1
|
Tabilin EJ, Gordon CA, Mu Y, Jiz M, Inobaya M, Avenido-Cervantes E, Gray D, Mationg ML, McManus DP, Egwang TG, Adriko M, Sako Y, Sato MO, Sato M, You H, Kelly M, Cai P. Development of a latex microsphere-based lateral flow immunoassay for the diagnosis of schistosomiasis japonica. PLoS Negl Trop Dis 2024; 18:e0012742. [PMID: 39680611 DOI: 10.1371/journal.pntd.0012742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Zoonotic schistosomiasis, caused by Schistosoma japonicum, is prevalent in China, the Philippines and Indonesia. Rapid point-of-care (POC) diagnostics are attractive and promising tools for evaluating the efficacy of intervention strategies for schistosomiasis control. METHODOLOGY The diagnostic potential of five recombinant antigens was tested by enzyme-linked immunosorbent assay (ELISA) using sera from individuals with positive Kato-Katz (KK) results for S. japonicum (n = 28) and non-endemic controls (n = 12). A latex microsphere (LM)-based lateral flow immunoassay (LFIA) incorporating the recombinant SjSAP4 (rSjSAP4) was developed for the diagnosis of schistosomiasis japonica. The test conditions including diluent, dilution factor and reaction time, were optimised for the developed LFIA. Under the optimised conditions, serum samples from individuals living in a barangay endemic for S. japonicum (n = 549) and non-endemic controls (n = 50) were tested with the established LFIA cassettes. The results were imaged by a smartphone and analysed by the ImageJ program. The intensity ratio of the test line to the control line (T/C ratio) was calculated for each cassette. MAIN FINDINGS ELISA confirmed that rSjSAP4 was the optimal candidate for serological diagnosis of schistosomiasis japonica. Under optimal testing conditions, the developed LFIA strips had a sensitivity of 80.6% and a specificity of 98.0% at a cut-off T/C ratio of 0.1031. Moreover, the results of the LM-based LFIA was positively correlated with those obtained from the rSjSAP4-ELISA (r = 0.8270, 95% CI, 0.7990-0.8514; p < 0.0001). The schistosomiasis prevalence determined by the LFIA strips was about 1.8 times greater than that obtained with the 6-slide KK procedure performed on three stool samples. CONCLUSIONS/SIGNIFICANCE The developed LFIA represents a POC diagnostic tool that is suitable for onsite screening of human S. japonicum infection with minimal equipment needed. The established immunochromatographic assay complies with most of the WHO's ASSURED criteria for POC diagnostics.
Collapse
Affiliation(s)
- Emmanuel John Tabilin
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Catherine A Gordon
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Yi Mu
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mario Jiz
- Research Institute for Tropical Medicine, Department of Health, Manila, Philippines
| | - Marianette Inobaya
- Research Institute for Tropical Medicine, Department of Health, Manila, Philippines
| | | | - Darren Gray
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mary Lorraine Mationg
- Research Institute for Tropical Medicine, Department of Health, Manila, Philippines
- Department of Applied Epidemiology, National Centre for Epidemiology and Population Health, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Donald P McManus
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Thomas G Egwang
- Department of Immunology and Parasitology, Med Biotech Laboratories, Kampala, Uganda
| | - Moses Adriko
- Vector Borne & Neglected Tropical Disease Control Division, Ministry of Health, Kampala, Uganda
| | | | - Marcello Otake Sato
- Faculty of Medical Technology, Division of Global Environment Parasitology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, Japan
| | - Megumi Sato
- Graduate School of Health Sciences, Niigata University, Niigata, Japan
| | - Hong You
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Matthew Kelly
- Department of Applied Epidemiology, National Centre for Epidemiology and Population Health, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Pengfei Cai
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
2
|
Archer J, Cunningham LJ, Juhász A, Jones S, O’Ferrall AM, Rollason S, Mainga B, Chammudzi P, Kapira DR, Lally D, Namacha G, Makaula P, LaCourse JE, Kayuni SA, Webster BL, Musaya J, Stothard JR. Molecular epidemiology and population genetics of Schistosoma mansoni infecting school-aged children situated along the southern shoreline of Lake Malawi, Malawi. PLoS Negl Trop Dis 2024; 18:e0012504. [PMID: 39374309 PMCID: PMC11458004 DOI: 10.1371/journal.pntd.0012504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/02/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND In areas of low disease endemicity, highly sensitive diagnostic tools to identify, diagnose, and monitor intestinal schistosomiasis transmission are needed to reliably measure the burden and risk of infection. Here, we used highly sensitive molecular diagnostic methods to investigate Schistosoma mansoni prevalence and transmission along the southern shoreline of Lake Malawi, five years post-disease outbreak. METHODOLOGY AND PRINCIPAL FINDINGS Faecal and urine samples were provided by school-aged children situated along the southern shoreline of Lake Malawi. Kato-Katz faecal-egg microscopy and point-of-care circulating cathodic antigen (POC-CCA) rapid diagnostic tests were then performed to diagnose infection with S. mansoni. Urine-egg microscopy was also used to diagnose infection with Schistosoma haematobium. In addition, Schistosoma miracidia were isolated from faecal material using a standard miracidium hatching technique. A two-step real-time PCR approach was then used to diagnose infection with S. mansoni using DNA isolated from faecal samples. Furthermore, isolated miracidia were genotyped to species level through PCR and Sanger sequencing. Phylogenetic analyses were then carried out to identify which previously defined S. mansoni cox1 lineage group S. mansoni miracidia were most closely related to. The measured prevalence of S. mansoni infection varied considerably depending on which diagnostic assay was used. When compared to real-time PCR, faecal-egg microscopy had a sensitivity of 9% and a specificity of 100%. When POC-CCA 'trace' results were considered positive, POC-CCA had a sensitivity of 73% and a specificity of 81% when compared to real-time PCR. However, when considered negative, POC-CCA sensitivity was reduced to 56%, whereas specificity was increased to 90%. In addition, a high degree of S. haematobium DNA was detected in DNA isolated from faecal samples and motile S. haematobium miracidia were recovered from faecal samples. Schistosoma mansoni miracidia were closely related to two independent cox1 lineage groups, suggesting multiple recent introduction and colonisation events originating from surrounding east African countries. CONCLUSIONS AND SIGNIFICANCE Intestinal schistosomiasis is now highly prevalent along the southern shoreline of Lake Malawi just five years post-disease outbreak. In addition, a high prevalence of urogenital schistosomiasis persists. The revision of ongoing schistosomiasis control programmes in this area is therefore recommended. Our study also highlights the need for reliable diagnostic assays capable of distinguishing between Schistosoma species in multispecies co-endemic areas.
Collapse
Affiliation(s)
- John Archer
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Wolfson Wellcome Biomedical Laboratories, Department of Zoology, Natural History Museum, Cromwell Road, London, United Kingdom
| | - Lucas J. Cunningham
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Alexandra Juhász
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | - Sam Jones
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Angus M. O’Ferrall
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Sarah Rollason
- School of Biosciences, University of Cardiff, Cardiff, United Kingdom
| | - Bright Mainga
- Laboratory Department, Mangochi District Hospital, Mangochi, Malawi
| | - Priscilla Chammudzi
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Department of Pathology, School of Medicine and Oral Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi
| | - Donales R. Kapira
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Department of Pathology, School of Medicine and Oral Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi
| | - David Lally
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Department of Pathology, School of Medicine and Oral Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi
| | - Gladys Namacha
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Department of Pathology, School of Medicine and Oral Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi
| | - Peter Makaula
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
| | - James E. LaCourse
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Sekeleghe A. Kayuni
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Department of Pathology, School of Medicine and Oral Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi
| | - Bonnie L. Webster
- Wolfson Wellcome Biomedical Laboratories, Department of Zoology, Natural History Museum, Cromwell Road, London, United Kingdom
| | - Janelisa Musaya
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Queen Elizabeth Central Hospital, Blantyre, Malawi
- Department of Pathology, School of Medicine and Oral Health, Kamuzu University of Health Sciences (KUHeS), Blantyre, Malawi
| | - J. Russell Stothard
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
3
|
Graeff-Teixeira C, Marcolongo-Pereira C, Kersanach BB, Geiger SM, Negrão-Correa D. Descriptive study on risk of increased morbidity of schistosomiasis and graft loss after liver transplantation. Rev Soc Bras Med Trop 2024; 57:e00201. [PMID: 39082515 PMCID: PMC11290851 DOI: 10.1590/0037-8682-0097-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/29/2024] [Indexed: 08/02/2024] Open
Abstract
Solid-organ transplantation procedures have witnessed a surge in frequency. Consequently, increased attention to associated infections and their impact on graft success is warranted. The liver is the principal target for infection by the flatworm Schistosoma mansoni. Hence, rigorous screening protocols for this parasite should be implemented for liver transplantation donors and recipients. This study investigated the risks posed by schistosomiasis-infected liver tissues for successful liver transplantation (LT), considering donors and recipients, by analyzing reported cases. Among the 43 patients undergoing LT (donors = 19; recipients = 24), 32 were infected with S. mansoni, five were infected with other Schistosoma species, and no identification was made in four patients. Reported follow-up periods ranged from 1 to 132 months, and all patients achieved successful recovery. As these helminths do not replicate in their vertebrate hosts, immunosuppressive treatment is not expected to promote increased morbidity or reactivation. Moreover, suspected or confirmed schistosomiasis infections often have a benign course, and generally, should not prevent LT. The available literature was reviewed and a provisional screening protocol has been proposed.
Collapse
Affiliation(s)
- Carlos Graeff-Teixeira
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia e Núcleo de Doenças Infecciosas, Vitória, ES, Brasil
| | - Clairton Marcolongo-Pereira
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia e Núcleo de Doenças Infecciosas, Vitória, ES, Brasil
- Centro Universitário do Espírito Santo, Faculdade de Medicina, Colatina, ES, Brasil
| | - Betina Bolina Kersanach
- Universidade Federal do Espírito Santo, Centro de Ciências da Saúde, Departamento de Patologia e Núcleo de Doenças Infecciosas, Vitória, ES, Brasil
| | - Stefan Michael Geiger
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| | - Deborah Negrão-Correa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Laboratório de Esquistossomose e Imuno-helmintologia - Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| |
Collapse
|
4
|
Okurut S, Boulware DR, Manabe YC, Tugume L, Skipper CP, Ssebambulidde K, Rhein J, Musubire AK, Akampurira A, Okafor E, Olobo JO, Janoff EN, Meya DB. Impact of Cerebrospinal Fluid Leukocyte Infiltration and Neuroimmmune Mediators on Survival with HIV-Associated Cryptococcal Meningitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.29.24308130. [PMID: 38854002 PMCID: PMC11160828 DOI: 10.1101/2024.05.29.24308130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Introduction Cryptococcal meningitis remains a prominent cause of death in persons with advanced HIV disease. CSF leukocyte infiltration predicts survival at 18 weeks; however, how CSF immune response relates to CSF leukocyte infiltration is unknown. Methods We enrolled 401 adults with HIV-associated cryptococcal meningitis in Uganda who received amphotericin and fluconazole induction therapy. We assessed the association of CSF leukocytes, chemokine, and cytokine responses with 18-week survival. Results Participants with CSF leukocytes ≥50/μL, had higher probability 68% (52/77) of 18-week survival compared with 52% (151/292) 18-week survival in those with ≤50 cells/μL (Hazard Ratio=1.63, 95% confidence intervals 1.14-2.23; p=0.008). Survival was also associated with higher expression of T helper (Th)-1, Th17 cytokines, and immune regulatory elements. CSF levels of Programmed Death-1 Ligand, CXCL10, and Interleukin (IL)-2 independently predicted survival. In multivariate analysis, CSF leukocytes were inversely associated with CSF fungal burden and positively associated with CSF protein, interferon-gamma (IFN-γ), IL-17A, tumor necrosis factor (TNF)-α, and peripheral blood CD4+ and CD8+ T cells expression. Conclusion 18-week survival after diagnosis of cryptococcal meningitis was associated with higher CSF leukocytes at baseline with greater T helper 1 (IFN-γ, IL-2 and TNF-α cytokines), T helper 17 (IL-17A cytokine) and CXCR3+ T cell (CXCL10 chemokine) responses. These results highlight the interdependent contribution of soluble and cellular immune responses in predicting survival with HIV-associated cryptococcal meningitis.
Collapse
Affiliation(s)
- Samuel Okurut
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Department of Medical Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - David R. Boulware
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Yukari C. Manabe
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Division of Infectious Diseases, Department of Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, MD, USA
| | - Lillian Tugume
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Caleb P. Skipper
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kenneth Ssebambulidde
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Joshua Rhein
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Abdu K. Musubire
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Andrew Akampurira
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Department of Medical Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Elizabeth Okafor
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Joseph O. Olobo
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Edward N. Janoff
- Mucosal and Vaccine Research Program Colorado, Department of Medicine, Division of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, CO. USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora CO, USA
| | - David B. Meya
- Research Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | | |
Collapse
|
5
|
Sanchez JC, Pierpont TM, Argueta-Zamora D, Wilson K, August A, Cerione RA. PTEN loss in glioma cell lines leads to increased extracellular vesicles biogenesis and PD-L1 cargo in a PI3K-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.26.550575. [PMID: 38464280 PMCID: PMC10925116 DOI: 10.1101/2023.07.26.550575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Phosphatase and Tensin Homologue (PTEN) is one of the most frequently lost tumor suppressors in cancer and the predominant negative regulator of the PI3K/AKT signaling axis. A growing body of evidence has highlighted the loss of PTEN with immuno-modulatory functions including the upregulation of the programmed death ligand-1 (PD-L1), an altered tumor derived secretome that drives an immunosuppressive tumor immune microenvironment (TIME), and resistance to certain immunotherapies. Given their roles in immunosuppression and tumor growth, we examined whether the loss of PTEN would impact the biogenesis, cargo, and function of extracellular vesicles (EVs) in the context of the anti-tumor associated cytokine interferon-γ (IFN-γ). Through genetic and pharmacological approaches, we show that PD-L1 expression is regulated by JAK/STAT signaling, not PI3K signaling. Instead, we observe that PTEN loss positively upregulates cell surface levels of PD-L1 and enhances the biogenesis of EVs enriched with PD-L1 in a PI3K-dependent manner. We demonstrate that because of these changes, EVs derived from glioma cells lacking PTEN have a greater ability to suppress T cell receptor (TCR) signaling. Taken together, these findings provide important new insights into how the loss of PTEN can contribute to an immunosuppressive TIME, facilitate immune evasion, and highlight a novel role for PI3K signaling in the regulation of EV biogenesis and the cargo they contain.
Collapse
Affiliation(s)
- Julio C Sanchez
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Timothy M Pierpont
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Dariana Argueta-Zamora
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Kristin Wilson
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Richard A Cerione
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
6
|
Oyeyemi OT, Ogundahunsi O, Schunk M, Fatem RG, Shollenberger LM. Neglected tropical disease (NTD) diagnostics: current development and operations to advance control. Pathog Glob Health 2024; 118:1-24. [PMID: 37872790 PMCID: PMC10769148 DOI: 10.1080/20477724.2023.2272095] [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: 10/25/2023] Open
Abstract
Neglected tropical diseases (NTDs) have become important public health threats that require multi-faceted control interventions. As late treatment and management of NTDs contribute significantly to the associated burdens, early diagnosis becomes an important component for surveillance and planning effective interventions. This review identifies common NTDs and highlights the progress in the development of diagnostics for these NTDs. Leveraging existing technologies to improve NTD diagnosis and improving current operational approaches for deployment of developed diagnostics are crucial to achieving the 2030 NTD elimination target. Point-of-care NTD (POC-NTD) diagnostic tools are recommended preferred diagnostic options in resource-constrained areas for mapping risk zones and monitoring treatment efficacy. However, few are currently available commercially. Technical training of remote health care workers on the use of POC-NTD diagnostics, and training of health workers on the psychosocial consequences of these diagnostics are critical in harnessing POC-NTD diagnostic potential. While the COVID-19 pandemic has challenged the possibility of achieving NTD elimination in 2030 due to the disruption of healthcare services and dwindling financial support for NTDs, the possible contribution of NTDs in exacerbating COVID-19 pandemic should motivate NTD health system strengthening.
Collapse
Affiliation(s)
- Oyetunde T. Oyeyemi
- Department of Biosciences and Biotechnology, University of Medical Sciences, Ondo, Nigeria
- Department of Biological Sciences, Old Dominion University, Virginia, USA
| | - Olumide Ogundahunsi
- The Central Office for Research and Development (CORD), University of Medical Sciences, Ondo, Nigeria
| | - Mirjam Schunk
- Division of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich (LMU) institution, Munich, Germany
| | - Ramzy G. Fatem
- Schistosome Biological Supply Center, Theodor Bilharz Research Institute, Giza, Egypt
| | | |
Collapse
|
7
|
Reagin KL, Lee RL, Cocciolone L, Funk KE. Antigen non-specific CD8 + T cells accelerate cognitive decline in aged mice following respiratory coronavirus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.02.573675. [PMID: 38260669 PMCID: PMC10802364 DOI: 10.1101/2024.01.02.573675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Primarily a respiratory infection, numerous patients infected with SARS-CoV-2 present with neurologic symptoms, some continuing long after viral clearance as a persistent symptomatic phase termed "long COVID". Advanced age increases the risk of severe disease, as well as incidence of long COVID. We hypothesized that perturbations in the aged immune response predispose elderly individuals to severe coronavirus infection and post-infectious sequelae. Using a murine model of respiratory coronavirus, mouse hepatitis virus strain A59 (MHV-A59), we found that aging increased clinical illness and lethality to MHV infection, with aged animals harboring increased virus in the brain during acute infection. This was coupled with an unexpected increase in activated CD8+ T cells within the brains of aged animals but reduced antigen specificity of those CD8+ T cells. Aged animals demonstrated spatial learning impairment following MHV infection, which correlated with increased neuronal cell death and reduced neuronal regeneration in aged hippocampus. Using primary cell culture, we demonstrated that activated CD8+ T cells induce neuronal death, independent of antigen-specificity. Specifically, higher levels of CD8+ T cell-derived IFN-γ correlated with neuronal death. These results support the evidence that CD8+ T cells in the brain directly contribute to cognitive dysfunction following coronavirus infection in aged individuals.
Collapse
Affiliation(s)
- Katie L. Reagin
- Department of Biological Sciences, University of North Carolina at Charlotte
| | - Rae-Ling Lee
- Department of Biological Sciences, University of North Carolina at Charlotte
| | - Loren Cocciolone
- Department of Biological Sciences, University of North Carolina at Charlotte
| | - Kristen E. Funk
- Department of Biological Sciences, University of North Carolina at Charlotte
| |
Collapse
|
8
|
Rivera J, Mu Y, Gordon CA, Jones MK, Cheng G, Cai P. Current and upcoming point-of-care diagnostics for schistosomiasis. Trends Parasitol 2024; 40:60-74. [PMID: 38000956 DOI: 10.1016/j.pt.2023.10.005] [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: 09/17/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023]
Abstract
Point-of-care (POC) diagnostics are simple and effective portable tools that can be used for fast mapping of helminthic diseases and monitoring control programs. Most POC tests (POCTs) available for schistosomiasis diagnosis are lateral flow immunoassays (LFIAs). The emergence of simple and rapid DNA isolation methods, along with isothermal nucleic acid amplification strategies - for example, loop-mediated isothermal amplification (LAMP) and recombinase polymerase amplification (RPA) - and recent clustered regularly interspaced short palindromic repeats (CRISPR)-based diagnostic methods facilitate the development of molecular-based POC diagnostics for schistosomiasis. Furthermore, smartphone-based techniques increase real-time connectivity and readout accuracy of POCTs. This review discusses the recent advances in immunological-, molecular-based POCTs and mobile phone microscopes for the diagnosis/screening of schistosomiasis.
Collapse
Affiliation(s)
- Jonas Rivera
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Yi Mu
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Catherine A Gordon
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Malcolm K Jones
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Veterinary Science, The University of Queensland, Brisbane, Australia
| | - Guofeng Cheng
- Shanghai Tenth People's Hospital, Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, China
| | - Pengfei Cai
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
9
|
Zuhair R, Eastwood M, Jones M, Cross A, Hester J, Issa F, Ginty F, Sailem H. Decoding mTOR signalling heterogeneity in the tumour microenvironment using multiplexed imaging and graph convolutional networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.30.573693. [PMID: 38234756 PMCID: PMC10793449 DOI: 10.1101/2023.12.30.573693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Evaluating the contribution of the tumour microenvironment (TME) in tumour progression has proven a complex challenge due to the intricate interactions within the TME. Multiplexed imaging is an emerging technology that allows concurrent assessment of multiple of these components simultaneously. Here we utilise a highly multiplexed dataset of 61 markers across 746 colorectal tumours to investigate how complex mTOR signalling in different tissue compartments influences patient prognosis. We found that the signalling of mTOR pathway can have heterogeneous activation patterns in tumour and immune compartments which correlate with patient prognosis. Using graph neural networks, we determined the most predictive features of mTOR activity in immune cells and identified relevant cellular subpopulations. We validated our observations using spatial transcriptomics data analysis in an independent patient cohort. Our work provides a framework for studying complex cell signalling and reveals important insights for developing mTOR-based therapies.
Collapse
|
10
|
Turner CN, Camilo Sanchez Arcila J, Huerta N, Quiguoe AR, Jensen KDC, Hoyer KK. T cell exhaustion dynamics in systemic autoimmune disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.23.573167. [PMID: 38187518 PMCID: PMC10769367 DOI: 10.1101/2023.12.23.573167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Unlike in infection and cancer, T cell exhaustion in autoimmune disease has not been clearly defined. Here we set out to understand inhibitory protein (PD-1, Tim3, CTLA4, Lag3) expression in CXCR5- and CXCR5+ CD8 and CD4 T cells in systemic lupus erythematosus. CXCR5+ CD8 and CD4 T cells express PD-1 and engage B cells in germinal center reactions, leading to autoantibody formation in autoimmunity. We hypothesized that CXCR5+ CD8 T cells develop an exhausted phenotype as SLE autoimmunity expands from initial to chronic, self-perpetuating disease due to chronic self-antigen exposure. Our results indicate that there is no exhaustion frequency differences between sexes, although disease kinetics vary by sex. CXCR5+ CD8 T cells express primarily IFNγ, known to promote autoimmune disease development, whereas CXCR5-CD8 T cells express TNFα and IFNγ as disease progresses from 2-6 months. Tim3 is the highest expressed inhibitory marker for all CD4 and CD8 T cell populations demonstrating potential for terminally exhausted populations. CTLA4 expression on CD4 T cells suggests potential tolerance induction in these cells. We identified exhaustion phenotypes within autoimmune disease that progress with increasing lupus erythematosus severity and possibly provide a feedback mechanism for immunological tolerance. Highlights CXCR5- and CXCR5+ CD8 T cells expand with rate of disease in SLE mouse model.CXCR5+ CD8 T cells are low contributors to TNFα disease progression unlike CXCR5-CD8 T cells but may increase disease mechanisms through high IFNγ production.Inhibitory markers upregulate in frequency with the highest amounts seen in Tim3+ populations. Tim3+Lag3+ expression may be an indicator of terminal differentiation for all populations.Inhibitory marker expression frequency was unrelated to sex.
Collapse
|
11
|
Hasan MF, Campbell AR, Croom-Perez TJ, Oyer JL, Dieffenthaller TA, Robles-Carrillo LD, Cash CA, Eloriaga JE, Kumar S, Andersen BW, Naeimi Kararoudi M, Tullius BP, Lee DA, Copik AJ. Knockout of the inhibitory receptor TIGIT enhances the antitumor response of ex vivo expanded NK cells and prevents fratricide with therapeutic Fc-active TIGIT antibodies. J Immunother Cancer 2023; 11:e007502. [PMID: 38081778 PMCID: PMC10729131 DOI: 10.1136/jitc-2023-007502] [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] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Inhibitory receptor T-cell Immunoreceptor with Ig and ITIM domains (TIGIT) expressed by Natural Killer (NK) and T cells regulates cancer immunity and has been touted as the next frontier in the development of cancer immunotherapeutics. Although early results of anti-TIGIT and its combinations with antiprogrammed death-ligand 1 were highly exciting, results from an interim analysis of phase III trials are disappointing. With mixed results, there is a need to understand the effects of therapeutic anti-TIGIT on the TIGIT+ immune cells to support its clinical use. Most of the TIGIT antibodies in development have an Fc-active domain, which binds to Fc receptors on effector cells. In mouse models, Fc-active anti-TIGIT induced superior immunity, while Fc receptor engagement was required for its efficacy. NK-cell depletion compromised the antitumor immunity of anti-TIGIT indicating the essential role of NK cells in the efficacy of anti-TIGIT. Since NK cells express TIGIT and Fc-receptor CD16, Fc-active anti-TIGIT may deplete NK cells via fratricide, which has not been studied. METHODS CRISPR-Cas9-based TIGIT knockout (KO) was performed in expanded NK cells. Phenotypic and transcriptomic properties of TIGIT KO and wild-type (WT) NK cells were compared with flow cytometry, CyTOF, and RNA sequencing. The effect of TIGIT KO on NK-cell cytotoxicity was determined by calcein-AM release and live cell imaging-based cytotoxicity assays. The metabolic properties of TIGIT KO and WT NK cells were compared with a Seahorse analyzer. The effect of the Fc-component of anti-TIGIT on NK-cell fratricide was determined by co-culturing WT and TIGIT KO NK cells with Fc-active and Fc-inactive anti-TIGIT. RESULTS TIGIT KO increased the cytotoxicity of NK cells against multiple cancer cell lines including spheroids. TIGIT KO NK cells upregulated mTOR complex 1 (mTORC1) signaling and had better metabolic fitness with an increased basal glycolytic rate when co-cultured with cancer cells compared with WT NK cells. Importantly, TIGIT KO prevented NK-cell fratricide when combined with Fc-active anti-TIGIT. CONCLUSIONS TIGIT KO in ex vivo expanded NK cells increased their cytotoxicity and metabolic fitness and prevented NK-cell fratricide when combined with Fc-active anti-TIGIT antibodies. These fratricide-resistant TIGIT KO NK cells have therapeutic potential alone or in combination with Fc-active anti-TIGIT antibodies to enhance their efficacy.
Collapse
Affiliation(s)
- Md Faqrul Hasan
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Amanda R Campbell
- Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Tayler J Croom-Perez
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Jeremiah L Oyer
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | | | - Liza D Robles-Carrillo
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Catherine A Cash
- Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jonathan E Eloriaga
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Sanjana Kumar
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Brendan W Andersen
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, School of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Brian P Tullius
- Pediatric Cellular Therapies, AdventHealth for Children, Orlando, Florida, USA
| | - Dean A Lee
- Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Alicja J Copik
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
12
|
Bamgbose TT, Schilke RM, Igiehon OO, Nkadi EH, Custis D, Bharrhan S, Schwarz B, Bohrnsen E, Bosio CM, Scott RS, Yurdagul A, Finck BN, Woolard MD. Lipin-1 restrains macrophage lipid synthesis to promote inflammation resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563587. [PMID: 37961352 PMCID: PMC10634750 DOI: 10.1101/2023.10.23.563587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Macrophages are critical to maintaining and restoring tissue homeostasis during inflammation. The lipid metabolic state of macrophages influences their function, but a deeper understanding of how lipid metabolism is regulated in pro-resolving macrophage responses is needed. Lipin-1 is a phosphatidic acid phosphatase with a transcriptional coregulatory activity (TC) that regulates lipid metabolism. We previously demonstrated that lipin-1 supports pro-resolving macrophage responses, and here, myeloid-associated lipin-1 is required for inflammation resolution, yet how lipin-1-regulated cellular mechanisms promote macrophage pro-resolution responses is unknown. We demonstrated that the loss of lipin-1 in macrophages led to increased free fatty acid, neutral lipid, and ceramide content and increased phosphorylation of acetyl-CoA carboxylase. The inhibition of the first step of lipid synthesis and transport of citrate from the mitochondria in macrophages reduced lipid content and restored efferocytosis and inflammation resolution in lipin-1mKO macrophages and mice. Our findings suggest macrophage-associated lipin-1 restrains lipid synthesis, promoting pro-resolving macrophage function in response to pro-resolving stimuli.
Collapse
Affiliation(s)
- Temitayo T. Bamgbose
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Robert M. Schilke
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Oluwakemi O. Igiehon
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ebubechukwu H. Nkadi
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - David Custis
- Research Core Facility, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Sushma Bharrhan
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Benjamin Schwarz
- Proteins & Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Eric Bohrnsen
- Proteins & Chemistry Section, Research and Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Disease, Hamilton, MT
| | - Rona S. Scott
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Brian N. Finck
- Division of Nutritional Sciences and Obesity Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Matthew D. Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Center for Applied Immunology and Pathological Processes (CAIPP), Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
13
|
Comelli A, Genovese C, Gobbi F, Brindicci G, Capone S, Corpolongo A, Crosato V, Mangano VD, Marrone R, Merelli M, Prato M, Santoro CR, Scarso S, Vanino E, Marchese V, Antinori S, Mastroianni C, Raglio A, Bruschi F, Minervini A, Donà D, Garazzino S, Galli L, Lo Vecchio A, Galli A, Dragoni G, Cricelli C, Colacurci N, Ferrazzi E, Pieralli A, Montresor A, Richter J, Calleri G, Bartoloni A, Zammarchi L. Schistosomiasis in non-endemic areas: Italian consensus recommendations for screening, diagnosis and management by the Italian Society of Tropical Medicine and Global Health (SIMET), endorsed by the Committee for the Study of Parasitology of the Italian Association of Clinical Microbiologists (CoSP-AMCLI), the Italian Society of Parasitology (SoIPa), the Italian Society of Gastroenterology and Digestive Endoscopy (SIGE), the Italian Society of Gynaecology and Obstetrics (SIGO), the Italian Society of Colposcopy and Cervico-Vaginal Pathology (SICPCV), the Italian Society of General Medicine and Primary Care (SIMG), the Italian Society of Infectious and Tropical Diseases (SIMIT), the Italian Society of Pediatrics (SIP), the Italian Society of Paediatric Infectious Diseases (SITIP), the Italian Society of Urology (SIU). Infection 2023; 51:1249-1271. [PMID: 37420083 PMCID: PMC10545632 DOI: 10.1007/s15010-023-02050-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/08/2023] [Indexed: 07/09/2023]
Affiliation(s)
- Agnese Comelli
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Camilla Genovese
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- II Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Federico Gobbi
- Department of Infectious-Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
- University of Brescia, Brescia, Italy
| | - Gaetano Brindicci
- AOU Consorziale Policlinico di Bari, Infectious Diseases Unit, Bari, Italy
| | - Susanna Capone
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Angela Corpolongo
- National Institute for Infectious Diseases 'Lazzaro Spallanzani' (IRCCS), Rome, Italy
| | - Verena Crosato
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Valentina Dianora Mangano
- Department of Translational Research, N.T.M.S, Università di Pisa, Pisa, Italy
- Programma Di Monitoraggio Delle Parassitosi e f.a.d, AOU Pisana, Pisa, Italy
| | - Rosalia Marrone
- National Institute for Health, Migration and Poverty, Rome, Italy
| | - Maria Merelli
- Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy
| | - Marco Prato
- Department of Infectious-Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | | | - Salvatore Scarso
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Elisa Vanino
- Unit of Infectious Diseases, Ospedale "Santa Maria delle Croci", AUSL Romagna, Ravenna, Italy
| | - Valentina Marchese
- Department of Infectious and Tropical Diseases, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Spinello Antinori
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Claudio Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Annibale Raglio
- Committee for the Study of Parasitology of the Italian Association of Clinical Microbiologists (CoSP-AMCLI), Milan, Italy
| | - Fabrizio Bruschi
- Department of Translational Research, N.T.M.S, Università di Pisa, Pisa, Italy
- Programma Di Monitoraggio Delle Parassitosi e f.a.d, AOU Pisana, Pisa, Italy
| | - Andrea Minervini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Department of Urology, University of Florence, Florence, Italy
| | - Daniele Donà
- Division of Paediatric Infectious Diseases, Department for Women's and Children's Health, University of Padua, Padua, Italy
| | - Silvia Garazzino
- Paediatric Infectious Disease Unit, Regina Margherita Children's Hospital, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Galli
- Infectious Diseases Unit, Meyer Children's Hospital, IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Andrea Lo Vecchio
- Department of Translational Medical Sciences, Paediatric Infectious Disease Unit, University of Naples Federico II, Naples, Italy
| | - Andrea Galli
- Gastroenterology Research Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Gabriele Dragoni
- Gastroenterology Research Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Claudio Cricelli
- Health Search-Istituto di Ricerca della SIMG (Italian Society of General Medicine and Primary Care), Florence, Italy
| | - Nicola Colacurci
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Enrico Ferrazzi
- Department of Woman, New-Born and Child, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Annalisa Pieralli
- Ginecologia Chirurgica Oncologica, Careggi University and Hospital, Florence, Italy
| | - Antonio Montresor
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Joachim Richter
- Institute of International Health, Charité Universitätsmedizin, Corporate Member of Freie und Humboldt Universität Berlin and Berlin Institute of Health, Berlin, Germany
| | - Guido Calleri
- Amedeo Di Savoia Hospital, ASL Città di Torino, Turin, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
14
|
Greene KS, Choi A, Chen M, Yang N, Li R, Qiu Y, Lukey MJ, Rojas KS, Shen J, Wilson KF, Katt WP, Whittaker GR, Cerione RA. Inhibiting Glutamine Metabolism Blocks Coronavirus Replication in Mammalian Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559756. [PMID: 37808692 PMCID: PMC10557708 DOI: 10.1101/2023.09.27.559756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Developing therapeutic strategies against COVID-19 has gained widespread interest given the likelihood that new viral variants will continue to emerge. Here we describe one potential therapeutic strategy which involves targeting members of the glutaminase family of mitochondrial metabolic enzymes (GLS and GLS2), which catalyze the first step in glutamine metabolism, the hydrolysis of glutamine to glutamate. We show three examples where GLS expression increases during coronavirus infection of host cells, and another in which GLS2 is upregulated. The viruses hijack the metabolic machinery responsible for glutamine metabolism to generate the building blocks for biosynthetic processes and satisfy the bioenergetic requirements demanded by the 'glutamine addiction' of virus-infected host cells. We demonstrate how genetic silencing of glutaminase enzymes reduces coronavirus infection and that newer members of two classes of small molecule allosteric inhibitors targeting these enzymes, designated as SU1, a pan-GLS/GLS2 inhibitor, and UP4, which is specific for GLS, block viral replication in mammalian epithelial cells. Overall, these findings highlight the importance of glutamine metabolism for coronavirus replication in human cells and show that glutaminase inhibitors can block coronavirus infection and thereby may represent a novel class of anti-viral drug candidates. Teaser Inhibitors targeting glutaminase enzymes block coronavirus replication and may represent a new class of anti-viral drugs.
Collapse
|
15
|
Pantoja IEM, Ding L, Leite PEC, Marques SA, Romero JC, Din DMAE, Zack DJ, Chamling X, Smirnova L. A novel approach to increase glial cell populations in brain microphysiological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557775. [PMID: 37745321 PMCID: PMC10515937 DOI: 10.1101/2023.09.14.557775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Brain microphysiological systems (bMPS), which recapitulate human brain cellular architecture and functionality more closely than traditional monolayer cultures, have become a practical, non-invasive, and increasingly relevant platform for the study of neurological function in health and disease. These models include 3D spheroids and organoids as well as organ-on-chip models. Currently, however, existing 3D brain models vary in reflecting the relative populations of the different cell types present in the human brain. Most of the models consist mainly of neurons, while glial cells represent a smaller portion of the cell populations. Here, by means of a chemically defined glial-enriched medium (GEM), we present an improved method to expand the population of astrocytes and oligodendrocytes without compromising neuronal differentiation in bMPS. An important finding is that astrocytes not only increased in number but also changed in morphology when cultured in GEM, more closely recapitulating primary culture astrocytes. We demonstrate oligodendrocyte and astrocyte enrichment in GEM bMPS using a variety of complementary methods. We found that GEM bMPS are electro-chemically active and showed different patterns of Ca +2 staining and flux. Synaptic vesicles and terminals observed by electron microscopy were also present. No significant changes in neuronal differentiation were observed by gene expression, however, GEM enhanced neurite outgrowth and cell migration, and differentially modulated neuronal maturation in two different iPSC lines. Our results have the potential to significantly improve in vivo-like functionality of bMPS for the study of neurological diseases and drug discovery, contributing to the unmet need for safe human models.
Collapse
|
16
|
Gao KM, Chiang K, Korkmaz FT, Janardhan HP, Trivedi CM, Quinton LJ, Gingras S, Fitzgerald KA, Marshak-Rothstein A. Expression of a STING Gain-of-function Mutation in Endothelial Cells Initiates Lymphocytic Infiltration of the Lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550897. [PMID: 37547024 PMCID: PMC10402179 DOI: 10.1101/2023.07.27.550897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Patients afflicted with STING gain-of-function mutations frequently present with debilitating interstitial lung disease ( ILD ) that is recapitulated in mice expressing the STING V154M mutation ( VM ). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in the initiation of ILD. To identify STING-expressing non-hematopoietic cell types relevant to ILD, we generated a conditional knock-in ( CKI ) model in which expression of the VM allele was directed to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted expression of the mutant allele resulted in the recruitment of immune cells to the lung and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of SAVI patients or patients afflicted with other ILD-related disorders. Summary Patients with STING gain-of-function (GOF) mutations develop life-threatening lung autoinflammation. In this study, Gao et al. utilize a mouse model of conditional STING GOF to demonstrate a role for endothelial STING GOF in initiating immune cell recruitment into lung tissues of SAVI mice.
Collapse
|
17
|
Grzegorek K, Kroidl I, Prazeres da Costa C, Rothe C. Spectrum of Helminth Infections in Migrants from Sub-Saharan Africa to Europe: A Literature Review. Am J Trop Med Hyg 2023; 108:1096-1104. [PMID: 37094791 PMCID: PMC10540113 DOI: 10.4269/ajtmh.22-0354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/21/2022] [Indexed: 04/26/2023] Open
Abstract
Sub-Saharan Africa (SSA) is endemic to numerous neglected tropical diseases, including many helminth diseases. With the migration of people from this part of the world to Europe, as has been happening on a large scale since 2015, these diseases are becoming more relevant to European physicians. This work aims to summarize the recent literature on this topic and to raise awareness of helminth diseases afflicting SSA migrants. The databases PubMed, Embase, and MEDLINE were screened for literature published in English and German between January 1, 2015 and December 31, 2020. In total, 74 articles were included in this review. The spectrum of helminth infections in migrants from SSA found in the literature review is broad; current research, however, is particularly focused on infections with Schistosoma spp. and Strongyloides stercoralis. Both diseases are often characterized by a long course, with few or no symptoms, with the risk of long-term organ damage. Successful and reliable screening for schistosomiasis and strongyloidiasis is strongly recommended. However, the current diagnostic methods lack sensitivity and specificity, rendering the diagnosis challenging and reliable assessment of disease prevalence difficult. Novel diagnostic methods and a greater awareness of these diseases are urgently needed.
Collapse
Affiliation(s)
- Katharina Grzegorek
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Inge Kroidl
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
- German Center for Infection Research, Munich, Germany
| | - Clarissa Prazeres da Costa
- German Center for Infection Research, Munich, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
- Center for Global Health, Technical University Munich, Munich, Germany
| | - Camilla Rothe
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| |
Collapse
|
18
|
Cherkaoui D, Mesquita SG, Huang D, Lugli EB, Webster BL, McKendry RA. CRISPR-assisted test for Schistosoma haematobium. Sci Rep 2023; 13:4990. [PMID: 36973334 PMCID: PMC10042105 DOI: 10.1038/s41598-023-31238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
Schistosomiasis is a major neglected tropical disease targeted for elimination as a public health issue by 2030, however there is an urgent need for more sensitive and specific diagnostic tests suitable to resource-limited settings. Here we developed CATSH, a CRISPR-assisted diagnostic test for Schistosoma haematobium, utilising recombinase polymerase amplification, Cas12a-targeted cleavage and portable real-time fluorescence detection. CATSH showed high analytical sensitivity, consistent detection of a single parasitic egg and specificity for urogenital Schistosoma species. Thanks to a novel CRISPR-compatible sample preparation developed using simulated urine samples containing parasitic eggs, CATSH had a sample-to-result within 2 h. The components of CATSH can be lyophilised, reducing cold chain dependence and widening access to lower and middle-income countries. This work presents a new application of CRISPR diagnostics for highly sensitive and specific detection of parasitic pathogens in remote areas and could have a significant impact on the elimination of neglected tropical diseases.
Collapse
Affiliation(s)
- Dounia Cherkaoui
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK.
- Division of Medicine, University College London, London, WC1E 6BT, UK.
| | - Silvia G Mesquita
- Wolfson Wellcome Biomedical Laboratories, Department of Science, Natural History Museum, Cromwell Road, London, SW7 5BD, UK
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
- London Centre for Neglected Tropical Disease Research (LCNTDR), London, W21 PG, UK
| | - Da Huang
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK
| | - Elena B Lugli
- Wolfson Wellcome Biomedical Laboratories, Department of Science, Natural History Museum, Cromwell Road, London, SW7 5BD, UK
- London Centre for Neglected Tropical Disease Research (LCNTDR), London, W21 PG, UK
| | - Bonnie L Webster
- Wolfson Wellcome Biomedical Laboratories, Department of Science, Natural History Museum, Cromwell Road, London, SW7 5BD, UK.
- London Centre for Neglected Tropical Disease Research (LCNTDR), London, W21 PG, UK.
| | - Rachel A McKendry
- London Centre for Nanotechnology, University College London, London, WC1H 0AH, UK.
- Division of Medicine, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
19
|
Carlson PM, Patel RB, Birstler J, Rodriquez M, Sun C, Erbe AK, Bates AM, Marsh I, Grudzinski J, Hernandez R, Pieper AA, Feils AS, Rakhmilevich AL, Weichert JP, Bednarz BP, Sondel PM, Morris ZS. Radiation to all macroscopic sites of tumor permits greater systemic antitumor response to in situ vaccination. J Immunother Cancer 2023; 11:e005463. [PMID: 36639155 PMCID: PMC9843201 DOI: 10.1136/jitc-2022-005463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The antitumor effects of external beam radiation therapy (EBRT) are mediated, in part, by an immune response. We have reported that a single fraction of 12 Gy EBRT combined with intratumoral anti-GD2 hu14.18-IL2 immunocytokine (IC) generates an effective in situ vaccine (ISV) against GD2-positive murine tumors. This ISV is effective in eradicating single tumors with sustained immune memory; however, it does not generate an adequate abscopal response against macroscopic distant tumors. Given the immune-stimulatory capacity of radiation therapy (RT), we hypothesized that delivering RT to all sites of disease would augment systemic antitumor responses to ISV. METHODS We used a syngeneic B78 murine melanoma model consisting of a 'primary' flank tumor and a contralateral smaller 'secondary' flank tumor, treated with 12 Gy EBRT and intratumoral IC immunotherapy to the primary and additional EBRT to the secondary tumor. As a means of delivering RT to all sites of disease, both known and occult, we also used a novel alkylphosphocholine analog, NM600, conjugated to 90Y as a targeted radionuclide therapy (TRT). Tumor growth, overall survival, and cause of death were measured. Flow cytometry was used to evaluate immune population changes in both tumors. RESULTS Abscopal effects of local ISV were amplified by delivering as little as 2-6 Gy of EBRT to the secondary tumor. When the primary tumor ISV regimen was delivered in mice receiving 12 Gy EBRT to the secondary tumor, we observed improved overall survival and more disease-free mice with immune memory compared with either ISV or 12 Gy EBRT alone. Similarly, TRT combined with ISV resulted in improved overall survival and a trend towards reduced tumor growth rates when compared with either treatment alone. Using flow cytometry, we identified an influx of CD8+ T cells with a less exhausted phenotype in both the ISV-targeted primary and the distant secondary tumor following the combination of secondary tumor EBRT or TRT with primary tumor ISV. CONCLUSIONS We report a novel use for low-dose RT, not as a direct antitumor modality but as an immunomodulator capable of driving and expanding antitumor immunity against metastatic tumor sites following ISV.
Collapse
Affiliation(s)
- Peter M Carlson
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ravi B Patel
- Radiation Oncology, University of Pittsburgh Medical Center Health System, Pittsburgh, Pennsylvania, USA
| | - Jen Birstler
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew Rodriquez
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Claire Sun
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amy K Erbe
- Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amber M Bates
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ian Marsh
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joseph Grudzinski
- Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Alexander A Pieper
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Arika S Feils
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alexander L Rakhmilevich
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jamey P Weichert
- Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Bryan P Bednarz
- Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Paul M Sondel
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zachary S Morris
- Human Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
20
|
Brauneck F, Fischer B, Witt M, Muschhammer J, Oelrich J, da Costa Avelar PH, Tsoka S, Bullinger L, Seubert E, Smit DJ, Bokemeyer C, Ackermann C, Wellbrock J, Haag F, Fiedler W. TIGIT blockade repolarizes AML-associated TIGIT + M2 macrophages to an M1 phenotype and increases CD47-mediated phagocytosis. J Immunother Cancer 2022; 10:jitc-2022-004794. [PMID: 36549780 PMCID: PMC9791419 DOI: 10.1136/jitc-2022-004794] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Leukemia-associated macrophages (LAMs) represent an important cell population within the tumor microenvironment, but little is known about the phenotype, function, and plasticity of these cells. The present study provides an extensive characterization of macrophages in patients with acute myeloid leukemia (AML). METHODS The phenotype and expression of coregulatory markers were assessed on bone marrow (BM)-derived LAM populations, using multiparametric flow cytometry. BM and blood aspirates were obtained from patients with newly diagnosed acute myeloid leukemia (pAML, n=59), patients in long-term remission (lrAML, n=8), patients with relapsed acute myeloid leukemia (rAML, n=7) and monocyte-derived macrophages of the blood from healthy donors (HD, n=17). LAM subpopulations were correlated with clinical parameters. Using a blocking anti-T-cell immunoreceptor with Ig and ITIM domains (TIGIT) antibody or mouse IgG2α isotype control, we investigated polarization, secretion of cytokines, and phagocytosis on LAMs and healthy monocyte-derived macrophages in vitro. RESULTS In pAML and rAML, M1 LAMs were reduced and the predominant macrophage population consisted of immunosuppressive M2 LAMs defined by expression of CD163, CD204, CD206, and CD86. M2 LAMs in active AML highly expressed inhibitory receptors such as TIGIT, T-cell immunoglobulin and mucin-domain containing-3 protein (TIM-3), and lymphocyte-activation gene 3 (LAG-3). High expression of CD163 was associated with a poor overall survival (OS). In addition, increased frequencies of TIGIT+ M2 LAMs were associated with an intermediate or adverse risk according to the European Leukemia Network criteria and the FLT3 ITD mutation. In vitro blockade of TIGIT shifted the polarization of primary LAMs or peripheral blood-derived M2 macrophages toward the M1 phenotype and increased secretion of M1-associated cytokines and chemokines. Moreover, the blockade of TIGIT augmented the anti-CD47-mediated phagocytosis of AML cell lines and primary AML cells. CONCLUSION Our findings suggest that immunosuppressive TIGIT+ M2 LAMs can be redirected into an efficient effector population that may be of direct clinical relevance in the near future.
Collapse
Affiliation(s)
- Franziska Brauneck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Brit Fischer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marius Witt
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Muschhammer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jennyfer Oelrich
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, UK
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Elisa Seubert
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel J Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christin Ackermann
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Mesquita SG, Lugli EB, Matera G, Fonseca CT, Caldeira RL, Webster B. Development of real-time and lateral flow recombinase polymerase amplification assays for rapid detection of Schistosoma mansoni. Front Microbiol 2022; 13:1043596. [PMID: 36466644 PMCID: PMC9716991 DOI: 10.3389/fmicb.2022.1043596] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
Background Accurate diagnosis followed by timely treatment is an effective strategy for the prevention of complications together with reducing schistosomiasis transmission. Recombinase Polymerase Amplification (RPA) is a simple, rapid, sensitive, and specific isothermal method with low resource needs. This research aimed at the development and optimisation of a real-time (RT) and a lateral flow (LF) RPA assay for the detection of Schistosoma mansoni. Methodology Recombinase Polymerase Amplification reactions were performed at full- (as recommended) and half-volumes (to reduce costs), with RT or LF detection systems targeting the S. mansoni mitochondrial minisatellite region. The specificity was assessed using gDNA from other Schistosoma species, helminths co-endemic with S. mansoni, human stool, and urine, and Biomphalaria snail hosts. The analytical sensitivity was evaluated using serial dilutions of gDNA, synthetic copies of the target, and single eggs. The ability of both assays to detect the S. mansoni DNA in human urine and stool samples was also tested. The long-term stability of the RT-RPA reagents was evaluated by storing the reaction components in different temperature conditions for up to 3 weeks. Results The RT- and the LF-RPA (SmMIT- and SmMIT-LF-RPA, respectively) presented similar results when used full- and half-volumes, thus the latter was followed in all experiments. The SmMIT-RPA was 100% specific to S. mansoni, able to detect a single egg, with a limit of detection (LOD) of down to 1 fg of gDNA and one synthetic copy of the target. The assay was able to detect S. mansoni DNA from stool containing 1 egg/g and in spiked urine at a concentration of 10 fg/μl. SmMIT-RPA reagents were stable for up to 3 weeks when kept at 19°C, and 2 weeks when stored at 27°C. The SmMIT-LF-RPA cross-reacted with Clinostomidae, presented the LOD of 10 fg and one synthetic copy of the target, being able to detect a single egg and 1 egg/g in a stool sample. The LOD in spiked urine samples was 10 pg/μl. Conclusion The half-volume SmMIT-RPA is a promising method to be used in the field. It is specific, sensitive, robust, and tolerant to inhibitors, with a long-term stability of the reaction components and the real-time visualisation of results.
Collapse
Affiliation(s)
- Silvia Gonçalves Mesquita
- Grupo de Pesquisa em Helmintologia e Malacologia Médica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil,Wolfson Wellcome Laboratories, Department of Science, Natural History Museum, London, United Kingdom
| | - Elena Birgitta Lugli
- Wolfson Wellcome Laboratories, Department of Science, Natural History Museum, London, United Kingdom
| | - Giovanni Matera
- Department of Health Sciences, Unit of Microbiology, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Cristina Toscano Fonseca
- Grupo de Pesquisa em Biologia e Imunologia Parasitária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Roberta Lima Caldeira
- Grupo de Pesquisa em Helmintologia e Malacologia Médica, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil,*Correspondence: Bonnie Webster, ; Roberta Lima Caldeira,
| | - Bonnie Webster
- Wolfson Wellcome Laboratories, Department of Science, Natural History Museum, London, United Kingdom,*Correspondence: Bonnie Webster, ; Roberta Lima Caldeira,
| |
Collapse
|
22
|
Yamamoto Y, Kadoya K, Terkawi MA, Endo T, Konno K, Watanabe M, Ichihara S, Hara A, Kaneko K, Iwasaki N, Ishijima M. Neutrophils delay repair process in Wallerian degeneration by releasing NETs outside the parenchyma. Life Sci Alliance 2022; 5:e202201399. [PMID: 35961782 PMCID: PMC9375156 DOI: 10.26508/lsa.202201399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/05/2022] Open
Abstract
Although inflammation is indispensable for the repair process in Wallerian degeneration (WD), the role of neutrophils in the WD repair process remains unclear. After peripheral nerve injury, neutrophils accumulate at the epineurium but not the parenchyma in the WD region because of the blood-nerve barrier. An increase or decrease in the number of neutrophils delayed or promoted macrophage infiltration from the epineurium into the parenchyma and the repair process in WD. Abundant neutrophil extracellular traps (NETs) were formed around neutrophils, and its inhibition dramatically increased macrophage infiltration into the parenchyma. Furthermore, inhibition of either MIF or its receptor, CXCR4, in neutrophils decreased NET formation, resulting in enhanced macrophage infiltration into the parenchyma. Moreover, inhibiting MIF for just 2 h after peripheral nerve injury promoted the repair process. These findings indicate that neutrophils delay the repair process in WD from outside the parenchyma by inhibiting macrophage infiltration via NET formation and that neutrophils, NETs, MIF, and CXCR4 are therapeutic targets for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yasuhiro Yamamoto
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamad Alaa Terkawi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Endo
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Ichihara
- Department of Orthopaedic Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Akira Hara
- Department of Orthopaedic Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Kazuo Kaneko
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Yang M, Mahanty A, Jin C, Wong ANN, Yoo JS. Label-free metabolic imaging for sensitive and robust monitoring of anti-CD47 immunotherapy response in triple-negative breast cancer. J Immunother Cancer 2022; 10:jitc-2022-005199. [PMID: 36096527 PMCID: PMC9472253 DOI: 10.1136/jitc-2022-005199] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2022] [Indexed: 11/22/2022] Open
Abstract
Background Immunotherapy is revolutionizing cancer treatment from conventional radiotherapies and chemotherapies to immune checkpoint inhibitors which use patients’ immune system to recognize and attack cancer cells. Despite the huge clinical success and vigorous development of immunotherapies, there is a significant unmet need for a robust tool to identify responders to specific immunotherapy. Early and accurate monitoring of immunotherapy response is indispensable for personalized treatment and effective drug development. Methods We established a label-free metabolic intravital imaging (LMII) technique to detect two-photon excited autofluorescence signals from two coenzymes, NAD(P)H (reduced nicotinamide adenine dinucleotide (phosphate) hydrogen) and FAD (flavin adenine dinucleotide) as robust imaging markers to monitor metabolic responses to immunotherapy. Murine models of triple-negative breast cancer (TNBC) were established and tested with different therapeutic regimens including anti-cluster of differentiation 47 (CD47) immunotherapy to monitor time-course treatment responses using the developed metabolic imaging technique. Results We first imaged the mechanisms of the CD47-signal regulatory protein alpha pathway in vivo, which unravels macrophage-mediated antibody-dependent cellular phagocytosis and illustrates the metabolism of TNBC cells and macrophages. We further visualized the autofluorescence of NAD(P)H and FAD and found a significant increase during tumor growth. Following anti-CD47 immunotherapy, the imaging signal was dramatically decreased demonstrating the sensitive monitoring capability of NAD(P)H and FAD imaging for therapeutic response. NAD(P)H and FAD intravital imaging also showed a marked decrease after chemotherapy and radiotherapy. A comparative study with conventional whole-body bioluminescence and fluorescent glucose imaging demonstrated superior sensitivity of metabolic imaging. Flow cytometry validated metabolic imaging results. In vivo immunofluorescent staining revealed the targeting ability of NAD(P)H imaging mainly for tumor cells and a small portion of immune-active cells and that of FAD imaging mainly for immunosuppressive cells such as M2-like tumor-associated macrophages. Conclusions Collectively, this study showcases the potential of the LMII technique as a powerful tool to visualize dynamic changes of heterogeneous cell metabolism of cancer cells and immune infiltrates in response to immunotherapy thus providing sensitive and complete monitoring. Leveraged on ability to differentiate cancer cells and immunosuppressive macrophages, the presented imaging approach provides particularly useful imaging biomarkers for emerged innate immune checkpoint inhibitors such as anti-CD47 therapy.
Collapse
Affiliation(s)
- Minfeng Yang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Arpan Mahanty
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Chunjing Jin
- The Affiliated Chuzhou Hospital of Anhui Medical University, The First People's Hospital of Chuzhou, Chuzhou, China
| | - Alex Ngai Nick Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Jung Sun Yoo
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| |
Collapse
|
24
|
Sánchez Martínez D, Tirado N, Mensurado S, Martínez-Moreno A, Romecín P, Gutiérrez Agüera F, Correia DV, Silva-Santos B, Menéndez P. Generation and proof-of-concept for allogeneic CD123 CAR-Delta One T (DOT) cells in acute myeloid leukemia. J Immunother Cancer 2022; 10:jitc-2022-005400. [PMID: 36162920 PMCID: PMC9516293 DOI: 10.1136/jitc-2022-005400] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 12/04/2022] Open
Abstract
Background Chimeric antigen receptor (CAR)-T cells have emerged as a breakthrough treatment for relapse/refractory hematological tumors, showing impressive complete remission rates. However, around 50% of the patients relapse before 1-year post-treatment. T-cell ‘fitness’ is critical to prolong CAR-T persistence and activity. Allogeneic T cells from healthy donors are less dysfunctional or exhausted than autologous patient-derived T cells; in this context, Delta One T cells (DOTs), a recently described cellular product based on MHC/HLA-independent Vδ1+γδ T cells, represent a promising allogeneic platform. Methods Here we generated and preclinically validated, for the first time, 4-1BB-based CAR-DOTs directed against the interleukin-3α chain receptor (CD123), a target antigen widely expressed on acute myeloid leukemia (AML) blasts. Results CD123CAR-DOTs showed vigorous, superior to control DOTs, cytotoxicity against AML cell lines and primary samples both in vitro and in vivo, even on tumor rechallenge. Conclusions Our results provide the proof-of-concept for a DOT-based next-generation allogeneic CAR-T therapy for AML.
Collapse
Affiliation(s)
- Diego Sánchez Martínez
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain .,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Néstor Tirado
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Sofia Mensurado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Alba Martínez-Moreno
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Paola Romecín
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Francisco Gutiérrez Agüera
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029)
| | - Daniel V Correia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Pablo Menéndez
- Josep Carreras Leukaemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain .,Red Española de Terapias Avanzadas (TERAV) - Instituto de Salud Carlos III (ISCII) (RICORS, RD21/0017/0029).,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
25
|
Summers S, Bhattacharyya T, Allan F, Stothard JR, Edielu A, Webster BL, Miles MA, Bustinduy AL. A review of the genetic determinants of praziquantel resistance in Schistosoma mansoni: Is praziquantel and intestinal schistosomiasis a perfect match? FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.933097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease (NTD) caused by parasitic trematodes belonging to the Schistosoma genus. The mainstay of schistosomiasis control is the delivery of a single dose of praziquantel (PZQ) through mass drug administration (MDA) programs. These programs have been successful in reducing the prevalence and intensity of infections. Due to the success of MDA programs, the disease has recently been targeted for elimination as a public health problem in some endemic settings. The new World Health Organization (WHO) treatment guidelines aim to provide equitable access to PZQ for individuals above two years old in targeted areas. The scale up of MDA programs may heighten the drug selection pressures on Schistosoma parasites, which could lead to the emergence of PZQ resistant schistosomes. The reliance on a single drug to treat a disease of this magnitude is worrying should drug resistance develop. Therefore, there is a need to detect and track resistant schistosomes to counteract the threat of drug resistance to the WHO 2030 NTD roadmap targets. Until recently, drug resistance studies have been hindered by the lack of molecular markers associated with PZQ resistance. This review discusses recent significant advances in understanding the molecular basis of PZQ action in S. mansoni and proposes additional genetic determinants associated with PZQ resistance. PZQ resistance will also be analyzed in the context of alternative factors that may decrease efficacy within endemic field settings, and the most recent treatment guidelines recommended by the WHO.
Collapse
|
26
|
Maggi J, Carrascal M, Soto L, Neira O, Cuéllar MC, Aravena O, James EA, Abian J, Jaraquemada D, Catalan D, Aguillón JC. Isolation of HLA-DR-naturally presented peptides identifies T-cell epitopes for rheumatoid arthritis. Ann Rheum Dis 2022; 81:1096-1105. [PMID: 35459695 DOI: 10.1136/annrheumdis-2021-220371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/08/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) immunopathogenesis revolves around the presentation of poorly characterised self-peptides by human leucocyte antigen (HLA)-class II molecules on the surface of antigen-presenting cells to autoreactive CD4 +T cells. Here, we analysed the HLA-DR-associated peptidome of synovial tissue (ST) and of dendritic cells (DCs) pulsed with synovial fluid (SF) or ST, to identify potential T-cell epitopes for RA. METHODS HLA-DR/peptide complexes were isolated from RA ST samples (n=3) and monocyte-derived DCs, generated from healthy donors carrying RA-associated shared epitope positive HLA-DR molecules and pulsed with RA SF (n=7) or ST (n=2). Peptide sequencing was performed by high-resolution mass spectrometry. The immunostimulatory capacity of selected peptides was evaluated on peripheral blood mononuclear cells from patients with RA (n=29) and healthy subjects (n=12) by flow cytometry. RESULTS We identified between 103 and 888 HLA-DR-naturally presented peptides per sample. We selected 37 native and six citrullinated (cit)-peptides for stimulation assays. Six of these peptides increased the expression of CD40L on CD4 +T cells patients with RA, and specifically triggered IFN-γ expression on RA CD4 +T cells compared with healthy subjects. Finally, the frequency of IFN-γ-producing CD4 +T cells specific for a myeloperoxidase-derived peptide showed a positive correlation with disease activity. CONCLUSIONS We significantly expanded the peptide repertoire presented by HLA-DR molecules in a physiologically relevant context, identifying six new epitopes recognised by CD4 +T cells from patients with RA. This information is important for a better understanding of the disease immunopathology, as well as for designing tolerising antigen-specific immunotherapies.
Collapse
Affiliation(s)
- Jaxaira Maggi
- Immune Regulation and Tolerance Research Group, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile Facultad de Medicina, Santiago, Chile
| | - Montserrat Carrascal
- Biological and Environmental Proteomics Group, IIBB-CSIC, IDIBAPS, Barcelona, Spain
| | - Lilian Soto
- Immune Regulation and Tolerance Research Group, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile Facultad de Medicina, Santiago, Chile
- Departamento de Medicina, Unidad del Dolor, Hospital Clinico de la Universidad de Chile Jose Joaquin Aguirre, Santiago, Chile
| | - Oscar Neira
- Servicio de Reumatología, Hospital del Salvador, Universidad de Chile, Santiago, Chile
| | - María C Cuéllar
- Servicio de Reumatología, Hospital del Salvador, Universidad de Chile, Santiago, Chile
| | - Octavio Aravena
- Immune Regulation and Tolerance Research Group, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile Facultad de Medicina, Santiago, Chile
| | - Eddie A James
- Translational Research Program, Benaroya Research Institute, Seattle, Washington, USA
| | - Joaquin Abian
- Biological and Environmental Proteomics Group, IIBB-CSIC, IDIBAPS, Barcelona, Spain
| | - Dolores Jaraquemada
- Immunology Unit, Cell Biology, Physiology and Immunology Department, Institut de Biotecnologia i Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Diego Catalan
- Immune Regulation and Tolerance Research Group, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile Facultad de Medicina, Santiago, Chile
| | - Juan C Aguillón
- Immune Regulation and Tolerance Research Group, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile Facultad de Medicina, Santiago, Chile
| |
Collapse
|
27
|
Marelli G, Morina N, Portale F, Pandini M, Iovino M, Di Conza G, Ho PC, Di Mitri D. Lipid-loaded macrophages as new therapeutic target in cancer. J Immunother Cancer 2022; 10:jitc-2022-004584. [PMID: 35798535 PMCID: PMC9263925 DOI: 10.1136/jitc-2022-004584] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/04/2022] Open
Abstract
Macrophages are main players of the innate immune system. They show great heterogeneity and play diverse functions that include support to development, sustenance of tissue homeostasis and defense against infections. Dysfunctional macrophages have been described in multiple pathologies including cancer. Indeed tumor-associated macrophages (TAMs) are abundant in most tumors and sustain cancer growth, promote invasion and mediate immune evasion. Importantly, lipid metabolism influences macrophage activation and lipid accumulation confers pathogenic features on macrophages. Notably, a subset of lipid-loaded macrophages has been recently identified in many tumor types. Lipid-loaded TAMs support tumor growth and progression and exert immune-suppressive activities. In this review, we describe the role of lipid metabolism in macrophage activation in physiology and pathology and we discuss the impact of lipid accumulation in macrophages in the context of cancer.
Collapse
Affiliation(s)
- Giulia Marelli
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Nicolò Morina
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy.,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| | - Federica Portale
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Marta Pandini
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy.,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| | - Marta Iovino
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy
| | - Giusy Di Conza
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.,Ludwig Institute of Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Diletta Di Mitri
- Tumor Microenvironment Unit, IRCCS Humanitas Research Hospital, Lombardia, Italy .,Department of Biomedical Sciences, Humanitas University, Lombardia, Italy
| |
Collapse
|
28
|
Bartlett AW, Sousa-Figueiredo JC, van Goor RC, Monaghan P, Lancaster W, Mugizi R, Mendes EP, Nery SV, Lopes S. Burden and factors associated with schistosomiasis and soil-transmitted helminth infections among school-age children in Huambo, Uige and Zaire provinces, Angola. Infect Dis Poverty 2022; 11:73. [PMID: 35752864 PMCID: PMC9233808 DOI: 10.1186/s40249-022-00975-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/17/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Schistosomiasis and soil-transmitted helminths (STHs) contribute high disease burdens amongst the neglected tropical diseases (NTDs) and are public health problems in Angola. This study reports the prevalence, intensity and risk factors for schistosomiasis and STH infection in Huambo, Uige and Zaire provinces, Angola, to inform a school-based preventive chemotherapy program. METHODS A two-stage cluster design was used to select schools and schoolchildren to participate in parasitological and water, sanitation and hygiene (WASH) surveys across Huambo, Uige, and Zaire provinces. Point-of-care circulating cathodic antigen and urinalysis rapid diagnostic tests (RDTs) were used to determine the prevalence of Schistosoma mansoni and S. haematobium, respectively. Kato-Katz was used to identify and quantify STH species and quantify and compare with RDTs for S. mansoni. Urine filtration was used to quantify and compare with RDTs for S. haematobium. Descriptive statistics were used for prevalence and infection intensity of schistosomiasis and STH infection. Performance of RDTs was assessed through specificity and Cohen's Kappa agreement with microscopy. A multivariate regression analysis was used to determine demographic and WASH factors associated with schistosomiasis and STH infection. RESULTS A total 575 schools and 17,093 schoolchildren participated in the schistosomiasis survey, of which 121 schools and 3649 schoolchildren participated in the STH survey. Overall prevalence of S. mansoni was 21.2% (municipality range 0.9-74.8%) and S. haematobium 13.6% (range 0-31.2%), with an overall prevalence of schistosomiasis of 31.4% (range 5.9-77.3%). Overall prevalence of Ascaris lumbricoides was 25.1% (range 0-89.7%), hookworm 5.2% (range 0-42.6%), and Trichuris trichiura 3.6% (range 0-24.2%), with an overall prevalence of STH infection of 29.5% (range 0.8-89.7%). Ecological zone and ethnicity were factors associated with schistosomiasis and STH infection, with older age and female sex additional risk factors for S. haematobium. CONCLUSIONS Most municipalities met World Health Organization defined prevalence thresholds for a schistosomiasis preventive chemotherapy program. A STH preventive chemotherapy program is indicated for nearly all municipalities in Uige and select municipalities in Huambo and Zaire. The association between ecological zone and ethnicity with schistosomiasis and STH infection necessitates further evaluation of home and school environmental, sociodemographic and behavioural factors to inform targeted control strategies to complement preventive chemotherapy programs.
Collapse
Affiliation(s)
- Adam W Bartlett
- Global Health Program, Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Jose C Sousa-Figueiredo
- Department of Life Sciences, Natural History Museum, Wolfson Wellcome Biomedical Laboratories, London, UK
- Health Research Center Angola, Hospital Provincial, Bengo, Angola
| | | | | | | | | | - Elsa P Mendes
- Section for Control of Neglected Tropical Diseases, Department of Disease Control, National Directorate of Public Health, Ministry of Health, Luanda, Angola
| | - Susana Vaz Nery
- Global Health Program, Kirby Institute, University of New South Wales, New South Wales, Australia
| | - Sergio Lopes
- The Mentor Initiative, Luanda, Angola.
- The Mentor Initiative, 4Th Floor (South Suite), Burns House, Harlands Road, Haywards Heath, R16 1PG, UK.
| |
Collapse
|
29
|
Ogongo P, Nyakundi RK, Chege GK, Ochola L. The Road to Elimination: Current State of Schistosomiasis Research and Progress Towards the End Game. Front Immunol 2022; 13:846108. [PMID: 35592327 PMCID: PMC9112563 DOI: 10.3389/fimmu.2022.846108] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The new WHO Roadmap for Neglected Tropical Diseases targets the global elimination of schistosomiasis as a public health problem. To date, control strategies have focused on effective diagnostics, mass drug administration, complementary and integrative public health interventions. Non-mammalian intermediate hosts and other vertebrates promote transmission of schistosomiasis and have been utilized as experimental model systems. Experimental animal models that recapitulate schistosomiasis immunology, disease progression, and pathology observed in humans are important in testing and validation of control interventions. We discuss the pivotal value of these models in contributing to elimination of schistosomiasis. Treatment of schistosomiasis relies heavily on mass drug administration of praziquantel whose efficacy is comprised due to re-infections and experimental systems have revealed the inability to kill juvenile schistosomes. In terms of diagnosis, nonhuman primate models have demonstrated the low sensitivity of the gold standard Kato Katz smear technique. Antibody assays are valuable tools for evaluating efficacy of candidate vaccines, and sera from graded infection experiments are useful for evaluating diagnostic sensitivity of different targets. Lastly, the presence of Schistosomes can compromise the efficacy of vaccines to other infectious diseases and its elimination will benefit control programs of the other diseases. As the focus moves towards schistosomiasis elimination, it will be critical to integrate treatment, diagnostics, novel research tools such as sequencing, improved understanding of disease pathogenesis and utilization of experimental models to assist with evaluating performance of new approaches.
Collapse
Affiliation(s)
- Paul Ogongo
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Ruth K. Nyakundi
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Gerald K. Chege
- Primate Unit & Delft Animal Centre, South African Medical Research Council, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Lucy Ochola
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
- Department of Environmental Health, School of Behavioural and Lifestyle Sciences, Faculty of Health Sciences, Nelson Mandela University, Gqeberha, South Africa
| |
Collapse
|
30
|
Rolig AS, Rose DC, McGee GH, Rubas W, Kivimäe S, Redmond WL. Combining bempegaldesleukin (CD122-preferential IL-2 pathway agonist) and NKTR-262 (TLR7/8 agonist) improves systemic antitumor CD8 + T cell cytotoxicity over BEMPEG+RT. J Immunother Cancer 2022; 10:jitc-2021-004218. [PMID: 35444059 PMCID: PMC9021762 DOI: 10.1136/jitc-2021-004218] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Background Tumor cell death caused by radiation therapy (RT) triggers antitumor immunity in part because dying cells release adjuvant factors that amplify and sustain dendritic cell and T cell responses. We previously demonstrated that bempegaldesleukin (BEMPEG: NKTR-214, an immunostimulatory IL-2 cytokine prodrug) significantly enhanced the antitumor efficacy of RT through a T cell-dependent mechanism. Because RT can induce either immunogenic or tolerogenic cell death, depending on various factors (radiation dose, cell cycle phase), we hypothesized that providing a specific immunogenic adjuvant, like intratumoral therapy with a novel toll-like receptor (TLR) 7/8 agonist, NKTR-262, would improve systemic tumor-specific responses through the activation of local innate immunity. Therefore, we evaluated whether intratumoral NKTR-262 combined with systemic BEMPEG treatment would elicit improved tumor-specific immunity and survival compared with RT combined with BEMPEG. Methods Tumor-bearing mice (CT26; EMT6) received BEMPEG (0.8 mg/kg; intravenously), RT (12 Gy × 1), and/or intratumoral NKTR-262 (0.5 mg/kg). Flow cytometry was used to evaluate CD4+ and CD8+ T cell responses in the blood and tumor 7 days post-treatment. The contribution of specific immune subsets was determined by depletion of CD4+, CD8+, or NK cells. CD8+ T cell cytolytic activity was determined by an in vitro CTL assay. Data are representative of 1–2 independent experiments (n=5–14/group) and statistical significance was determined by 1-way analysis of variance (ANOVA) or repeated measures ANOVA (p value cut-off of 0.05). Results BEMPEG+NKTR-262 significantly improved survival compared with BEMPEG+RT in a CD8+ T cell-dependent manner. Response to BEMPEG+NKTR-262 was characterized by a significant expansion of activated CD8+ T cells (GzmA+; Ki-67+; ICOS+; PD-1+) in the blood, which correlated with reduced tumor size (p<0.05). In the tumor, BEMPEG+NKTR-262 induced higher frequencies of GzmA+ CD8+ T cells exhibiting reduced expression of suppressive molecules (PD-1+), compared with BEMPEG+RT (p<0.05). Further, BEMPEG+NKTR-262 treatment induced greater tumor-specific CD8+ T cell cytolytic function than BEMPEG+RT. Conclusions BEMPEG+NKTR-262 therapy elicited more robust expansion of activated CD8+ T cells compared with BEMPEG+RT, suggesting that intratumoral TLR stimulation provides superior antigen presentation and costimulatory activity compared with RT. A clinical trial of BEMPEG+NKTR-262 for patients with metastatic solid tumors is in progress (NCT03435640).
Collapse
Affiliation(s)
- Annah S Rolig
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Daniel C Rose
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Grace Helen McGee
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | | | - William L Redmond
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| |
Collapse
|
31
|
Le Gall C, Cammarata A, de Haas L, Ramos-Tomillero I, Cuenca-Escalona J, Schouren K, Wijfjes Z, Becker AMD, Bödder J, Dölen Y, de Vries IJM, Figdor CG, Flórez-Grau G, Verdoes M. Efficient targeting of NY-ESO-1 tumor antigen to human cDC1s by lymphotactin results in cross-presentation and antigen-specific T cell expansion. J Immunother Cancer 2022; 10:jitc-2021-004309. [PMID: 35428705 PMCID: PMC9014073 DOI: 10.1136/jitc-2021-004309] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 12/20/2022] Open
Abstract
Background Type 1 conventional dendritic cells (cDC1s) are characterized by their ability to induce potent CD8+ T cell responses. In efforts to generate novel vaccination strategies, notably against cancer, human cDC1s emerge as an ideal target to deliver antigens. cDC1s uniquely express XCR1, a seven transmembrane G protein-coupled receptor. Due to its restricted expression and endocytic nature, XCR1 represents an attractive receptor to mediate antigen-delivery to human cDC1s. Methods To explore tumor antigen delivery to human cDC1s, we used an engineered version of XCR1-binding lymphotactin (XCL1), XCL1(CC3). Site-specific sortase-mediated transpeptidation was performed to conjugate XCL1(CC3) to an analog of the HLA-A*02:01 epitope of the cancer testis antigen New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1). While poor epitope solubility prevented isolation of stable XCL1-antigen conjugates, incorporation of a single polyethylene glycol (PEG) chain upstream of the epitope-containing peptide enabled generation of soluble XCL1(CC3)-antigen fusion constructs. Binding and chemotactic characteristics of the XCL1-antigen conjugate, as well as its ability to induce antigen-specific CD8+ T cell activation by cDC1s, was assessed. Results PEGylated XCL1(CC3)-antigen conjugates retained binding to XCR1, and induced cDC1 chemoattraction in vitro. The model epitope was efficiently cross-presented by human cDC1s to activate NY-ESO-1-specific CD8+ T cells. Importantly, vaccine activity was increased by targeting XCR1 at the surface of cDC1s. Conclusion Our results present a novel strategy for the generation of targeted vaccines fused to insoluble antigens. Moreover, our data emphasize the potential of targeting XCR1 at the surface of primary human cDC1s to induce potent CD8+ T cell responses.
Collapse
Affiliation(s)
- Camille Le Gall
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, Oncode Institute, Nijmegen, The Netherlands
| | - Anna Cammarata
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Lukas de Haas
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Iván Ramos-Tomillero
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Jorge Cuenca-Escalona
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Kayleigh Schouren
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Zacharias Wijfjes
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Anouk M D Becker
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Johanna Bödder
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Yusuf Dölen
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, Oncode Institute, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, Oncode Institute, Nijmegen, The Netherlands
| | - Georgina Flórez-Grau
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Cobb DA, de Rossi J, Liu L, An E, Lee DW. Targeting of the alpha v beta 3 integrin complex by CAR-T cells leads to rapid regression of diffuse intrinsic pontine glioma and glioblastoma. J Immunother Cancer 2022; 10:jitc-2021-003816. [PMID: 35210306 PMCID: PMC8883284 DOI: 10.1136/jitc-2021-003816] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background Diffuse intrinsic pontine glioma (DIPG) and glioblastoma (GBM) are two highly aggressive and generally incurable gliomas with little therapeutic advancements made in the past several decades. Despite immense initial success of chimeric antigen receptor (CAR) T cells for the treatment of leukemia and lymphoma, significant headway into the application of CAR-T cells against solid tumors, including gliomas, is still forthcoming. The integrin complex alphav beta3 (αvβ3) is present on multiple and diverse solid tumor types and tumor vasculature with limited expression throughout most normal tissues, qualifying it as an appealing target for CAR-T cell-mediated immunotherapy. Methods Patient-derived DIPG and GBM cell lines were evaluated by flow cytometry for surface expression of αvβ3. Second-generation CAR-T cells expressing an anti-αvβ3 single-chain variable fragment were generated by retroviral transduction containing either a CD28 or 4-1BB costimulatory domain and CD3zeta. CAR-T cells were evaluated by flow cytometry for CAR expression, memory phenotype distribution, and inhibitory receptor profile. DIPG and GBM cell lines were orthotopically implanted into NSG mice via stereotactic injection and monitored with bioluminescent imaging to evaluate αvβ3 CAR-T cell-mediated antitumor responses. Results We found that patient-derived DIPG cells and GBM cell lines express high levels of surface αvβ3 by flow cytometry, while αvβ3 is minimally expressed on normal tissues by RNA sequencing and protein microarray. The manufactured CAR-T cells consisted of a substantial frequency of favorable early memory cells and a low inhibitory receptor profile. αvβ3 CAR-T cells demonstrated efficient, antigen-specific tumor cell killing in both cytotoxicity assays and in in vivo models of orthotopically and stereotactically implanted DIPG and GBM tumors into relevant locations in the brain of NSG mice. Tumor responses were rapid and robust with systemic CAR-T cell proliferation and long-lived persistence associated with long-term survival. Following tumor clearance, TCF-1+αvβ3 CAR-T cells were detectable, underscoring their ability to persist and undergo self-renewal. Conclusions These results highlight the potential of αvβ3 CAR-T cells for immunotherapeutic treatment of aggressive brain tumors with reduced risk of on-target, off-tumor mediated toxicity due to the restricted nature of αvβ3 expression in normal tissues.
Collapse
Affiliation(s)
- Dustin A Cobb
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| | - Jacopo de Rossi
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| | - Lixia Liu
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| | - Erin An
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel W Lee
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA .,University of Virginia Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
33
|
Ghosh AJ, Saferali A, Lee S, Chase R, Moll M, Morrow J, Yun J, Castaldi PJ, Hersh CP. Blood RNA sequencing shows overlapping gene expression across COPD phenotype domains. Thorax 2022; 77:115-122. [PMID: 34168019 PMCID: PMC8711128 DOI: 10.1136/thoraxjnl-2020-216401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/21/2021] [Indexed: 02/03/2023]
Abstract
RATIONALE COPD can be assessed using multidimensional grading systems with components from three domains: pulmonary function tests, symptoms and systemic features. Clinically, measures may be used interchangeably, though it is not known if they share similar pathobiology. OBJECTIVE To use RNA sequencing (RNA-seq) to determine if there is an overlap in the underlying biological mechanisms and consequences driving different components of the multidimensional grading systems. METHODS Whole blood was collected for RNA-seq from current and former smokers in the Genetic Epidemiology of COPD study. We tested the overlap in gene expression and biological pathways associated with case-control status and quantitative COPD phenotypes within and between the three domains. RESULTS In 2647 subjects, there were 3030 genes differentially expressed in any of the three domains or case-control status. There were five genes that overlapped between the three domains and case-control status, including G protein-coupled receptor 15(GPR15), sestrin 1 (SESN1) and interferon-induced guanylate-binding protein 1 (GBP1), which were associated with longitudinal decline in FEV1. The overlap between the three domains was enriched for pathways related to cellular components. CONCLUSIONS We identified gene sets and pathways that overlap between 12 COPD-related phenotypes and case-control status. There were no pathways represented in the overlap between the three domains and case-control status, but we identified multiple genes that demonstrated a consistent pattern of expression across several of the phenotypes. Patterns of gene expression correlation were generally similar to the correlation of clinical phenotypes in the PFT and symptom domains but not the systemic features.
Collapse
Affiliation(s)
- Auyon J Ghosh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sool Lee
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Robert Chase
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Matthew Moll
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jarrett Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jeong Yun
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
34
|
de Waal AM, Hiemstra PS, Ottenhoff TH, Joosten SA, van der Does AM. Lung epithelial cells interact with immune cells and bacteria to shape the microenvironment in tuberculosis. Thorax 2022; 77:408-416. [PMID: 35017314 PMCID: PMC8938665 DOI: 10.1136/thoraxjnl-2021-217997] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022]
Abstract
The lung epithelium has long been overlooked as a key player in tuberculosis disease. In addition to acting as a direct barrier to Mycobacterium tuberculosis (Mtb), epithelial cells (EC) of the airways and alveoli act as first responders during Mtb infections; they directly sense and respond to Mtb by producing mediators such as cytokines, chemokines and antimicrobials. Interactions of EC with innate and adaptive immune cells further shape the immune response against Mtb. These three essential components, epithelium, immune cells and Mtb, are rarely studied in conjunction, owing in part to difficulties in coculturing them. Recent advances in cell culture technologies offer the opportunity to model the lung microenvironment more closely. Herein, we discuss the interplay between lung EC, immune cells and Mtb and argue that modelling these interactions is of key importance to unravel early events during Mtb infection.
Collapse
Affiliation(s)
- Amy M de Waal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom Hm Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne M van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
35
|
Miyamoto E, Takahagi A, Ohsumi A, Martinu T, Hwang D, Boonstra KM, Joe B, Umana JM, Bei KF, Vosoughi D, Liu M, Cypel M, Keshavjee S, Juvet SC. Ex vivo delivery of regulatory T cells for control of alloimmune priming in the donor lung. Eur Respir J 2021; 59:13993003.00798-2021. [PMID: 34475226 DOI: 10.1183/13993003.00798-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/17/2021] [Indexed: 11/05/2022]
Abstract
Survival after lung transplantation (LTx) is hampered by uncontrolled inflammation and alloimmunity. Regulatory T cells (Tregs) are being studied as a cellular therapy in solid organ transplantation. Whether these systemically administered Tregs can function at the appropriate location and time is an important concern. We hypothesized that in vitro expanded, recipient-derived Tregs can be delivered to donor lungs prior to LTx via ex vivo lung perfusion (EVLP), maintaining their immunomodulatory ability.In a rat model, Wistar Kyoto (WKy) CD4+CD25high Tregs were expanded in vitro prior to EVLP. Expanded Tregs were administered to Fisher 344 (F344) donor lungs during EVLP; left lungs were transplanted into WKy recipients. Treg localisation and function post-transplant were assessed. In a proof-of-concept experiment, cryopreserved expanded human CD4+CD25+CD127low Tregs were thawed and injected into discarded human lungs during EVLP.Rat Tregs entered the lung parenchyma and retained suppressive function. Expanded Tregs had no adverse effect on donor lung physiology during EVLP; lung water as measured by wet-to-dry weight ratio was reduced by Treg therapy. The administered cells remained in the graft at 3 days post-transplant where they reduced activation of intragraft effector CD4+ T cells; these effects were diminished by day 7. Human Tregs entered the lung parenchyma during EVLP where they expressed key immunoregulatory molecules (CTLA4+, 4-1BB+, CD39+, and CD15s+).Pre-transplant Treg administration can inhibit alloimmunity within the lung allograft at early time points post- transplant. Our organ-directed approach has potential for clinical translation.
Collapse
Affiliation(s)
- Ei Miyamoto
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Takahagi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Ohsumi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hwang
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Kristen M Boonstra
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Betty Joe
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Juan Mauricio Umana
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ke F Bei
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Vosoughi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Stephen C Juvet
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Cimini A, Ricci M, Gigliotti PE, Pugliese L, Chiaravalloti A, Danieli R, Schillaci O. Medical Imaging in the Diagnosis of Schistosomiasis: A Review. Pathogens 2021; 10:pathogens10081058. [PMID: 34451522 PMCID: PMC8401107 DOI: 10.3390/pathogens10081058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
Schistosomiasis is one of the most important parasitic diseases and it is endemic in tropical and subtropical areas. Clinical and laboratory data are fundamental for the diagnosis of schistosomiasis, but diagnostic imaging techniques such as x-rays, ultrasound (US), computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography/computed tomography (PET/CT) may be helpful in the evaluation of disease severity and complications. In this context, the aim of this review is to explore the actual role of diagnostic imaging in the diagnosis of schistosomiasis, underlining advantages and drawbacks providing information about the utilization of diagnostic imaging techniques in this context. Furthermore, we aim to provide a useful guide regarding imaging features of schistosomiasis for radiology and nuclear medicine physicians of non-endemic countries: in fact, in the last years non-endemic countries have experienced important flows of migrants from endemic areas, therefore it is not uncommon to face cases of this disease in daily practice.
Collapse
Affiliation(s)
- Andrea Cimini
- Department of Biomedicine and Prevention, University Tor Vergata, Via Cracovia 50, 00133 Rome, Italy; (M.R.); (P.E.G.); (L.P.); (A.C.); (O.S.)
- Correspondence: ; Tel.: +39-(06)-20902467
| | - Maria Ricci
- Department of Biomedicine and Prevention, University Tor Vergata, Via Cracovia 50, 00133 Rome, Italy; (M.R.); (P.E.G.); (L.P.); (A.C.); (O.S.)
| | - Paola Elda Gigliotti
- Department of Biomedicine and Prevention, University Tor Vergata, Via Cracovia 50, 00133 Rome, Italy; (M.R.); (P.E.G.); (L.P.); (A.C.); (O.S.)
| | - Luca Pugliese
- Department of Biomedicine and Prevention, University Tor Vergata, Via Cracovia 50, 00133 Rome, Italy; (M.R.); (P.E.G.); (L.P.); (A.C.); (O.S.)
- Department of Radiology, San Giovanni Calibita Fatebenefratelli Hospital, Via di Ponte di Quattro Capi 39, 00186 Rome, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University Tor Vergata, Via Cracovia 50, 00133 Rome, Italy; (M.R.); (P.E.G.); (L.P.); (A.C.); (O.S.)
- Nuclear Medicine Section, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Roberta Danieli
- Department of Human Sciences and Promotion of the Quality of Life, University San Raffaele, Via di Val Cannuta 247, 00166 Rome, Italy;
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Via Cracovia 50, 00133 Rome, Italy; (M.R.); (P.E.G.); (L.P.); (A.C.); (O.S.)
- Nuclear Medicine Section, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| |
Collapse
|
37
|
Hewavisenti R, Ferguson A, Wang K, Jones D, Gebhardt T, Edwards J, Zhang M, Britton W, Yang J, Hong A, Palendira U. CD103+ tumor-resident CD8+ T cell numbers underlie improved patient survival in oropharyngeal squamous cell carcinoma. J Immunother Cancer 2021; 8:jitc-2019-000452. [PMID: 32527931 PMCID: PMC7292045 DOI: 10.1136/jitc-2019-000452] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Human Papillomavirus (HPV) associated oropharyngeal squamous cell carcinoma (OPSCC) is one of the fastest growing cancers in the Western world. When compared to OPSCCs induced by smoking or alcohol, patients with HPV+ OPSCC, have better survival and the mechanisms remain unclear. METHODS The Cancer Genome Atlas (TCGA) database was examined for genes associated with tissue-resident CD8+ T cells. Multiplex immunohistochemistry (IHC) staining was performed on tumor specimen taken from 35 HPV+ and 27 HPV- OPSCC patients. RESULTS TCGA database revealed that the expression of genes encoding CD103 and CD69 were significantly higher in HPV+ head and neck SCCs (HNSCC) than in HPV- HNSCC. Higher expression levels of these two genes were also associated with better overall survival. IHC staining showed that the proportion of CD103+ tumor-resident CD8+ T cells were significantly higher in HPV+ OPSCCs when compared to HPV- OPSCC. This higher level was also associated with both lower risk of loco-regional failure, and better overall survival. Importantly, patients with HPV- OPSCC who had comparable levels of CD103+ tumor-resident CD8+ T cells to those with HPV+ OPSCC demonstrated similar survival as those with HPV+OPSCC. CONCLUSION Our results show that CD103+ tumor-resident CD8+ T cells are critical for protective immunity in both types of OPSCCs. Our data further suggest that the enhanced local protective immunity provided by tumor-resident T cell responses is the underlying factor driving favorable clinical outcomes in HPV+ OPSCCs over HPV- OPSCCs.
Collapse
Affiliation(s)
| | - Angela Ferguson
- Centenary Institute, Newtown, New South Wales, Australia.,Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Kevin Wang
- School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales, Australia
| | - Deanna Jones
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Thomas Gebhardt
- Department of Immunology and Microbiology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jarem Edwards
- Centenary Institute, Newtown, New South Wales, Australia
| | - Mei Zhang
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Radiation Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | | | - Jean Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Hong
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia .,Department of Radiation Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Umaimainthan Palendira
- Centenary Institute, Newtown, New South Wales, Australia .,Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
Çınar Ö, Brzezicha B, Grunert C, Kloetzel PM, Beier C, Peuker CA, Keller U, Pezzutto A, Busse A. High-affinity T-cell receptor specific for MyD88 L265P mutation for adoptive T-cell therapy of B-cell malignancies. J Immunother Cancer 2021; 9:jitc-2021-002410. [PMID: 34330762 PMCID: PMC8327818 DOI: 10.1136/jitc-2021-002410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Adoptive transfer of engineered T cells has shown remarkable success in B-cell malignancies. However, the most common strategy of targeting lineage-specific antigens can lead to undesirable side effects. Also, a substantial fraction of patients have refractory disease. Novel treatment approaches with more precise targeting may be an appealing alternative. Oncogenic somatic mutations represent ideal targets because of tumor specificity. Mutation-derived neoantigens can be recognized by T-cell receptors (TCRs) in the context of MHC-peptide presentation. METHODS Here we have generated T-cell lines from healthy donors by autologous in vitro priming, targeting a missense mutation on the adaptor protein MyD88, changing leucine at position 265 to proline (MyD88 L265P), which is one of the most common driver mutations found in B-cell lymphomas. RESULTS Generated T-cell lines were selectively reactive against the mutant HLA-B*07:02-restricted epitope but not against the corresponding wild-type peptide. Cloned TCRs from these cell lines led to mutation-specific and HLA-restricted reactivity with varying functional avidity. T cells engineered with a mutation-specific TCR (TCR-T cells) recognized and killed B-cell lymphoma cell lines characterized by intrinsic MyD88 L265P mutation. Furthermore, TCR-T cells showed promising therapeutic efficacy in xenograft mouse models. In addition, initial safety screening did not indicate any sign of off-target reactivity. CONCLUSION Taken together, our data suggest that mutation-specific TCRs can be used to target the MyD88 L265P mutation, and hold promise for precision therapy in a significant subgroup of B-cell malignancies, possibly achieving the goal of absolute tumor specificity, a long sought-after dream of immunotherapy.
Collapse
Affiliation(s)
- Özcan Çınar
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany .,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Corinna Grunert
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Peter Michael Kloetzel
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christin Beier
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Caroline Anna Peuker
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Antonio Pezzutto
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Antonia Busse
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
39
|
Gillardie ML, Babba O, Mahinc C, Duthel M, de Bengy C, Morineaud C, Rivollier E, Flori P. Molecular approach to the epidemiology of urinary schistosomiasis in France. PLoS Negl Trop Dis 2021; 15:e0009515. [PMID: 34228747 PMCID: PMC8284649 DOI: 10.1371/journal.pntd.0009515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 07/16/2021] [Accepted: 05/28/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The diagnosis of urogenital schistosomiasis is based on the complementarity of serological technique and microscopic examination (ME). Between 2015 and 2019, the number of urinary schistosomiasis tests received in our laboratory increased sharply from 300 to 900 per year. Therefore, we wanted to evaluate the reliability of urine microscopic examination (ME, reference and routine technique) from urine sample by comparing it to other techniques (antigenic technique and PCR). To this end, we optimized two real-time PCRs targeting respectively Schistosoma haematobium (Sh) and Schistosoma mansoni (Sm). METHODOLOGY/PRINCIPAL FINDINGS 914 urine samples from 846 patients suspected of urogenital schistosomiasis were prescribed and analyzed by PCR and also by antigenic technique for the first 143 samples. The antigenic technique evaluated was Schisto POC-CCA, Rapid Medical Diagnostics. These results (antigenic technique and PCR) were compared to ME which was performed from all urines. The percentage of 14% (128/914) positive cases with the PCR technique and the percentage of 6.0% (54/914) positive cases with ME is significantly different (Chi 2 test, p<0.001). These 128 positive PCRs correspond to 120 different patients, 88.3% (106/120) of them were young migrants and 11.7% (14/120) were French patients returning from travel. Among these migrants, more than 75% (80/106) came from French-speaking West Africa. In addition, the Schisto POC-CCA showed a specificity of 39% (46/117), too poor to be used as a screening tool in low or non-endemic areas. CONCLUSION/SIGNIFICANCE Targeted Sh and Sm PCRs in urine are reliable techniques compared to ME (reference technique). In view of our results, we decided to screen urinary schistosomiasis by direct ME always coupled by the PCR technique, which has shown better reliability criteria.
Collapse
Affiliation(s)
- Marie-Laure Gillardie
- University of Saint-Etienne, GIMAP-EA-3064, Saint Etienne, France
- Parasitology and Mycology, department of Infectious Agents and Hygiene, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Oussama Babba
- University of Saint-Etienne, GIMAP-EA-3064, Saint Etienne, France
- Parasitology and Mycology, department of Infectious Agents and Hygiene, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Caroline Mahinc
- Parasitology and Mycology, department of Infectious Agents and Hygiene, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Maureen Duthel
- University of Saint-Etienne, GIMAP-EA-3064, Saint Etienne, France
- Parasitology and Mycology, department of Infectious Agents and Hygiene, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Claire de Bengy
- University of Saint-Etienne, GIMAP-EA-3064, Saint Etienne, France
- Parasitology and Mycology, department of Infectious Agents and Hygiene, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Clotilde Morineaud
- Department of Public Health, University Hospital of Poitiers, Poitiers, France
| | - Elisabeth Rivollier
- Department PASS, University Hospital of Saint-Etienne, Saint Etienne, France
| | - Pierre Flori
- University of Saint-Etienne, GIMAP-EA-3064, Saint Etienne, France
- Parasitology and Mycology, department of Infectious Agents and Hygiene, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
40
|
Cavalcanti MG, Engel DC, de Araujo Cunha AF, Peralta JM. Case Report: Diagnosis and Assessment of Cure Approaches for Acute Schistosomiasis in Pre-School Children. Front Immunol 2021; 12:624736. [PMID: 34054799 PMCID: PMC8149760 DOI: 10.3389/fimmu.2021.624736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/27/2021] [Indexed: 02/02/2023] Open
Abstract
Acute schistosomiasis (AS) manifests with a broad spectrum of clinical features in pediatric populations. Diagnosis may be difficult in the absence of detectable numbers of eggs. As a result, new approaches may be required to achieve an accurate diagnosis. Optimal praziquantel (PZQ) treatment regimen for young children is debatable. Also, the post-treatment response is still poorly evaluated due to the lack of reliable markers. A group of 6 children (a toddler and 5 pre-school children) and one pre-adolescent were investigated for AS clinical manifestations and followed-up for two years after treatment. Ova detection was performed by Kato-Katz (KK) and presence of Schistosoma mansoni DNA was assessed by real-time PCR (rt-PCR) in stool samples. IgG and IgE anti-Schistosoma levels and urinary antigen were detected by ELISA and point-of-care circulating cathodic antigen (POC-CCA) testing in serum and urine, respectively. AS clinical symptoms were present in 5/7 (71.4%) of the infected children, and hypereosinophilia was detected in all of them. Ova detection and serology were positive in only 3/7 (44.9%) and 4/7 (57.1%), respectively. However, real-time PCR (rt-PCR) showed the presence of Schistosoma DNA in 6/7 (85.7%) of the cases, and urinary antigen was detected in all infected children. The long-term follow-up after treatment with three doses of PZQ (80mg/kg/dose), showed high cure rates (CR) as demonstrated by the DNA-based assay as well as reduced levels of side effects. CR based on urinary antigen detection ranged from 28.6 to 100%, being the highest CR due to double testing the 2-year post-treatment samples. The results suggest that high dose and repeated treatment with PZQ might be effective for AS in young children. Also, new laboratory markers should be considered to diagnosis and monitor the drug response.
Collapse
Affiliation(s)
- Marta G Cavalcanti
- Serviço de Doenças Infecciosas e Parasitárias, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departmento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Délia Celser Engel
- Serviço de Doenças Infecciosas e Parasitárias, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Fernandes de Araujo Cunha
- Departmento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Mauro Peralta
- Departmento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Kießler M, Plesca I, Sommer U, Wehner R, Wilczkowski F, Müller L, Tunger A, Lai X, Rentsch A, Peuker K, Zeissig S, Seifert AM, Seifert L, Weitz J, Bachmann M, Bornhäuser M, Aust D, Baretton G, Schmitz M. Tumor-infiltrating plasmacytoid dendritic cells are associated with survival in human colon cancer. J Immunother Cancer 2021; 9:jitc-2020-001813. [PMID: 33762320 PMCID: PMC7993360 DOI: 10.1136/jitc-2020-001813] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/19/2022] Open
Abstract
Background Plasmacytoid dendritic cells (pDCs) play a key role in the induction and maintenance of antitumor immunity. Conversely, they can act as tolerogenic DCs by inhibiting tumor-directed immune responses. Therefore, pDCs may profoundly influence tumor progression. To gain novel insights into the role of pDCs in colon cancer, we investigated the frequency and clinical relevance of pDCs in primary tumor tissues from patients with colon cancer with different clinicopathological characteristics. Methods Immunohistochemical stainings were performed to explore the frequency of tumor-infiltrating BDCA-2+ pDCs in patients with colon cancer. Statistical analyses were conducted to determine an association between the pDC density and clinicopathological characteristics of the patients. Furthermore, we used multiplex immunofluorescence stainings to evaluate the localization and phenotype of pDCs in stroma and tertiary lymphoid structures (TLS) of colon cancer tissues. Results An increased density of infiltrating pDCs was associated with lower Union for International Cancer Control (UICC) stages. Furthermore, a higher pDC frequency was significantly correlated with increased progression-free and overall survival of patients with colon cancer. Moreover, a lower number of coloncancer-infiltrating pDCs was significantly and independently linked to worse prognosis. In addition, we found that a proportion of pDCs shows a nuclear expression of the transcription factor interferon regulatory factor 7 (IRF7), which is characteristic for an activated phenotype. In various tumor stroma regions, IRF7+ pDCs were located in the neighborhood of granzyme B-expressing CD8+ T cells. Moreover, pDCs were identified as a novel component of the T cell zone of colon cancer-associated TLS, which are major regulators of adaptive antitumor immunity. A proportion of TLS-associated pDCs displayed a nuclear IRF7 expression and was preferentially located close to CD4+ T cells. Conclusions These results indicate that higher densities of tumor-infiltrating pDCs are associated with prolonged survival of patients with colon cancer. Moreover, colon cancer-infiltrating pDCs may represent a novel prognostic factor. The colocalization of activated pDCs and T cells in tumor stroma and within TLS may contribute to the correlation between higher pDC densities and better prognosis. In addition, our findings may have implications for the design of novel immunotherapeutic strategies that are based on targeting colon cancer-infiltrating pDCs.
Collapse
Affiliation(s)
- Maximilian Kießler
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ioana Plesca
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Rebekka Wehner
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Friederike Wilczkowski
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Luise Müller
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Xixi Lai
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Anke Rentsch
- University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Kenneth Peuker
- Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Sebastian Zeissig
- Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Adrian M Seifert
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Lena Seifert
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Jürgen Weitz
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Bachmann
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany.,Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Martin Bornhäuser
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Daniela Aust
- Institute of Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumor and normal tissue bank of the University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Gustavo Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumor and normal tissue bank of the University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany .,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| |
Collapse
|
42
|
Huang Q, Li CD, Yang YR, Qin XF, Wang JJ, Zhang X, Du XN, Yang X, Wang Y, Li L, Mu M, Lv Z, Cui Y, Huang K, Corrigan CJ, Wang W, Ying S. Role of the IL-33/ST2 axis in cigarette smoke-induced airways remodelling in chronic obstructive pulmonary disease. Thorax 2021; 76:thoraxjnl-2020-214712. [PMID: 33589512 DOI: 10.1136/thoraxjnl-2020-214712] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 01/17/2021] [Accepted: 01/23/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Efficient therapy and potential prophylaxis are confounded by current ignorance of the pathogenesis of airway remodelling and blockade in COPD. OBJECTIVE To explore the role of the IL-33/ST2 axis in cigarette smoke (CS) exposure-induced airways remodelling. METHODS C57BL/6, BALB/c and IL-1RL1 -/- mice exposed to CS were used to establish an animal surrogate of COPD (air-exposed=5~8, CS-exposed=6~12). Hallmarks of remodelling were measured in mice. Cigarette smoke extract (CSE)-induced proliferation and protein production in vitro by fibroblasts in the presence of anti-interleukin-33 (anti-IL-33) or hST2 antibodies were measured. Expression of IL-33 and ST2 and other remodelling hallmarks were measured, respectively, in bronchoalveolar lavage fluid (BALF) (controls=20, COPD=20), serum (controls=59, COPD=90) and lung tissue sections (controls=11, COPD=7) from patients with COPD and controls. RESULTS Wild-type mice exposed to CS elevated expression of hallmarks of tissue remodelling in the lungs and also in the heart, spleen and kidneys, which were significantly abrogated in the IL-1RL1 -/- mice. Fibroblasts exposed to CSE, compared with control, exhibited early cellular translocation of IL-33, accompanied by proliferation and elevated protein synthesis, all inhabitable by blockade of IL-33/ST2 signalling. Expression of IL-33 and ST2 and hallmarks of tissue remodelling were significantly and proportionally elevated in BALF, serum and tissue samples from patients with COPD. CONCLUSIONS Exposure to CS induces remodelling changes in multiple organs. The data support the hypothesis that CS-induced lung collagen deposition is at least partly a result of CS-induced IL-33 translocation and release from local fibroblasts.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chen Duo Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yi Ran Yang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao Feng Qin
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing Jing Wang
- Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao Nan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xia Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Wang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lun Li
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mi Mu
- Department of Respiratory Medicine, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kewu Huang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Faculty of Life Sciences & Medicine, School of Immunology & Microbial Sciences, Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Kocher K, Delot-Vilain A, Spencer D, LoTempio J, Délot EC. Paucity and Disparity of Publicly Available Sex-Disaggregated Data for the COVID-19 Epidemic Hamper Evidence-Based Decision-Making. ARCHIVES OF SEXUAL BEHAVIOR 2021; 50:407-426. [PMID: 33398705 DOI: 10.1101/2020.04.29.20083709] [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: 04/30/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 05/28/2023]
Abstract
COVID-19 has joined the long list of sexually dimorphic human disorders. Higher lethality in men, evident in the first reports from China, was confirmed in the subsequent Italian outbreak. Newspapers and scientific journals commented on this finding and the preexisting conditions, biological processes, and behavioral differences that may underlie it. However, little appeared to be released about sex differences in severity of disease, comorbidities, rate of recovery, length of hospital stay, or number of tests performed. Systematic analysis of official websites for 20 countries and 6 US states revealed a wide disparity in sex-disaggregated data made available to the public and scholars. Only a handful reported cases by sex. None of the other characteristics, including deaths, were stratified by sex at the time. Beyond suboptimal sex disaggregation, we found a paucity of usable raw data sets and a generalized lack of standardization of captured data, making comparisons difficult. A second round of data capture in April found more complete, but even more disparate, information. Our analysis revealed a wide range of sex ratios among confirmed cases. In countries where a male bias was initially reported, the proportion of women dramatically increased in 3 weeks. Analysis also revealed a complex pattern of sex ratio variation with age. Accurate, peer-reviewed, analysis of harmonized, sex-disaggregated data for characteristics of epidemics, such as availability of testing, suspected source of infection, or comorbidities, will be critical to understand where the observed disparities come from and to generate evidence-based recommendations for decision-making by governments.
Collapse
Affiliation(s)
- Kristen Kocher
- Children's National Research Institute, Center for Genetic Medicine Research, 111 Michigan Avenue NW, Washington, DC, 20010, USA
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA
| | | | - D'Andre Spencer
- Children's National Research Institute, Center for Genetic Medicine Research, 111 Michigan Avenue NW, Washington, DC, 20010, USA
| | - Jonathan LoTempio
- Children's National Research Institute, Center for Genetic Medicine Research, 111 Michigan Avenue NW, Washington, DC, 20010, USA
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA
| | - Emmanuèle C Délot
- Children's National Research Institute, Center for Genetic Medicine Research, 111 Michigan Avenue NW, Washington, DC, 20010, USA.
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, USA.
- Children's National Research Institute, Center for Translational Research, Washington, DC, USA.
| |
Collapse
|
44
|
Sen G, Gordon P, Sado DM. Cardiac manifestations of rheumatological disease: a synopsis for the cardiologist. Heart 2020; 107:1173-1181. [PMID: 33310886 DOI: 10.1136/heartjnl-2019-316460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Gautam Sen
- Cardiology, King's College Hospital NHS Foundation Trust, London, UK
| | - Patrick Gordon
- Department of Rheumatology, King's College Hospital, London, UK
| | - Daniel M Sado
- Cardiology, King's College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
45
|
Colley DG, King CH, Kittur N, Ramzy RMR, Secor WE, Fredericks-James M, Ortu G, Clements MN, Ruberanziza E, Umulisa I, Wittmann U, Campbell CH. Evaluation, Validation, and Recognition of the Point-of-Care Circulating Cathodic Antigen, Urine-Based Assay for Mapping Schistosoma mansoni Infections. Am J Trop Med Hyg 2020; 103:42-49. [PMID: 32400347 PMCID: PMC7351311 DOI: 10.4269/ajtmh.19-0788] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Efforts to control Schistosoma mansoni infection depend on the ability of programs to effectively detect and quantify infection levels and adjust programmatic approaches based on these levels and program goals. One of the three major objectives of the Schistosomiasis Consortium for Operational Research and Evaluation (SCORE) has been to develop and/or evaluate tools that would assist Neglected Tropical Disease program managers in accomplishing this fundamental task. The advent of a widely available point-of-care (POC) assay to detect schistosome circulating cathodic antigen (CCA) in urine with a rapid diagnostic test (the POC-CCA) in 2008 led SCORE and others to conduct multiple evaluations of this assay, comparing it with the Kato–Katz (KK) stool microscopy assay—the standard used for more than 45 years. This article describes multiple SCORE-funded studies comparing the POC-CCA and KK assays, the pros and cons of these assays, the use of the POC-CCA assay for mapping of S. mansoni infections in areas across the spectrum of prevalence levels, and the validation and recognition that the POC-CCA, although not infallible, is a highly useful tool to detect low-intensity infections in low-to-moderate prevalence areas. Such an assay is critical, as control programs succeed in driving down prevalence and intensity and seek to either maintain control or move to elimination of transmission of S. mansoni.
Collapse
Affiliation(s)
- Daniel G Colley
- Schistosomiasis Consortium for Operational Research and Evaluation (SCORE), Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Microbiology, University of Georgia, Athens, Georgia
| | - Charles H King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio
| | - Nupur Kittur
- Schistosomiasis Consortium for Operational Research and Evaluation (SCORE), Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Reda M R Ramzy
- National Nutrition Institute, General Organization for Teaching Hospitals and Institutes, Cairo, Egypt
| | - William Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | - Michelle N Clements
- Medical Research Council, Clinical Research Trials Unit, University College London, London, United Kingdom.,Schistosomiasis Control Initiative, London, United Kingdom
| | - Eugene Ruberanziza
- Malaria and Other Parasitic Diseases Division, Neglected Tropical Diseases and Other Parasitic Diseases Unit, Rwanda Biomedical Center, Ministry of Health, Kigali, Rwanda
| | - Irenee Umulisa
- African Leaders Malaria Alliance, Dar-es-Salam, Tanzania.,Malaria and Other Parasitic Diseases Division, Neglected Tropical Diseases and Other Parasitic Diseases Unit, Rwanda Biomedical Center, Ministry of Health, Kigali, Rwanda
| | - Udo Wittmann
- Consult AG Statistical Services, Zurich, Switzerland.,Schistosomiasis Control Initiative, London, United Kingdom
| | - Carl H Campbell
- Schistosomiasis Consortium for Operational Research and Evaluation (SCORE), Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| |
Collapse
|
46
|
van Pul KM, Vuylsteke RJCLM, de Beijer MTA, van de Ven R, van den Tol MP, Stockmann HBAC, de Gruijl TD. Breast cancer-induced immune suppression in the sentinel lymph node is effectively countered by CpG-B in conjunction with inhibition of the JAK2/STAT3 pathway. J Immunother Cancer 2020; 8:jitc-2020-000761. [PMID: 33046620 PMCID: PMC7552844 DOI: 10.1136/jitc-2020-000761] [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] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We previously showed selectively hampered activation of lymph node-resident (LNR) dendritic cell (DC) subsets in the breast cancer (BrC) sentinel lymph node (SLN) to precede a state of profound T cell anergy. Reactivating these DC subsets by intratumoral delivery of the Toll-like receptor-9 (TLR9) agonist CpG-B could potentially offer a promising immune therapeutic strategy to combat this immune suppression and prevent disease spread. Unfortunately, CpG-B can limit its own immune stimulatory activity through direct TLR9-mediated activation of signal transducer and activator of transcription 3 (STAT3), pinpointed as a key regulator of immune suppression in the tumor microenvironment. Here, we have investigated whether in vitro exposure to CpG-B, with or without simultaneous inhibition of STAT3 signaling, could overcome immune suppression in BrC SLN. METHODS Immune modulatory effects of CpG-B (CPG7909) with or without the JAK2/STAT3 inhibitor (STAT3i) AG490 were assessed in ex vivo cultured BrC SLN-derived single-cell suspensions (N=29). Multiparameter flow cytometric analyses were conducted for DC and T cell subset characterization and assessment of (intracellular) cytokine profiles. T cell reactivity against the BrC-associated antigen Mammaglobin-A was determined by means of interferon-γ ELISPOT assay. RESULTS Although CpG-B alone induced activation of all DC subsets, combined inhibition of the JAK2/STAT3 pathway resulted in superior DC maturation (ie, increased CD83 expression), with most profound activation and maturation of LNR DC subsets. Furthermore, combined CpG-B and JAK2/STAT3 inhibition promoted Th1 skewing by counterbalancing the CpG-induced Th2/regulatory T cell response and significantly enhanced Mammaglobin-A specific T cell reactivity. CONCLUSION Ex vivo immune modulation of the SLN by CpG-B and simultaneous JAK2/STAT3 inhibition can effectively overcome BrC-induced immune suppression by preferential activation of LNR DC, ultimately restoring type 1-mediated antitumor immunity, thereby securing a BrC-specific T cell response. These findings provide a clear rationale for clinical exploration of SLN-immune potentiation through local CpG/STAT3i administration in patients with BrC.
Collapse
Affiliation(s)
- Kim M van Pul
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands.,Surgical Oncology, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | | | - Monique T A de Beijer
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Medical Oncology and Otolaryngology-Head and Neck Surgery-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | | | | | - Tanja D de Gruijl
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Diab RG, Tolba MM, Ghazala RA, Abu-Sheasha GA, Webster BL, Mady RF. Intestinal schistosomiasis: Can a urine sample decide the infection? Parasitol Int 2020; 80:102201. [PMID: 33010472 DOI: 10.1016/j.parint.2020.102201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/19/2020] [Accepted: 09/01/2020] [Indexed: 12/17/2022]
Abstract
Intestinal schistosomiasis, one of the neglected tropical diseases whose control depends on accurate diagnosis of the disease prevalence. The use of low sensitive Kato Katz (KK) fecal egg detection method as a reference gold standard is not an accurate indication especially in low transmission areas. Latent class analysis frameworks especially the Bayesian could be used instead to compare between different diagnostic tests without the use of a gold standard method as a reference. Thus, this study compared two urine-based tests for the detection of circulating antigen and cell free DNA of Schistosoma mansoni versus KK method using the Bayesian latent class analytical framework and in two models where the trace results of point of contact - assay of circulating cathodic antigen (POC-CCA) were once estimated as positive, and as negative in the other model. The Bayesian framework in the trace CCA positive model showed an estimate of disease prevalence of 26% (95% BCI:0 to 60%). POC-CCA showed the highest sensitivity (74% with BCI: 9 to 91%) and lowest specificity for (20% with BCI: 0% to 37%) and the reverse for KK. For POC-CCA with traces considered negative, it was found that results between the three tests were moderated where the positivity for infection by Schistosoma antigen detection and PCR for cell free DNA approached that estimated by the Bayesian framework (44%), and the specificity for point of contact assay(81%; 95%BCI: 59% to 100%) rose in hand with its sensitivity(77%, 95% BCI:53% to 100%) and with results for PCR test (sensitivity = 80%; 95% BCI: 61% to 100%, specificity = 69%; 95% BIC: 47% to 100%). KK remains with the highest specificity while its sensitivity in the two models never exceeded 22%. Thus, we conclude that the use of a single urine sample could be very sensitive and highly specific in the diagnosis of intestinal schistosomiasis using either the trace negative model of point of contact assay, or conventional PCR, when compared to the fecal egg detection using duplicate KK. However, the use of a single tool restricts the management of the disease in areas of low endemicity.
Collapse
Affiliation(s)
- Radwa Galal Diab
- Medical Parasitology Department, Faculty of Medicine, University of Alexandria, Egypt.
| | - Mona Mohamed Tolba
- Parasitology Department, Medical Research Institute, University of Alexandria, Egypt.
| | | | - Ghada Ahmed Abu-Sheasha
- Department of Biomedical Informatics and Medical Statistics, Medical Research Institute, University of Alexandria, Egypt.
| | | | - Rasha Fadly Mady
- Medical Parasitology Department, Faculty of Medicine, University of Alexandria, Egypt.
| |
Collapse
|
48
|
Simeni Njonnou SR, Deuson J, Royer-Chardon C, Vandergheynst FA, Wilde VD. Unexplained cause of thrombocytopenia, fever, anasarca and hypothyroidism: TAFRO syndrome with thrombotic microangiopathy renal histology. BMJ Case Rep 2020; 13:13/6/e234155. [PMID: 32606113 DOI: 10.1136/bcr-2019-234155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TAFRO (thrombocytopenia, anasarca, fever, reticulin myelofibrosis or renal dysfunction and organomegaly) syndrome is a systemic inflammatory disease characterised by thrombocytopenia, anasarca, fever or inflammatory syndrome, reticulin myelofibrosis or renal dysfunction and organomegaly. It was first described as a subtype of idiopathic multicentric Castleman disease. Here, we report the case of a 42-year-old woman presenting with thrombocytopenia, anasarca, inflammatory syndrome, renal insufficiency, reticulin myelofibrosis at bone marrow biopsy and cervical and axillary lymph nodes. Kidney biopsy showed double contours of the glomerular basement membrane, mesangiolysis and endothelial swelling compatible with thrombotic microangiopathy (TMA) as well as with TAFRO syndrome. She was successfully treated by corticosteroids, tocilizumab and rituximab. This new case description of TAFRO syndrome underlines three features of this disease rarely described in the literature and never simultaneously in the same patient: the association to severe hypothyroidism, the presence of TMA-like lesions on kidney biopsy and the treatment by the association of steroids, tocilizumab and rituximab.
Collapse
Affiliation(s)
- Sylvain Raoul Simeni Njonnou
- Internal Medicine, Hopital Erasme, Brussels, Belgium
- Internal Medicine and Specialties, Faculty of Medicine and Pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | | | | | | | | |
Collapse
|
49
|
Ong E, Wong MU, Huffman A, He Y. COVID-19 coronavirus vaccine design using reverse vaccinology and machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.03.20.000141. [PMID: 32511333 PMCID: PMC7239068 DOI: 10.1101/2020.03.20.000141] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
To ultimately combat the emerging COVID-19 pandemic, it is desired to develop an effective and safe vaccine against this highly contagious disease caused by the SARS-CoV-2 coronavirus. Our literature and clinical trial survey showed that the whole virus, as well as the spike (S) protein, nucleocapsid (N) protein, and membrane (M) protein, have been tested for vaccine development against SARS and MERS. However, these vaccine candidates might lack the induction of complete protection and have safety concerns. We then applied the Vaxign reverse vaccinology tool and the newly developed Vaxign-ML machine learning tool to predict COVID-19 vaccine candidates. By investigating the entire proteome of SARS-CoV-2, six proteins, including the S protein and five non-structural proteins (nsp3, 3CL-pro, and nsp8-10), were predicted to be adhesins, which are crucial to the viral adhering and host invasion. The S, nsp3, and nsp8 proteins were also predicted by Vaxign-ML to induce high protective antigenicity. Besides the commonly used S protein, the nsp3 protein has not been tested in any coronavirus vaccine studies and was selected for further investigation. The nsp3 was found to be more conserved among SARS-CoV-2, SARS-CoV, and MERS-CoV than among 15 coronaviruses infecting human and other animals. The protein was also predicted to contain promiscuous MHC-I and MHC-II T-cell epitopes, and linear B-cell epitopes localized in specific locations and functional domains of the protein. By applying reverse vaccinology and machine learning, we predicted potential vaccine targets for effective and safe COVID-19 vaccine development. We then propose that an "Sp/Nsp cocktail vaccine" containing a structural protein(s) (Sp) and a non-structural protein(s) (Nsp) would stimulate effective complementary immune responses.
Collapse
Affiliation(s)
- Edison Ong
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mei U Wong
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anthony Huffman
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yongqun He
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
50
|
Liu Y, Zhang X, Wang M, Ma Y, Tang W. Uncovering the effect of solvents on solid-liquid phase equilibrium of praziquantel. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2019.111917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|