falciparum malaria successfully treated with a conservative approach. Parasitol Int 2023; 94:102736. [PMID: 36750153 DOI: 10.1016/j.parint.2023.102736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/26/2023] [Indexed: 02/07/2023]
Abstract
Malaria is frequently associated with splenomegaly. However, spontaneous splenic rupture is a rare and life-threatening complication. It is mostly seen in acute infection in non-immune adults and Plasmodium vivax and Plasmodium falciparum have been associated with the majority of cases. We describe a case of splenic rupture in an adult with complicated malaria by Plasmodium falciparum in which a conservative approach was used.
Collapse
Affiliation(s)
- Lúcia Ribeiro Dias
- Infectious Diseases Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal.
| | - João Paulo Caldas
- Infectious Diseases Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Virgília Gomes
- General Surgery Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Alexandra Babo
- General Surgery Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Ana Sofia Faustino
- Infectious Diseases Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Ricardo Baptista
- Infectious Diseases Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Sofia R Valdoleiros
- Infectious Diseases Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | - Lurdes Santos
- Infectious Diseases Department, Centro Hospitalar Universitário de São João, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
19
|
Pathogenesis of Anemia in Canine Babesiosis: Possible Contribution of Pro-Inflammatory Cytokines and Chemokines-A Review. Pathogens 2023; 12:pathogens12020166. [PMID: 36839438 PMCID: PMC9962459 DOI: 10.3390/pathogens12020166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Canine babesiosis is a tick-borne protozoan disease caused by intraerythrocytic parasites of the genus Babesia. The infection may lead to anemia in infected dogs. However, anemia is not directly caused by the pathogen. The parasite's developmental stages only have a marginal role in contributing to a decreased red blood cell (RBC) count. The main cause of anemia in affected dogs is the immune response to the infection. This response includes antibody production, erythrophagocytosis, oxidative damage of RBCs, complement activation, and antibody-dependent cellular cytotoxicity. Moreover, both infected and uninfected erythrocytes are retained in the spleen and sequestered in micro-vessels. All these actions are driven by pro-inflammatory cytokines and chemokines, especially IFN-γ, TNF-α, IL-6, and IL-8. Additionally, imbalance between the actions of pro- and anti-inflammatory cytokines plays a role in patho-mechanisms leading to anemia in canine babesiosis. This article is a review of the studies on the pathogenesis of anemia in canine babesiosis and related diseases, such as bovine or murine babesiosis and human or murine malaria, and the role of pro-inflammatory cytokines and chemokines in the mechanisms leading to anemia in infected dogs.
Collapse
|
20
|
DKHIL MA, ALJAWDAH HMA, ABDEL-GABER R, THAGFAN FA, DELIC D, AL-QURAISHY S. The effect of Eucalyptus camaldulensis leaf extracts from different environmental harvesting locations on Plasmodium chabaudi-induced malaria outcome. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.006723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
21
|
Porto-Pedrosa MLM, de Miranda CDM, Lopes ME, Nakagaki BN, Mafra K, de Paula CMP, Diniz AB, Costa KMDO, Antunes MM, Oliveira AG, Balderas R, Lopes RP, Menezes GB. High-dimensional intravital microscopy reveals major changes in splenic immune system during postnatal development. Front Immunol 2022; 13:1002919. [PMID: 36531990 PMCID: PMC9755845 DOI: 10.3389/fimmu.2022.1002919] [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: 07/25/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
Spleen is a key organ for immunologic surveillance, acting as a firewall for antigens and parasites that spread through the blood. However, how spleen leukocytes evolve across the developmental phase, and how they spatially organize and interact in vivo is still poorly understood. Using a novel combination of selected antibodies and fluorophores to image in vivo the spleen immune environment, we described for the first time the dynamics of immune development across postnatal period. We found that spleens from adults and infants had similar numbers and arrangement of lymphoid cells. In contrast, splenic immune environment in newborns is sharply different from adults in almost all parameters analysed. Using this in vivo approach, B cells were the most frequent subtype throughout the development. Also, we revealed how infections - using a model of malaria - can change the spleen immune profile in adults and infants, which could become the key to understanding different severity grades of infection. Our new imaging solutions can be extremely useful for different groups in all areas of biological investigation, paving a way for new intravital approaches and advances.
Collapse
Affiliation(s)
- Maria Luiza Mundim Porto-Pedrosa
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Camila Dutra Moreira de Miranda
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mateus Eustáquio Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kassiana Mafra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina Maria Pinto de Paula
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ariane Barros Diniz
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karen Marques de Oliveira Costa
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maisa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - André Gustavo Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Robert Balderas
- BD Biosciences, Department of Biological Sciences, San Jose, CA, United States
| | - Rodrigo Pestana Lopes
- BD Biosciences, Department of Medical & Scientific Affairs, São Paulo, São Paulo, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil,*Correspondence: Gustavo Batista Menezes,
| |
Collapse
|
22
|
Royo J, Camara A, Bertrand B, Batigne P, Coste A, Pipy B, Aubouy A. Kinetics of monocyte subpopulations during experimental cerebral malaria and its resolution in a model of late chloroquine treatment. Front Cell Infect Microbiol 2022; 12:952993. [PMID: 36310859 PMCID: PMC9614070 DOI: 10.3389/fcimb.2022.952993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral malaria (CM) is one of the most severe forms of malaria and is a neuropathology that can lead to death. Monocytes have been shown to accumulate in the brain microvasculature at the onset of neurological symptoms during CM. Monocytes have a remarkable ability to adapt their function to their microenvironment from pro-inflammatory to resolving activities. This study aimed to describe the behavior of monocyte subpopulations during infection and its resolution. C57BL/6 mice were infected with the Plasmodium berghei ANKA strain and treated or not with chloroquine (CQ) on the first day of the onset of neurological symptoms (day 6) for 4 days and followed until day 12 to mimic neuroinflammation and its resolution during experimental CM. Ly6C monocyte subpopulations were identified by flow cytometry of cells from the spleen, peripheral blood, and brain and then quantified and characterized at different time points. In the brain, the Ly6Cint and Ly6Clow monocytes were associated with neuroinflammation, while Ly6Chi and Ly6Cint were mobilized from the peripheral blood to the brain for resolution. During neuroinflammation, CD36 and CD163 were both involved via splenic monocytes, whereas our results suggest that the low CD36 expression in the brain during the neuroinflammation phase was due to degradation. The resolution phase was characterized by increased expressions of CD36 and CD163 in blood Ly6Clow monocytes, a higher expression of CD36 in the microglia, and restored high expression levels of CD163 in Ly6Chi monocytes localized in the brain. Thus, our results suggest that increasing the expressions of CD36 and CD163 specifically in the brain during the neuroinflammatory phase contributes to its resolution.
Collapse
Affiliation(s)
- Jade Royo
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
| | - Aissata Camara
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
- Pharmacy Department, Institut de Recherche et de Développement des Plantes Médicinales et Alimentaires de Guinée (IRDPMAG), Dubréka, Guinea
| | - Benedicte Bertrand
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
| | - Philippe Batigne
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
| | - Agnes Coste
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
| | - Bernard Pipy
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
| | - Agnes Aubouy
- Unité Mixte de Recherche (UMR152) Pharmcochimie et biologie pour le développement (PHARMADEV), Université de Toulouse, French National Research Institue for Sustainable Development (IRD), UPS, Toulouse, France
- *Correspondence: Agnes Aubouy,
| | | |
Collapse
|
23
|
Konozy EHE, Osman MEFM, Ghartey-Kwansah G, Abushama HM. The striking mimics between COVID-19 and malaria: A review. Front Immunol 2022; 13:957913. [PMID: 36081516 PMCID: PMC9445119 DOI: 10.3389/fimmu.2022.957913] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives COVID-19 is a transmissible illness triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since its onset in late 2019 in Wuhan city of China, it continues to spread universally, leading to an ongoing pandemic that shattered all efforts to restrain it. On the other hand, in Africa, the COVID-19 infection may be influenced by malaria coinfection. Hence, in this review article, we aimed to give a comprehensive account of the similarities between COVID-19 and malaria in terms of symptoms, clinical, immunological, and molecular perspectives. Methodology In this article, we reviewed over 50 research papers to highlight the multilayered similarities between COVID-19 and malaria infections that might influence the ontology of COVID-19. Results Despite the poor health and fragile medical system of many sub-Saharan African countries, they persisted with a statistically significantly low number of COVID-19 cases. This was attributed to many factors such as the young population age, the warm weather, the lack of proper diagnosis, previous infection with malaria, the use of antimalarial drugs, etc. Additionally, population genetics appears to play a significant role in shaping the COVID-19 dynamics. This is evident as recent genomic screening analyses of the angiotensin-converting enzyme 2 (ACE2) and malaria-associated-variants identified 6 candidate genes that might play a role in malaria and COVID-19 incidence and severity. Moreover, the clinical and pathological resemblances between the two diseases have made considerable confusion in the diagnosis and thereafter curb the disease in Africa. Therefore, possible similarities between the diseases in regards to the clinical, pathological, immunological, and genetical ascription were discussed. Conclusion Understanding the dynamics of COVID-19 infection in Sub-Saharan Africa and how it is shaped by another endemic disease like malaria can provide insights into how to tailor a successful diagnostic, intervention, and control plans that lower both disease morbidity and mortality.
Collapse
Affiliation(s)
| | | | - George Ghartey-Kwansah
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | | |
Collapse
|
24
|
Aparici Herraiz I, Caires HR, Castillo-Fernández Ó, Sima N, Méndez-Mora L, Risueño RM, Sattabongkot J, Roobsoong W, Hernández-Machado A, Fernandez-Becerra C, Barrias CC, del Portillo HA. Advancing Key Gaps in the Knowledge of Plasmodium vivax Cryptic Infections Using Humanized Mouse Models and Organs-on-Chips. Front Cell Infect Microbiol 2022; 12:920204. [PMID: 35873153 PMCID: PMC9302440 DOI: 10.3389/fcimb.2022.920204] [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: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite representing 36.3% of disease burden in the South-East Asia region and the most predominant species in the region of the Americas. Recent estimates indicate that 3.3 billion of people are under risk of infection with circa 7 million clinical cases reported each year. This burden is certainly underestimated as the vast majority of chronic infections are asymptomatic. For centuries, it has been widely accepted that the only source of cryptic parasites is the liver dormant stages known as hypnozoites. However, recent evidence indicates that niches outside the liver, in particular in the spleen and the bone marrow, can represent a major source of cryptic chronic erythrocytic infections. The origin of such chronic infections is highly controversial as many key knowledge gaps remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Due to ethical and technical considerations, working with the liver, bone marrow and spleen from natural infections is very difficult. Recent advances in the development of humanized mouse models and organs-on-a-chip models, offer novel technological frontiers to study human diseases, vaccine validation and drug discovery. Here, we review current data of these frontier technologies in malaria, highlighting major challenges ahead to study P. vivax cryptic niches, which perpetuate transmission and burden.
Collapse
Affiliation(s)
- Iris Aparici Herraiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Hugo R. Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Óscar Castillo-Fernández
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Núria Sima
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Méndez-Mora
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Aurora Hernández-Machado
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Hernando A. del Portillo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- *Correspondence: Hernando A. del Portillo,
| |
Collapse
|
25
|
IL-10 Producing Regulatory B Cells Mediated Protection against Murine Malaria Pathogenesis. BIOLOGY 2022; 11:biology11050669. [PMID: 35625397 PMCID: PMC9138363 DOI: 10.3390/biology11050669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 02/06/2023]
Abstract
Simple Summary The immunomodulatory role of B cell subset called regulatory B cells was evaluated during Plasmodium infection to study their role in susceptibility or resistance during infection. The expansion of regulatory B cells during Plasmodium infection indicated their important role in regulating the immune response. Adoptive transfer of regulatory B cells following infection with a lethal parasite resulted in enhanced survival of mice and inhibited growth of the Plasmodium parasite. Moreover, by inhibiting the production of the pro-inflammatory cytokine, IFN-γ, and stimulating anti-inflammatory IL-10 production, regulatory B cells may serve as an important contributor to protective immune response. Abstract Various immune cells are known to participate in combating infection. Regulatory B cells represent a subset of B cells that take part in immunomodulation and control inflammation. The immunoregulatory function of regulatory B cells has been shown in various murine models of several disorders. In this study, a comparable IL-10 competent B-10 cell subset (regulatory B cells) was characterized during lethal and non-lethal infection with malaria parasites using the mouse model. We observed that infection of Balb/c mice with P. yoelii I 7XL was lethal, and a rapid increase in dynamics of IL-10 producing B220+CD5+CD1d+ regulatory B cells over the course of infection was observed. However, animals infected with a less virulent strain of the parasite P. yoelii I7XNL attained complete resistance. It was observed that there is an increase in the population of regulatory B cells with an increase of parasitemia; however, a sudden drop in the frequency of these cells was observed with parasite clearance. Adoptive transfer of regulatory B cells to naïve mice followed by infection results in slow parasite growth and enhancement of survival in P. yoelii 17XL (lethal) infected animals. Adoptively transferred regulatory B cells also resulted in decreased production of pro-inflammatory cytokine (IFN-γ) and enhanced production of anti-inflammatory cytokine (IL-10). It infers that these regulatory B cells may contribute in immune protection by preventing the inflammation associated with disease and inhibiting the parasite growth.
Collapse
|
26
|
Rodriguez-Muñoz D, Sánchez Á, Pérez-Benavente S, Contreras-Jurado C, Montero-Pedrazuela A, Toledo-Castillo M, Gutiérrez-Hernández M, Rodrigues-Díez R, Folgueira C, Briones AM, Sabio G, Monedero-Cobeta I, Chávez-Coira I, Castejón D, Fernández-Valle E, Regadera J, Bautista JM, Aranda A, Alemany S. Hypothyroidism confers tolerance to cerebral malaria. SCIENCE ADVANCES 2022; 8:eabj7110. [PMID: 35385300 PMCID: PMC8985923 DOI: 10.1126/sciadv.abj7110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The modulation of the host's metabolism to protect tissue from damage induces tolerance to infections increasing survival. Here, we examined the role of the thyroid hormones, key metabolic regulators, in the outcome of malaria. Hypothyroidism confers protection to experimental cerebral malaria by a disease tolerance mechanism. Hypothyroid mice display increased survival after infection with Plasmodium berghei ANKA, diminishing intracranial pressure and brain damage, without altering pathogen burden, blood-brain barrier disruption, or immune cell infiltration. This protection is reversed by treatment with a Sirtuin 1 inhibitor, while treatment of euthyroid mice with a Sirtuin 1 activator induces tolerance and reduces intracranial pressure and lethality. This indicates that thyroid hormones and Sirtuin 1 are previously unknown targets for cerebral malaria treatment, a major killer of children in endemic malaria areas.
Collapse
Affiliation(s)
- Diego Rodriguez-Muñoz
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ángela Sánchez
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Pérez-Benavente
- Department of Biochemistry and Molecular Biology, Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Constanza Contreras-Jurado
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Alfonso X El Sabio, Villanueva de la Cañada, Madrid, Spain
| | - Ana Montero-Pedrazuela
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Toledo-Castillo
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Gutiérrez-Hernández
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Raquel Rodrigues-Díez
- Departament of Pharmacology, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| | - Cintia Folgueira
- Departament of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares, ISCIII, 28029 Madrid, Spain
| | - Ana M. Briones
- Departament of Pharmacology, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| | - Guadalupe Sabio
- Departament of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares, ISCIII, 28029 Madrid, Spain
| | | | - Irene Chávez-Coira
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Castejón
- MNR Unit (CAI de Bioimagen), Universidad Complutense de Madrid, Madrid, Spain
| | | | - Javier Regadera
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology, Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Aranda
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Biomedicine Unit (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Madrid, Spain
- Corresponding author. (A.A.); (S.A.)
| | - Susana Alemany
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Biomedicine Unit (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Corresponding author. (A.A.); (S.A.)
| |
Collapse
|
27
|
Matthews K, Lamoureux ES, Myrand-Lapierre ME, Duffy SP, Ma H. Technologies for measuring red blood cell deformability. LAB ON A CHIP 2022; 22:1254-1274. [PMID: 35266475 DOI: 10.1039/d1lc01058a] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Human red blood cells (RBCs) are approximately 8 μm in diameter, but must repeatedly deform through capillaries as small as 2 μm in order to deliver oxygen to all parts of the body. The loss of this capability is associated with the pathology of many diseases, and is therefore a potential biomarker for disease status and treatment efficacy. Measuring RBC deformability is a difficult problem because of the minute forces (∼pN) that must be exerted on these cells, as well as the requirements for throughput and multiplexing. The development of technologies for measuring RBC deformability date back to the 1960s with the development of micropipette aspiration, ektacytometry, and the cell transit analyzer. In the past 10 years, significant progress has been made using microfluidics by leveraging the ability to precisely control fluid flow through microstructures at the size scale of individual RBCs. These technologies have now surpassed traditional methods in terms of sensitivity, throughput, consistency, and ease of use. As a result, these efforts are beginning to move beyond feasibility studies and into applications to enable biomedical discoveries. In this review, we provide an overview of both traditional and microfluidic techniques for measuring RBC deformability. We discuss the capabilities of each technique and compare their sensitivity, throughput, and robustness in measuring bulk and single-cell RBC deformability. Finally, we discuss how these tools could be used to measure changes in RBC deformability in the context of various applications including pathologies caused by malaria and hemoglobinopathies, as well as degradation during storage in blood bags prior to blood transfusions.
Collapse
Affiliation(s)
- Kerryn Matthews
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, V6T 1Z4, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Erik S Lamoureux
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, V6T 1Z4, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Marie-Eve Myrand-Lapierre
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, V6T 1Z4, Canada.
| | - Simon P Duffy
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- British Columbia Institute of Technology, Vancouver, BC, Canada
| | - Hongshen Ma
- Department of Mechanical Engineering, University of British Columbia, 2054-6250 Applied Science Lane, Vancouver, BC, V6T 1Z4, Canada.
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Science, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
28
|
Wunderlich F, Delic D, Gerovska D, Araúzo-Bravo MJ. Vaccination Accelerates Liver-Intrinsic Expression of Megakaryocyte-Related Genes in Response to Blood-Stage Malaria. Vaccines (Basel) 2022; 10:vaccines10020287. [PMID: 35214745 PMCID: PMC8880532 DOI: 10.3390/vaccines10020287] [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: 11/29/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Erythropoiesis and megakaryo-/thrombopoiesis occur in the bone marrow proceeding from common, even bipotent, progenitor cells. Recently, we have shown that protective vaccination accelerates extramedullary hepatic erythroblastosis in response to blood-stage malaria of Plasmodium chabaudi. Here, we investigated whether protective vaccination also accelerates extramedullary hepatic megakaryo-/thrombopoiesis. Female Balb/c mice were twice vaccinated with a non-infectious vaccine before infecting with 106 P. chabaudi-parasitized erythrocytes. Using gene expression microarrays and quantitative real-time PCR, transcripts of genes known to be expressed in the bone marrow by cells of the megakaryo-/thrombocytic lineage were compared in livers of vaccination-protected and unprotected mice on days 0, 1, 4, 8, and 11 p.i. Livers of vaccination-protected mice responded with expression of megakaryo-/thrombocytic genes faster to P. chabaudi than those of unvaccinated mice, evidenced at early patency on day 4 p.i., when livers exhibited significantly higher levels of malaria-induced transcripts of the genes Selp and Pdgfb (p-values < 0.0001), Gp5 (p-value < 0.001), and Fli1, Runx1, Myb, Mpl, Gp1ba, Gp1bb, Gp6, Gp9, Pf4, and Clec1b (p-values < 0.01). Together with additionally analyzed genes known to be related to megakaryopoiesis, our data suggest that protective vaccination accelerates liver-intrinsic megakaryo-/thrombopoiesis in response to blood-stage malaria that presumably contributes to vaccination-induced survival of otherwise lethal blood-stage malaria.
Collapse
Affiliation(s)
- Frank Wunderlich
- Department of Biology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Denis Delic
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, 68167 Heidelberg, Germany
- Correspondence: (D.D.); (M.J.A.-B.)
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- TransBioNet Thematic Network of Excellence for Transitional Bioinformatics, Barcelona Supercomputing Center, 08034 Barcelona, Spain
- Correspondence: (D.D.); (M.J.A.-B.)
| |
Collapse
|
29
|
Fernandez-Becerra C, Aparici-Herraiz I, Del Portillo HA. Cryptic erythrocytic infections in Plasmodium vivax, another challenge to its elimination. Parasitol Int 2021; 87:102527. [PMID: 34896615 DOI: 10.1016/j.parint.2021.102527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/04/2021] [Accepted: 12/04/2021] [Indexed: 12/26/2022]
Abstract
Human malaria caused by Plasmodium vivax infection (vivax malaria) is a major global health issue. It is the most geographically widespread form of the disease, accounting for 7 million annual clinical cases, the majority of cases in America and Asia and an estimation of over 2.5 billion people living under risk of infection. The general perception towards vivax malaria has shifted recently, following a series of reports, from being viewed as a benign infection to the recognition of its potential for more severe manifestations including fatal cases. However, the underlying pathogenic mechanisms of vivax malaria remain largely unresolved. Asymptomatic carriers of malaria parasites are a major challenge for malaria elimination. In the case of P. vivax, it has been widely accepted that the only source of cryptic parasites is hypnozoite dormant stages. Here, we will review new evidence indicating that cryptic erythrocytic niches outside the liver, in particular in the spleen and bone marrow, can represent a major source of asymptomatic infections. The origin of such parasites is being controversial and many key gaps in the knowledge of such infections remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Last, we will glimpse into the role of reticulocyte-derived exosomes, extracellular vesicles of endocytic origin, as intercellular communicators likely involved in the formation of such cryptic erythrocytic infections.
Collapse
Affiliation(s)
- Carmen Fernandez-Becerra
- ISGlobal, Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain.
| | | | - Hernando A Del Portillo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, 08036 Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
30
|
Morphology of the Spleen in Oreochromis niloticus: Splenic Subregions and the Blood-Spleen Barrier. Animals (Basel) 2021; 11:ani11102934. [PMID: 34679955 PMCID: PMC8532917 DOI: 10.3390/ani11102934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
The spleen is a separate organ of the teleost, playing an essential role in immune reactions. The morphology of the spleen is different from the fish species. Little knowledge about the spleen structure and the blood splenic barrier (BSB) in Nile tilapia has been reported. To address this issue, we studied the histology of the spleen and the BSB in healthy Nile tilapia. The morphology of the spleen was observed, then H&E staining, modified Jame's staining, and ultrastructural techniques were performed to portion the spleen into three subregions and analyze the location of components and fibers. Thereafter, vital staining of Nile tilapia with Trypan blue was conducted to elucidate the composition and function of BSB. Histologically, the spleen could be divided into three subregions (inner, middle, and outer). The venules, clumps of lymphocytes, and vessels were separately characterized features of the outer, middle, and inner layers. Post injection, Trypan blue was intercepted in the endotheliocytes of ellipsoids in the middle layer (i.p.) or was deposited to the reticular fibers surrounding the ellipsoids (i.v.). Additionally, the amount of Trypan blue was shown to be positively correlated to that of the Acid phosphatase expressed. In conclusion, the spleen could be portioned into three subregions, and the BSB lay in the middle layer, composed of the cuboidal-shaped endotheliocytes and the surrounding reticular fibers of the ellipsoid capillaries. The present study enriched the research of immune tissues and system in tilapia and provided reference for the study of spleen in other fish species.
Collapse
|
31
|
Deroost K, Alder C, Hosking C, McLaughlin S, Lin JW, Lewis MD, Saavedra-Torres Y, Addy JWG, Levy P, Giorgalli M, Langhorne J. Tissue macrophages and interferon-gamma signalling control blood-stage Plasmodium chabaudi infections derived from mosquito-transmitted parasites. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:104-119. [PMID: 34532703 PMCID: PMC8428512 DOI: 10.1016/j.crimmu.2021.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 11/19/2022] Open
Abstract
Natural infection with Plasmodium parasites, the causative agents of malaria, occurs via mosquito vectors. However, most of our knowledge of the immune response to the blood stages of Plasmodium is from infections initiated by injection of serially blood-passaged infected red blood cells, resulting in an incomplete life cycle in the mammalian host. Vector transmission of the rodent malaria parasite, Plasmodium chabaudi chabaudi AS has been shown to give rise to a more attenuated blood-stage infection in C57Bl/6J mice, when compared to infections initiated with serially blood-passaged P. chabaudi-infected red blood cells. In mouse models, the host immune response induced by parasites derived from natural mosquito transmission is likely to more closely resemble the immune responses to Plasmodium infections in humans. It is therefore important to determine how the host response differs between the two types of infections. As the spleen is considered to be a major contributor to the protective host response to P. chabaudi, we carried out a comparative transcriptomic analysis of the splenic response to recently mosquito-transmitted and serially blood-passaged parasites in C57Bl/6J mice. The attenuated infection arising from recently mosquito-transmitted parasites is characterised by an earlier and stronger myeloid- and IFNγ-related response. Analyses of spleen lysates from the two infections similarly showed stronger or earlier inflammatory cytokine and chemokine production in the recently mosquito-transmitted blood-stage infections. Furthermore, tissue macrophages, including red pulp macrophages, and IFNγ-signalling in myeloid cells, are required for the early control of P. chabaudi recently mosquito-transmitted parasites, thus contributing to the attenuation of mosquito-transmitted infections. The molecules responsible for this early activation response to recently-transmitted blood-stage parasites in mice would be important to identify, as they may help to elucidate the nature of the initial interactions between blood-stage parasites and the host immune system in naturally transmitted malaria. Attenuation of recently transmitted malaria happens at blood stage of infection. Stronger or earlier inflammatory cytokine and chemokine production. Tissue macrophages, including red pulp macrophages, contribute to attenuation. IFNγ-signalling in myeloid cells is required for early control of P. chabaudi AS.
Collapse
|
32
|
Immunoprofiles associated with controlled human malaria infection and naturally acquired immunity identify a shared IgA pre-erythrocytic immunoproteome. NPJ Vaccines 2021; 6:115. [PMID: 34518543 PMCID: PMC8438027 DOI: 10.1038/s41541-021-00363-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Knowledge of the Plasmodium falciparum antigens that comprise the human liver stage immunoproteome is important for pre-erythrocytic vaccine development, but, compared with the erythrocytic stage immunoproteome, more challenging to classify. Previous studies of P. falciparum antibody responses report IgG and rarely IgA responses. We assessed IgG and IgA antibody responses in adult sera collected during two controlled human malaria infection (CHMI) studies in malaria-naïve volunteers and in 1- to 6-year-old malaria-exposed Malian children on a 251 P. falciparum antigen protein microarray. IgG profiles in the two CHMI groups were equivalent and differed from Malian children. IgA profiles were robust in the CHMI groups and a subset of Malian children. We describe immunoproteome differences in naïve vs. exposed individuals and report pre-erythrocytic proteins recognized by the immune system. IgA responses detected in this study expand the list of pre-erythrocytic antigens for further characterization as potential vaccine candidates.
Collapse
|
33
|
Vieira-Santos F, Leal-Silva T, de Lima Silva Padrão L, Ruas ACL, Nogueira DS, Kraemer L, Oliveira FMS, Caliari MV, Russo RC, Fujiwara RT, Bueno LL. Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malar J 2021; 20:296. [PMID: 34210332 PMCID: PMC8248286 DOI: 10.1186/s12936-021-03824-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/17/2021] [Indexed: 12/22/2022] Open
Abstract
Background Ascariasis and malaria are highly prevalent parasitic diseases in tropical regions and often have overlapping endemic areas, contributing to high morbidity and mortality rates in areas with poor sanitary conditions. Several studies have previously aimed to correlate the effects of Ascaris-Plasmodium coinfections but have obtained contradictory and inconclusive results. Therefore, the present study aimed to investigate parasitological and immunopathological aspects of the lung during murine experimental concomitant coinfection by Plasmodium berghei and Ascaris suum during larvae ascariasis. Methods C57BL/6J mice were inoculated with 1 × 104P. berghei strain NK65-NY-infected red blood cells (iRBCs) intraperitoneally and/or 2500 embryonated eggs of A. suum by oral gavage. P. berghei parasitaemia, morbidity and the survival rate were assessed. On the seventh day postinfection (dpi), A. suum lung burden analysis; bronchoalveolar lavage (BAL); histopathology; NAG, MPO and EPO activity measurements; haematological analysis; and respiratory mechanics analysis were performed. The concentrations of interleukin (IL)-1β, IL-12/IL-23p40, IL-6, IL-4, IL-33, IL-13, IL-5, IL-10, IL-17A, IFN-γ, TNF and TGF-β were assayed by sandwich ELISA. Results Animals coinfected with P. berghei and A. suum show decreased production of type 1, 2, and 17 and regulatory cytokines; low leukocyte recruitment in the tissue; increased cellularity in the circulation; and low levels of NAG, MPO and EPO activity that lead to an increase in larvae migration, as shown by the decrease in larvae recovered in the lung parenchyma and increase in larvae recovered in the airway. This situation leads to severe airway haemorrhage and, consequently, an impairment respiratory function that leads to high morbidity and early mortality. Conclusions This study demonstrates that the Ascaris-Plasmodium interaction is harmful to the host and suggests that this coinfection may potentiate Ascaris-associated pathology by dampening the Ascaris-specific immune response, resulting in the early death of affected animals. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03824-w.
Collapse
Affiliation(s)
- Flaviane Vieira-Santos
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Leal-Silva
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza de Lima Silva Padrão
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Loiola Ruas
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Silva Nogueira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Kraemer
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Laboratory of Protozooses, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Institute of Biological Sciences, Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
34
|
Cervantes-Candelas LA, Aguilar-Castro J, Buendía-González FO, Fernández-Rivera O, Cervantes-Sandoval A, Morales-Montor J, Legorreta-Herrera M. Tamoxifen Suppresses the Immune Response to Plasmodium berghei ANKA and Exacerbates Symptomatology. Pathogens 2021; 10:pathogens10060743. [PMID: 34204678 PMCID: PMC8231265 DOI: 10.3390/pathogens10060743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/25/2022] Open
Abstract
Malaria is the most lethal parasitic disease in the world. Mortality and severity in symptoms are higher in men than women, suggesting that oestrogens, which are in higher concentration in females than in males, may regulate the immune response against malaria. Tamoxifen, a selective oestrogen receptor modulator used in breast cancer treatment due to its antagonistic effect on oestrogen receptors α and β, is also studied because of its potential therapeutic use for several parasitic diseases. However, most studies, including one in malaria, have not addressed the immunomodulatory role of tamoxifen. In this work, we evaluated the effect of tamoxifen on the immune response of CBA/Ca mice against Plasmodium berghei ANKA. This study showed for the first time that tamoxifen increased parasite load, aggravated symptoms by decreasing body temperature and body weight, and worsened anaemia. Additionally, tamoxifen significantly increased the splenic index and the percentages of CD4+ and NK+ cells on day eight post-infection. By contrast, tamoxifen decreased both CD8+ and B220+ populations in the spleen and decreased the serum levels of IL-2, IL-6, and IL-17. Our findings support the notion that tamoxifen is a potent immunomodulator in malaria-infected mice and suggest caution when administering it to malaria-infected women with breast cancer.
Collapse
Affiliation(s)
- Luis Antonio Cervantes-Candelas
- Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Laboratorio de Inmunología Molecular, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (L.A.C.-C.); (J.A.-C.); (F.O.B.-G.); (O.F.-R.)
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, Mexico
| | - Jesús Aguilar-Castro
- Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Laboratorio de Inmunología Molecular, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (L.A.C.-C.); (J.A.-C.); (F.O.B.-G.); (O.F.-R.)
| | - Fidel Orlando Buendía-González
- Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Laboratorio de Inmunología Molecular, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (L.A.C.-C.); (J.A.-C.); (F.O.B.-G.); (O.F.-R.)
| | - Omar Fernández-Rivera
- Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Laboratorio de Inmunología Molecular, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (L.A.C.-C.); (J.A.-C.); (F.O.B.-G.); (O.F.-R.)
| | - Armando Cervantes-Sandoval
- Laboratorio de Aplicaciones Computacionales, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico;
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, AP 70228, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Martha Legorreta-Herrera
- Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Laboratorio de Inmunología Molecular, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico; (L.A.C.-C.); (J.A.-C.); (F.O.B.-G.); (O.F.-R.)
- Correspondence: ; Tel.: +52-5556230700 (ext. 83207)
| |
Collapse
|
35
|
Galán-Salinas A, Corral-Ruíz G, Pérez-Vega MJ, Fabila-Castillo L, Silva-García R, Marquina-Castillo B, León-Contreras JC, Barrios-Payán J, Francisco-Cruz A, Montecillo-Aguado M, Huerta-Yepez S, Calderón-Amador J, Flores-Romo L, Hernández-Pando R, Sánchez-Torres LE. Monocyte Locomotion Inhibitory Factor confers neuroprotection and prevents the development of murine cerebral malaria. Int Immunopharmacol 2021; 97:107674. [PMID: 34044183 DOI: 10.1016/j.intimp.2021.107674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/30/2021] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
Cerebral malaria (CM) is a neurological complication derived from the Plasmodium falciparum infection in humans. The mechanisms involved in the disease progression are still not fully understood, but both the sequestration of infected red blood cells (iRBC) and leukocytes and an exacerbated host inflammatory immune response are significant factors. In this study, we investigated the effect of Monocyte Locomotion Inhibitory Factor (MLIF), an anti-inflammatory peptide, in a well-characterized murine model of CM. Our data showed that the administration of MLIF increased the survival and avoided the neurological signs of CM in Plasmodium berghei ANKA (PbA) infected C57BL/6 mice. MLIF administration down-regulated systemic inflammatory mediators such as IFN-γ, TNF-α, IL-6, CXCL2, and CCL2, as well as the in situ expression of TNF-α in the brain. In the same way, MLIF reduced the expression of CD31, CD36, CD54, and CD106 in the cerebral endothelium of infected animals and prevented the sequestration of iRBC and leucocytes in the brain microvasculature. Furthermore, MLIF inhibited the activation of astrocytes and microglia and preserved the integrity of the blood-brain barrier (BBB). In conclusion, our results demonstrated that the administration of MLIF increased survival and conferred neuroprotection by decreasing neuroinflammation in murine CM.
Collapse
Affiliation(s)
- A Galán-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - G Corral-Ruíz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - M J Pérez-Vega
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - L Fabila-Castillo
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - R Silva-García
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, CMN-Siglo XXI, IMSS, México City, Mexico
| | - B Marquina-Castillo
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - J C León-Contreras
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - J Barrios-Payán
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - A Francisco-Cruz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Montecillo-Aguado
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México, Federico Gómez, México City, Mexico
| | - S Huerta-Yepez
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México, Federico Gómez, México City, Mexico
| | - J Calderón-Amador
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, Instituto Politécnico Nacional, México City, Mexico
| | - L Flores-Romo
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, Instituto Politécnico Nacional, México City, Mexico
| | - R Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico.
| | - L E Sánchez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico.
| |
Collapse
|
36
|
Mandal RK, Denny JE, Namazzi R, Opoka RO, Datta D, John CC, Schmidt NW. Dynamic modulation of spleen germinal center reactions by gut bacteria during Plasmodium infection. Cell Rep 2021; 35:109094. [PMID: 33979614 PMCID: PMC8141963 DOI: 10.1016/j.celrep.2021.109094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/08/2021] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
Gut microbiota educate the local and distal immune system in early life to imprint long-term immunological outcomes while maintaining the capacity to dynamically modulate the local mucosal immune system throughout life. It is unknown whether gut microbiota provide signals that dynamically regulate distal immune responses following an extra-gastrointestinal infection. We show here that gut bacteria composition correlated with the severity of malaria in children. Using the murine model of malaria, we demonstrate that parasite burden and spleen germinal center reactions are malleable to dynamic cues provided by gut bacteria. Whereas antibiotic-induced changes in gut bacteria have been associated with immunopathology or impairment of immunity, the data demonstrate that antibiotic-induced changes in gut bacteria can enhance immunity to Plasmodium. This effect is not universal but depends on baseline gut bacteria composition. These data demonstrate the dynamic communications that exist among gut bacteria, the gut-distal immune system, and control of Plasmodium infection.
Collapse
Affiliation(s)
- Rabindra K Mandal
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA; Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Joshua E Denny
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
| | - Ruth Namazzi
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Robert O Opoka
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Dibyadyuti Datta
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA; Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
37
|
Soon MSF, Nalubega M, Boyle MJ. T-follicular helper cells in malaria infection and roles in antibody induction. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab008. [PMID: 36845571 PMCID: PMC9914587 DOI: 10.1093/oxfimm/iqab008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/29/2023] Open
Abstract
Immunity to malaria is mediated by antibodies that block parasite replication to limit parasite burden and prevent disease. Cytophilic antibodies have been consistently shown to be associated with protection, and recent work has improved our understanding of the direct and Fc-mediated mechanisms of protective antibodies. Antibodies also have important roles in vaccine-mediated immunity. Antibody induction is driven by the specialized CD4+ T cells, T-follicular helper (Tfh) cells, which function within the germinal centre to drive B-cell activation and antibody induction. In humans, circulating Tfh cells can be identified in peripheral blood and are differentiated into subsets that appear to have pathogen/vaccination-specific roles in antibody induction. Tfh cell responses are essential for protective immunity from Plasmodium infection in murine models of malaria. Our understanding of the activation of Tfh cells during human malaria infection and the importance of different Tfh cell subsets in antibody development is still emerging. This review will discuss our current knowledge of Tfh cell activation and development in malaria, and the potential avenues and pitfalls of targeting Tfh cells to improve malaria vaccines.
Collapse
Affiliation(s)
- Megan S F Soon
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Mayimuna Nalubega
- Infectious Diseases Research Collaboration, Tororo District Hospital, Tororo, Uganda
| | - Michelle J Boyle
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia,Correspondence address. QIMR Berghofer Medical Research Institute, Brisbane, Australia. E-mail:
| |
Collapse
|
38
|
Cao H, Antonopoulos A, Henderson S, Wassall H, Brewin J, Masson A, Shepherd J, Konieczny G, Patel B, Williams ML, Davie A, Forrester MA, Hall L, Minter B, Tampakis D, Moss M, Lennon C, Pickford W, Erwig L, Robertson B, Dell A, Brown GD, Wilson HM, Rees DC, Haslam SM, Alexandra Rowe J, Barker RN, Vickers MA. Red blood cell mannoses as phagocytic ligands mediating both sickle cell anaemia and malaria resistance. Nat Commun 2021; 12:1792. [PMID: 33741926 PMCID: PMC7979802 DOI: 10.1038/s41467-021-21814-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
In both sickle cell disease and malaria, red blood cells (RBCs) are phagocytosed in the spleen, but receptor-ligand pairs mediating uptake have not been identified. Here, we report that patches of high mannose N-glycans (Man5-9GlcNAc2), expressed on diseased or oxidized RBC surfaces, bind the mannose receptor (CD206) on phagocytes to mediate clearance. We find that extravascular hemolysis in sickle cell disease correlates with high mannose glycan levels on RBCs. Furthermore, Plasmodium falciparum-infected RBCs expose surface mannose N-glycans, which occur at significantly higher levels on infected RBCs from sickle cell trait subjects compared to those lacking hemoglobin S. The glycans are associated with high molecular weight complexes and protease-resistant, lower molecular weight fragments containing spectrin. Recognition of surface N-linked high mannose glycans as a response to cellular stress is a molecular mechanism common to both the pathogenesis of sickle cell disease and resistance to severe malaria in sickle cell trait.
Collapse
Affiliation(s)
- Huan Cao
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | | | - Sadie Henderson
- grid.476695.f0000 0004 0495 4557Scottish National Blood Transfusion Service, Aberdeen, UK
| | - Heather Wassall
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - John Brewin
- grid.46699.340000 0004 0391 9020Department of Haematology, King’s College Hospital, London, UK
| | - Alanna Masson
- grid.417581.e0000 0000 8678 4766Department of Haematology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Jenna Shepherd
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Gabriela Konieczny
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Bhinal Patel
- grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, UK
| | - Maria-Louise Williams
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Adam Davie
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Megan A. Forrester
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Lindsay Hall
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Beverley Minter
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Dimitris Tampakis
- grid.13097.3c0000 0001 2322 6764Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University and Division of Cancer Studies, King’s College London, London, UK
| | - Michael Moss
- grid.476695.f0000 0004 0495 4557Scottish National Blood Transfusion Service, Aberdeen, UK
| | - Charlotte Lennon
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Wendy Pickford
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Lars Erwig
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Beverley Robertson
- grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, UK
| | - Anne Dell
- grid.46699.340000 0004 0391 9020Department of Haematology, King’s College Hospital, London, UK
| | - Gordon D. Brown
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK ,grid.8391.30000 0004 1936 8024Medical Medical Research Council Centre for Medical Mycology at the University of Exeter, Exeter, UK
| | - Heather M. Wilson
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - David C. Rees
- grid.46699.340000 0004 0391 9020Department of Haematology, King’s College Hospital, London, UK
| | - Stuart M. Haslam
- grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, UK
| | - J. Alexandra Rowe
- grid.4305.20000 0004 1936 7988Centre for Immunity, Infection and Evolution, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Robert N. Barker
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Mark A. Vickers
- grid.7107.10000 0004 1936 7291School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK ,grid.476695.f0000 0004 0495 4557Scottish National Blood Transfusion Service, Aberdeen, UK ,grid.417581.e0000 0000 8678 4766Department of Haematology, Aberdeen Royal Infirmary, Aberdeen, UK
| |
Collapse
|
39
|
Gualdrón-López M, Díaz-Varela M, Toda H, Aparici-Herraiz I, Pedró-Cos L, Lauzurica R, Lacerda MVG, Fernández-Sanmartín MA, Fernandez-Becerra C, Del Portillo HA. Multiparameter Flow Cytometry Analysis of the Human Spleen Applied to Studies of Plasma-Derived EVs From Plasmodium vivax Patients. Front Cell Infect Microbiol 2021; 11:596104. [PMID: 33732657 PMCID: PMC7957050 DOI: 10.3389/fcimb.2021.596104] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/05/2021] [Indexed: 11/13/2022] Open
Abstract
The spleen is a secondary lymphoid organ with multiple functions including the removal of senescent red blood cells and the coordination of immune responses against blood-borne pathogens, such as malaria parasites. Despite the major role of the spleen, the study of its function in humans is limited by ethical implications to access human tissues. Here, we employed multiparameter flow cytometry combined with cell purification techniques to determine human spleen cell populations from transplantation donors. Spleen immuno-phenotyping showed that CD45+ cells included B (30%), CD4+ T (16%), CD8+ T (10%), NK (6%) and NKT (2%) lymphocytes. Myeloid cells comprised neutrophils (16%), monocytes (2%) and DCs (0.3%). Erythrocytes represented 70%, reticulocytes 0.7% and hematopoietic stem cells 0.02%. Extracellular vesicles (EVs) are membrane-bound nanoparticles involved in intercellular communication and secreted by almost all cell types. EVs play several roles in malaria that range from modulation of immune responses to vascular alterations. To investigate interactions of plasma-derived EVs from Plasmodium vivax infected patients (PvEVs) with human spleen cells, we used size-exclusion chromatography (SEC) to separate EVs from the bulk of soluble plasma proteins and stained isolated EVs with fluorescent lipophilic dyes. The integrated cellular analysis of the human spleen and the methodology employed here allowed in vitro interaction studies of human spleen cells and EVs that showed an increased proportion of T cells (CD4+ 3 fold and CD8+ 4 fold), monocytes (1.51 fold), B cells (2.3 fold) and erythrocytes (3 fold) interacting with PvEVs as compared to plasma-derived EVs from healthy volunteers (hEVs). Future functional studies of these interactions can contribute to unveil pathophysiological processes involving the spleen in vivax malaria.
Collapse
Affiliation(s)
- Melisa Gualdrón-López
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain
| | | | - Haruka Toda
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| | | | - Laura Pedró-Cos
- IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Ricardo Lauzurica
- Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Marcus V G Lacerda
- Fundaçao de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Instituto Leônidas & Maria Deane (ILMD), Fiocruz, Manaus, Brazil
| | | | - Carmen Fernandez-Becerra
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Hernando A Del Portillo
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,IGTP: Germans Trias i Pujol Research Institute, Barcelona, Spain.,ICREA: Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| |
Collapse
|
40
|
Corbett Y, Parapini S, Perego F, Messina V, Delbue S, Misiano P, Falchi M, Silvestrini F, Taramelli D, Basilico N, D'Alessandro S. Phagocytosis and activation of bone marrow-derived macrophages by Plasmodium falciparum gametocytes. Malar J 2021; 20:81. [PMID: 33568138 PMCID: PMC7874634 DOI: 10.1186/s12936-021-03589-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/08/2021] [Indexed: 02/01/2023] Open
Abstract
Background The innate immune response against various life cycle stages of the malaria parasite plays an important role in protection against the disease and regulation of its severity. Phagocytosis of asexual erythrocytic stages is well documented, but little and contrasting results are available about phagocytic clearance of sexual stages, the gametocytes, which are responsible for the transmission of the parasites from humans to mosquitoes. Similarly, activation of host macrophages by gametocytes has not yet been carefully addressed. Methods Phagocytosis of early or late Plasmodium falciparum gametocytes was evaluated through methanol fixed cytospin preparations of immortalized mouse C57Bl/6 bone marrow-derived macrophages treated for 2 h with P. falciparum and stained with Giemsa, and it was confirmed through a standardized bioluminescent method using the transgenic P. falciparum 3D7elo1-pfs16-CBG99 strain. Activation was evaluated by measuring nitric oxide or cytokine levels in the supernatants of immortalized mouse C57Bl/6 bone marrow-derived macrophages treated with early or late gametocytes. Results The results showed that murine bone marrow-derived macrophages can phagocytose both early and late gametocytes, but only the latter were able to induce the production of inflammatory mediators, specifically nitric oxide and the cytokines tumour necrosis factor and macrophage inflammatory protein 2. Conclusions These results support the hypothesis that developing gametocytes interact in different ways with innate immune cells of the host. Moreover, the present study proposes that early and late gametocytes act differently as targets for innate immune responses.
Collapse
Affiliation(s)
- Yolanda Corbett
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy. .,Centro Interuniversitario di Ricerca sulla Malaria-Italian Malaria Network, Milan, Italy.
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria-Italian Malaria Network, Milan, Italy
| | - Federica Perego
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy
| | - Valeria Messina
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Serena Delbue
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy
| | - Paola Misiano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy
| | - Mario Falchi
- AIDS-Ricerca e sviluppo, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Silvestrini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy.,Centro Interuniversitario di Ricerca sulla Malaria-Italian Malaria Network, Milan, Italy
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria-Italian Malaria Network, Milan, Italy
| | - Nicoletta Basilico
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria-Italian Malaria Network, Milan, Italy
| | - Sarah D'Alessandro
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy. .,Centro Interuniversitario di Ricerca sulla Malaria-Italian Malaria Network, Milan, Italy. .,Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Pascal 36, 20133, Milan, Italy.
| |
Collapse
|
41
|
Djouwoug CN, Gounoue RK, Ngueguim FT, NankapTsakem JM, Gouni CD, Kandeda AC, Ngouela S, Lenta BN, Sewald N, Fekam FB, Dimo T. In vitro and in vivo antiplasmodial activity of hydroethanolic bark extract of Bridelia atroviridis müll. Arg. (Euphorbiaceae) and lc-ms-based phytochemical analysis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113424. [PMID: 33010404 DOI: 10.1016/j.jep.2020.113424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/03/2020] [Accepted: 09/24/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Malaria is a life-threatening health problem worldwide and treatment remains a major challenge. Natural products from medicinal plants are credible sources for better anti-malarial drugs. AIM OF THE STUDY This study aimed at assessing the in vitro and in vivo antiplasmodial activities of the hydroethanolic extract of Bridelia atroviridis bark. MATERIALS AND METHODS The phytochemical characterization of Bridelia atroviridis extract was carried out by High-Performance Liquid Chromatography-Mass spectrometry (HPLC-MS). The cytotoxicity test on Vero cells was carried out using the resazurin-based assay while the in vitro antiplasmodial activity was determined on Plasmodium falciparum (Dd2 strain, chloroquine resistant) using the SYBR green I-based fluorescence assay. The in vivo assay was performed on Plasmodium berghei-infected rats daily treated for 5 days with distilled water (10 mL/kg) for malaria control, 25 mg/kg of chloroquine sulfate for positive control and 50, 100 and 200 mg/kg of B. atroviridis extract for the three test groups. Parasitaemia was daily monitored using 10% giemsa-staining thin blood smears. At the end of the treatment, animals were sacrificed, blood was collected for hematological and biochemical analysis while organs were removed for biochemical and histopathological analyses. RESULTS The HPLC-MS analysis data of B. atroviridis revealed the presence of bridelionoside D, isomyricitrin, corilagin, myricetin and 5 others compounds not yet identified. Bridelia atroviridis exhibited good in vitro antiplasmodial activity with the IC50 evaluated at 8.08 μg/mL and low cytotoxicity with the median cytotoxic concentration (CC50) higher than 100 μg/mL. B. atroviridis extract significantly reduced the parasitemia (p < 0.05) with an effective dose-50 (ED-50) of 89 mg/kg. B. atroviridis also prevented anemia, leukocytosis and liver and kidneys impairment by decrease of transaminases, ALP, creatinine, uric acid, and triglycerides concentrations. As well, B. atroviridis extract decreased some pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) levels and significantly improved the anti-inflammatory status (P < 0.01) of infected animals marked by a decrease of IL-10 concentration. These results were further confirmed by the improved of antioxidant status and the quasi-normal microarchitecture of the liver, kidneys and spleen in test groups. Overall, the hydroethanolic bark extract of Bridelia atroviridis demonstrated antimalarial property and justified its use in traditional medicine to manage malaria disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Silvere Ngouela
- Laboratory of Natural Substances, Faculty of Science, University of Yaoundé I, Cameroon
| | - Bruno Ndjakou Lenta
- Laboratory of Natural Substances, High Teaching Training College, University of Yaounde I, Cameroon
| | - Nobert Sewald
- Laboratory of Organic and Bioorganic Chemistry, University of Bielefeld, Germany
| | - Fabrice Boyom Fekam
- Laboratory for Phytobiochemistry and Medicinal Plants Studies, Antimicrobial and Biocontrol Agents Unit, Faculty of Science, University of Yaounde I, Cameroon
| | - Théophile Dimo
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé I, Cameroon.
| |
Collapse
|
42
|
Tetrorchidium didymostemon leaf extract reduces Plasmodium berghei induced oxidative stress and hepatic injury in Swiss albino mice. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-021-00548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
Abstract
Lactic acidosis and hyperlactatemia are common metabolic disturbances in patients with severe malaria. Lactic acidosis causes physiological adverse effects, which can aggravate the outcome of malaria. Despite its clear association with mortality in malaria patients, the etiology of lactic acidosis is not completely understood. In this review, the possible contributors to lactic acidosis and hyperlactatemia in patients with malaria are discussed. Both increased lactate production and impaired lactate clearance may play a role in the pathogenesis of lactic acidosis. The increased lactate production is caused by several factors, including the metabolism of intraerythrocytic Plasmodium parasites, aerobic glycolysis by activated immune cells, and an increase in anaerobic glycolysis in hypoxic cells and tissues as a consequence of parasite sequestration and anemia. Impaired hepatic and renal lactate clearance, caused by underlying liver and kidney disease, might further aggravate hyperlactatemia. Multiple factors thus participate in the etiology of lactic acidosis in malaria, and further investigations are required to fully understand their relative contributions and the consequences of this major metabolic disturbance.
Collapse
Affiliation(s)
- Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Belgium
| |
Collapse
|
44
|
BCG Provides Short-Term Protection from Experimental Cerebral Malaria in Mice. Vaccines (Basel) 2020; 8:vaccines8040745. [PMID: 33316929 PMCID: PMC7768457 DOI: 10.3390/vaccines8040745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 11/17/2022] Open
Abstract
Clinical and experimental evidence suggests that the tuberculosis vaccine BCG offers protection against unrelated pathogens including the malaria parasite. Cerebral malaria (CM) is the most severe complication associated with Plasmodium falciparum infection in humans and is responsible for most of the fatalities attributed to malaria. We investigated whether BCG protected C57BL/6 mice from P. berghei ANKA (PbA)-induced experimental CM (ECM). The majority of PbA-infected mice that were immunized with BCG showed prolonged survival without developing clinical symptoms of ECM. However, this protective effect waned over time and was associated with the recovery of viable BCG from liver and spleen. Intriguingly, BCG-mediated protection from ECM was not associated with a reduction in parasite burden, indicating that BCG immunization did not improve anti-parasite effector mechanisms. Instead, we found a significant reduction in pro-inflammatory mediators and CD8+ T cells in brains of BCG-vaccinated mice. Together these data suggest that brain recruitment of immune cells involved in the pathogenesis of ECM decreased after BCG vaccination. Understanding the mechanisms underlying the protective effects of BCG on PbA-induced ECM can provide a rationale for developing effective adjunctive therapies to reduce the risk of death and brain damage in CM.
Collapse
|
45
|
Spontaneous Pathologic Splenic Rupture in a Patient with Plasmodium falciparum Infection, First Case Reported in Israel. Infect Dis Rep 2020; 12:121-126. [PMID: 33302479 PMCID: PMC7768459 DOI: 10.3390/idr12030022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/05/2022] Open
Abstract
Travelers exposed to malaria may develop severe disease and complications. A less well-known complication is spontaneous pathologic splenic rupture, which is still under-reported and has never been reported in Israel. In this paper, we report a 23 years old healthy young man presenting in the emergency department, two weeks after coming back from Sierra Leone, with intermittent fever, mild tachycardia and mild left upper quadrant abdominal pain. The patient was diagnosed with Plasmodium falciparum infection and developed rapidly after hospital admission spleen rupture. He was managed conservatively at first but ultimately underwent splenectomy after being hemodynamically unstable. In the recovery period, the patient developed acute respiratory distress syndrome and was reintubated. A high level of suspicion is recommended in every malaria patient presenting with left upper quadrant abdominal pain, even if minimal. Ultrasonography availability in the internal medicine department may be a critical diagnostic tool, especially in non-endemic areas.
Collapse
|
46
|
Monteiro W, Brito-Sousa JD, Elizalde-Torrent A, Bôtto-Menezes C, Melo GC, Fernandez-Becerra C, Lacerda M, Del Portillo HA. Cryptic Plasmodium chronic infections: was Maurizio Ascoli right? Malar J 2020; 19:440. [PMID: 33256745 PMCID: PMC7708240 DOI: 10.1186/s12936-020-03516-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 11/20/2020] [Indexed: 12/02/2022] Open
Abstract
Cryptic Plasmodium niches outside the liver possibly represent a major source of hypnozoite-unrelated recrudescences in malaria. Maurizio Ascoli, an Italian physician and scientist, suggested that infection was maintained as a result of the persistence of endoerythrocytic parasites in the circulatory bed of some internal organs, mainly the spleen. This would explain a proportion of the recurrences in patients, regardless of the Plasmodium species. Ascoli proposed a method that included the co-administration of adrenaline, in order to induce splenic contraction, and quinine to clear expelled forms in major vessels. Driven by controversy regarding safety and effectiveness, along with the introduction of new drugs, the Ascoli method was abandoned and mostly forgotten by the malaria research community. To date, however, the existence of cryptic parasites outside the liver is gaining supportive data. This work is a historical retrospective of cryptic malaria infections and the Ascoli method, highlighting key knowledge gaps regarding these possible parasite reservoirs.
Collapse
Affiliation(s)
- Wuelton Monteiro
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil. .,Universidade Do Estado Do Amazonas (UEA), Manaus, Amazonas, Brazil.
| | - José Diego Brito-Sousa
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil.,Universidade Do Estado Do Amazonas (UEA), Manaus, Amazonas, Brazil
| | - Aleix Elizalde-Torrent
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Institut D'Investigació Germans Trias I Pujol (IGTP), Badalona, Barcelona, Spain.,IrsiCaixa AIDS Research Institute, Badalona, Catalonia, Spain
| | - Camila Bôtto-Menezes
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil.,Universidade Do Estado Do Amazonas (UEA), Manaus, Amazonas, Brazil
| | - Gisely Cardoso Melo
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil.,Universidade Do Estado Do Amazonas (UEA), Manaus, Amazonas, Brazil
| | - Carmen Fernandez-Becerra
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Institut D'Investigació Germans Trias I Pujol (IGTP), Badalona, Barcelona, Spain
| | - Marcus Lacerda
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil.,Instituto Leônidas & Maria Deane (ILMD), Fiocruz, Manaus, Amazonas, Brazil
| | - Hernando A Del Portillo
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain. .,Institut D'Investigació Germans Trias I Pujol (IGTP), Badalona, Barcelona, Spain. .,Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
47
|
Smith NL, Nahrendorf W, Sutherland C, Mooney JP, Thompson J, Spence PJ, Cowan GJM. A Conserved TCRβ Signature Dominates a Highly Polyclonal T-Cell Expansion During the Acute Phase of a Murine Malaria Infection. Front Immunol 2020; 11:587756. [PMID: 33329568 PMCID: PMC7719809 DOI: 10.3389/fimmu.2020.587756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/27/2020] [Indexed: 01/31/2023] Open
Abstract
CD4+ αβ T-cells are key mediators of the immune response to a first Plasmodium infection, undergoing extensive activation and splenic expansion during the acute phase of an infection. However, the clonality and clonal composition of this expansion has not previously been described. Using a comparative infection model, we sequenced the splenic CD4+ T-cell receptor repertoires generated over the time-course of a Plasmodium chabaudi infection. We show through repeat replicate experiments, single-cell RNA-seq, and analyses of independent RNA-seq data, that following a first infection - within a highly polyclonal expansion - T-effector repertoires are consistently dominated by TRBV3 gene usage. Clustering by sequence similarity, we find the same dominant clonal signature is expanded across replicates in the acute phase of an infection, revealing a conserved pathogen-specific T-cell response that is consistently a hallmark of a first infection, but not expanded upon re-challenge. Determining the host or parasite factors driving this conserved response may uncover novel immune targets for malaria therapeutic purposes.
Collapse
Affiliation(s)
- Natasha L. Smith
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
48
|
Araúzo-Bravo MJ, Delic D, Gerovska D, Wunderlich F. Protective Vaccination Reshapes Hepatic Response to Blood-Stage Malaria of Genes Preferentially Expressed by NK Cells. Vaccines (Basel) 2020; 8:vaccines8040677. [PMID: 33202767 PMCID: PMC7712122 DOI: 10.3390/vaccines8040677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023] Open
Abstract
The role of natural killer (NK) cells in the liver as first-line post infectionem (p.i.) effectors against blood-stage malaria and their responsiveness to protective vaccination is poorly understood. Here, we investigate the effect of vaccination on NK cell-associated genes induced in the liver by blood-stage malaria of Plasmodium chabaudi. Female Balb/c mice were vaccinated at weeks 3 and 1 before being infected with 106P. chabaudi-parasitized erythrocytes. Genes preferentially expressed by NK cells were investigated in livers of vaccination-protected and non-protected mice on days 0, 1, 4, 8, and 11 p.i. using microarrays, qRT-PCR, and chromosome landscape analysis. Blood-stage malaria induces expression of specific genes in the liver at different phases of infection, i.e., Itga1 in expanding liver-resident NK (lrNK) cells, Itga2 in immigrating conventional NK (cNK) cells; Eomes and Tbx21 encoding transcription factors; Ncr1, Tnfsf10, Prf1, Gzma, Gzmb, Gzmc, Gzmm, and Gzmk encoding cytolytic effectors; natural killer gene complex (NKC)-localized genes encoding the NK cell receptors KLRG1, KLRK1, KLRAs1, 2, 5, 7, KLRD1, KLRC1, KLRC3, as well as the three receptors KLRB1A, KLRB1C, KLRB1F and their potential ligands CLEC2D and CLEC2I. Vaccination enhances this malaria-induced expression of genes, but impairs Gzmm expression, accelerates decline of Tnfsf10 and Clec2d expression, whereas it accelerates increased expression of Clec2i, taking a very similar time course as that of genes encoding plasma membrane proteins of erythroblasts, whose malaria-induced extramedullary generation in the liver is known to be accelerated by vaccination. Collectively, vaccination reshapes the response of the liver NK cell compartment to blood-stage malaria. Particularly, the malaria-induced expansion of lrNK cells peaking on day 4 p.i. is highly significantly (p < 0.0001) reduced by enhanced immigration of peripheral cNK cells, and KLRB1F:CLEC2I interactions between NK cells and erythroid cells facilitate extramedullary erythroblastosis in the liver, thus critically contributing to vaccination-induced survival of otherwise lethal blood-stage malaria of P. chabaudi.
Collapse
Affiliation(s)
- Marcos J. Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- TransBioNet Thematic Network of Excellence for Transitional Bioinformatics, Barcelona Supercomputing Center, 08034 Barcelona, Spain
- Correspondence: (M.J.A.-B.); (D.D.); Tel.: +34-943006108 (M.J.A.-B.); +49-735154143839 (D.D.)
| | - Denis Delic
- Boeringer Ingelheim Pharma, 88400 Biberach, Germany
- Correspondence: (M.J.A.-B.); (D.D.); Tel.: +34-943006108 (M.J.A.-B.); +49-735154143839 (D.D.)
| | - Daniela Gerovska
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain;
| | - Frank Wunderlich
- Department of Biology, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| |
Collapse
|
49
|
Murshed M, Dkhil MA, Al-Shaebi EM, Qasem MAA, Mares MM, Aljawdah HMA, Alojayri G, Abdel-Gaber R, Al-Quraishy S. Biosynthesized silver nanoparticles regulate the iron status in the spleen of Plasmodium chabaudi-infected mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:40054-40060. [PMID: 32651798 DOI: 10.1007/s11356-020-10027-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/06/2020] [Indexed: 06/11/2023]
Abstract
Malaria is a dangerous disease affecting millions around the globe. Biosynthesized nanoparticles are used against a variety of diseases including malaria worldwide. Here, silver nanoparticles (AgNPs) synthesized from the leaf extracts of Indigofera oblongifolia have been used in the treatment of mice infected with Plasmodium chabaudi to evaluate the expression of iron regulatory genes in the spleen. Infrared spectroscopy was used to identify the expected classes of compounds in the extract. AgNPs were able to decrease the parasitemia nearly similar to the used reference drug, chloroquine. In addition, AgNPs significantly decreased the spleen index after infection. Moreover, the iron distribution was increased after the treatment. Finally, AgNPs could regulate the mice spleen iron regulatory genes, Lipocalin 2 (Lcn2), transferrin receptor 1 (TFR1) and hepcidin antimicrobial peptide (Hamp). Taken together, our findings indicate that AgNPs have antimalarial activity and can control the state of iron in spleen. We need further investigations to determine mechanisms of action of the AgNPs.
Collapse
Affiliation(s)
- Mutee Murshed
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia.
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, 11795, Egypt.
| | - Esam M Al-Shaebi
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Mahmood A A Qasem
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Mohammed M Mares
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Hossam M A Aljawdah
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Ghada Alojayri
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| | - Rewaida Abdel-Gaber
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, P.O. Box: 2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
50
|
Al-Quraishy S, Murshed M, Delic D, Al-Shaebi EM, Qasem MAA, Mares MM, Dkhil MA. Plasmodium chabaudi-infected mice spleen response to synthesized silver nanoparticles from Indigofera oblongifolia extract. Lett Appl Microbiol 2020; 71:542-549. [PMID: 32749003 DOI: 10.1111/lam.13366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Malaria is a worldwide serious-threatening infectious disease caused by Plasmodium and the parasite resistance to antimalarial drugs has confirmed a significant obstacle to novel therapeutic antimalarial drugs. In this article, we assessed the antioxidant and anti-inflammatory activity of nanoparticles prepared from Indigofera oblongifolia extract (AgNPs) against the infection with Plasmodium chabaudi caused in mice spleen. AgNPs could significantly suppress the parasitaemia caused by the parasite to approximately 98% on day 7 postinfection with P. chabaudi and could improve the histopathological induced spleen damage. Also, AgNPs were able to increase the capsule thickness of the infected mice spleen. In addition, the AgNPs functioned as an antioxidant agent that affects the change in glutathione, nitric oxide and catalase levels in the spleen. Moreover spleen IL1β, IL-6 and TNF-α-mRNA expression was regulated by AgNPs administration to the infected mice. These results indicated the anti-oxidant and the anti-inflammatory protective role of AgNPs against P. chabaudi-induced spleen injury.
Collapse
Affiliation(s)
- S Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M Murshed
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - D Delic
- Boehringer Ingelheim Pharma, Biberach, Germany
| | - E M Al-Shaebi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M A A Qasem
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M M Mares
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M A Dkhil
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|