1
|
Gervas-Arruga J, Barba-Romero MÁ, Fernández-Martín JJ, Gómez-Cerezo JF, Segú-Vergés C, Ronzoni G, Cebolla JJ. In Silico Modeling of Fabry Disease Pathophysiology for the Identification of Early Cellular Damage Biomarker Candidates. Int J Mol Sci 2024; 25:10329. [PMID: 39408658 PMCID: PMC11477023 DOI: 10.3390/ijms251910329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal disease whose ultimate consequences are the accumulation of sphingolipids and subsequent inflammatory events, mainly at the endothelial level. The outcomes include different nervous system manifestations as well as multiple organ damage. Despite the availability of known biomarkers, early detection of FD remains a medical need. This study aimed to develop an in silico model based on machine learning to identify candidate vascular and nervous system proteins for early FD damage detection at the cellular level. A combined systems biology and machine learning approach was carried out considering molecular characteristics of FD to create a computational model of vascular and nervous system disease. A data science strategy was applied to identify risk classifiers by using 10 K-fold cross-validation. Further biological and clinical criteria were used to prioritize the most promising candidates, resulting in the identification of 36 biomarker candidates with classifier abilities, which are easily measurable in body fluids. Among them, we propose four candidates, CAMK2A, ILK, LMNA, and KHSRP, which have high classification capabilities according to our models (cross-validated accuracy ≥ 90%) and are related to the vascular and nervous systems. These biomarkers show promise as high-risk cellular and tissue damage indicators that are potentially applicable in clinical settings, although in vivo validation is still needed.
Collapse
Affiliation(s)
| | - Miguel Ángel Barba-Romero
- Department of Internal Medicine, Albacete University Hospital, 02006 Albacete, Spain;
- Albacete Medical School, Castilla-La Mancha University, 02006 Albacete, Spain
| | | | - Jorge Francisco Gómez-Cerezo
- Department of Internal Medicine, Infanta Sofía University Hospital, 28702 Madrid, Spain;
- Faculty of Medicine, European University of Madrid, 28670 Madrid, Spain
| | | | | | | |
Collapse
|
2
|
Hannon ER, Marsit CJ, Dent AE, Embury P, Ogolla S, Midem D, Williams SM, Kazura JW. Transcriptome- and DNA methylation-based cell-type deconvolutions produce similar estimates of differential gene expression and differential methylation. BioData Min 2024; 17:21. [PMID: 38992677 PMCID: PMC11241886 DOI: 10.1186/s13040-024-00374-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Changing cell-type proportions can confound studies of differential gene expression or DNA methylation (DNAm) from peripheral blood mononuclear cells (PBMCs). We examined how cell-type proportions derived from the transcriptome versus the methylome (DNAm) influence estimates of differentially expressed genes (DEGs) and differentially methylated positions (DMPs). METHODS Transcriptome and DNAm data were obtained from PBMC RNA and DNA of Kenyan children (n = 8) before, during, and 6 weeks following uncomplicated malaria. DEGs and DMPs between time points were detected using cell-type adjusted modeling with Cibersortx or IDOL, respectively. RESULTS Most major cell types and principal components had moderate to high correlation between the two deconvolution methods (r = 0.60-0.96). Estimates of cell-type proportions and DEGs or DMPs were largely unaffected by the method, with the greatest discrepancy in the estimation of neutrophils. CONCLUSION Variation in cell-type proportions is captured similarly by both transcriptomic and methylome deconvolution methods for most major cell types.
Collapse
Affiliation(s)
- Emily R Hannon
- Center for Global Health and Diseases, Case Western Reserve University, 10900 Euclid Avenue LC:4983, Cleveland, OH, 44106, USA.
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Arlene E Dent
- Center for Global Health and Diseases, Case Western Reserve University, 10900 Euclid Avenue LC:4983, Cleveland, OH, 44106, USA
- Division of Pediatric Infectious Diseases, Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA
| | - Paula Embury
- Center for Global Health and Diseases, Case Western Reserve University, 10900 Euclid Avenue LC:4983, Cleveland, OH, 44106, USA
| | | | - David Midem
- Chulaimbo Sub-county Hospital, Kisumu County, Kenya
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - James W Kazura
- Center for Global Health and Diseases, Case Western Reserve University, 10900 Euclid Avenue LC:4983, Cleveland, OH, 44106, USA
| |
Collapse
|
3
|
Friedman-Klabanoff DJ, Berry AA, Travassos MA, Shriver M, Cox C, Butts J, Lundeen JS, Strauss KA, Joshi S, Shrestha B, Mo AX, Nomicos EYH, Deye GA, Regules JA, Bergmann-Leitner ES, Pasetti MF, Laurens MB. Recombinant Full-length Plasmodium falciparum Circumsporozoite Protein-Based Vaccine Adjuvanted With Glucopyranosyl Lipid A-Liposome Quillaja saponaria 21: Results of Phase 1 Testing With Malaria Challenge. J Infect Dis 2024; 229:1883-1893. [PMID: 38330357 PMCID: PMC11175675 DOI: 10.1093/infdis/jiae062] [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: 08/26/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Malaria is preventable yet causes >600 000 deaths annually. RTS,S, the first marketed malaria vaccine, has modest efficacy, but improvements are needed for eradication. METHODS We conducted an open-label, dose escalation phase 1 study of a full-length recombinant circumsporozoite protein vaccine (rCSP) administered with adjuvant glucopyranosyl lipid A-liposome Quillaja saponaria 21 formulation (GLA-LSQ) on days 1, 29, and 85 or 1 and 490 to healthy, malaria-naive adults. The primary end points were safety and reactogenicity. The secondary end points were antibody responses and Plasmodium falciparum parasitemia after homologous controlled human malaria infection. RESULTS Participants were enrolled into 4 groups receiving rCSP/GLA-LSQ: 10 µg × 3 (n = 20), 30 µg × 3 (n = 10), 60 µg × 3 (n = 10), or 60 µg × 2 (n = 9); 10 participants received 30 µg rCSP alone × 3, and there were 6 infectivity controls. Participants experienced no serious adverse events. Rates of solicited and unsolicited adverse events were similar among groups. All 26 participants who underwent controlled human malaria infection 28 days after final vaccinations developed malaria. Increasing vaccine doses induced higher immunoglobulin G titers but did not achieve previously established RTS,S benchmarks. CONCLUSIONS rCSP/GLA-LSQ had favorable safety results. However, tested regimens did not induce protective immunity. Further investigation could assess whether adjuvant or schedule adjustments improve efficacy. CLINICAL TRIALS REGISTRATION NCT03589794.
Collapse
Affiliation(s)
- DeAnna J Friedman-Klabanoff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrea A Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mark A Travassos
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mallory Shriver
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Kathleen A Strauss
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sudhaunshu Joshi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Annie X Mo
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Effie Y H Nomicos
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gregory A Deye
- Parasitology and International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jason A Regules
- Biologics Research & Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Elke S Bergmann-Leitner
- Biologics Research & Development, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Senkpeil L, Bhardwaj J, Little MR, Holla P, Upadhye A, Fusco EM, Swanson PA, Wiegand RE, Macklin MD, Bi K, Flynn BJ, Yamamoto A, Gaskin EL, Sather DN, Oblak AL, Simpson E, Gao H, Haining WN, Yates KB, Liu X, Murshedkar T, Richie TL, Sim BKL, Otieno K, Kariuki S, Xuei X, Liu Y, Polidoro RB, Hoffman SL, Oneko M, Steinhardt LC, Schmidt NW, Seder RA, Tran TM. Innate immune activation restricts priming and protective efficacy of the radiation-attenuated PfSPZ malaria vaccine. JCI Insight 2024; 9:e167408. [PMID: 38687615 PMCID: PMC11382880 DOI: 10.1172/jci.insight.167408] [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: 12/05/2022] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.
Collapse
Affiliation(s)
- Leetah Senkpeil
- Division of Infectious Diseases, Department of Medicine
- Department of Microbiology and Immunology, and
| | | | - Morgan R Little
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Prasida Holla
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aditi Upadhye
- Division of Infectious Diseases, Department of Medicine
| | | | - Phillip A Swanson
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Ryan E Wiegand
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Kevin Bi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Barbara J Flynn
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Ayako Yamamoto
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Erik L Gaskin
- Division of Infectious Diseases, Department of Medicine
| | - D Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Edward Simpson
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hongyu Gao
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kathleen B Yates
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Xiaowen Liu
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | | - Kephas Otieno
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Xiaoling Xuei
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yunlong Liu
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rafael B Polidoro
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Laura C Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, and
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert A Seder
- Cellular Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Tuan M Tran
- Division of Infectious Diseases, Department of Medicine
- Department of Microbiology and Immunology, and
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
5
|
van Dorst MMAR, Pyuza JJ, Nkurunungi G, Kullaya VI, Smits HH, Hogendoorn PCW, Wammes LJ, Everts B, Elliott AM, Jochems SP, Yazdanbakhsh M. Immunological factors linked to geographical variation in vaccine responses. Nat Rev Immunol 2024; 24:250-263. [PMID: 37770632 DOI: 10.1038/s41577-023-00941-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Vaccination is one of medicine's greatest achievements; however, its full potential is hampered by considerable variation in efficacy across populations and geographical regions. For example, attenuated malaria vaccines in high-income countries confer almost 100% protection, whereas in low-income regions these same vaccines achieve only 20-50% protection. This trend is also observed for other vaccines, such as bacillus Calmette-Guérin (BCG), rotavirus and yellow fever vaccines, in terms of either immunogenicity or efficacy. Multiple environmental factors affect vaccine responses, including pathogen exposure, microbiota composition and dietary nutrients. However, there has been variable success with interventions that target these individual factors, highlighting the need for a better understanding of their downstream immunological mechanisms to develop new ways of modulating vaccine responses. Here, we review the immunological factors that underlie geographical variation in vaccine responses. Through the identification of causal pathways that link environmental influences to vaccine responsiveness, it might become possible to devise modulatory compounds that can complement vaccines for better outcomes in regions where they are needed most.
Collapse
Affiliation(s)
- Marloes M A R van Dorst
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Jeremia J Pyuza
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Gyaviira Nkurunungi
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Vesla I Kullaya
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Linda J Wammes
- Department of Medical Microbiology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Alison M Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Simon P Jochems
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
6
|
Blanot M, Casaroli-Marano RP, Mondéjar-Medrano J, Sallén T, Ramírez E, Segú-Vergés C, Artigas L. Aflibercept Off-Target Effects in Diabetic Macular Edema: An In Silico Modeling Approach. Int J Mol Sci 2024; 25:3621. [PMID: 38612432 PMCID: PMC11011561 DOI: 10.3390/ijms25073621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/14/2024] Open
Abstract
Intravitreal aflibercept injection (IAI) is a treatment for diabetic macular edema (DME), but its mechanism of action (MoA) has not been completely elucidated. Here, we aimed to explore IAI's MoA and its multi-target nature in DME pathophysiology with an in silico (computer simulation) disease model. We used the Therapeutic Performance Mapping System (Anaxomics Biotech property) to generate mathematical models based on the available scientific knowledge at the time of the study, describing the relationship between the modulation of vascular endothelial growth factor receptors (VEGFRs) by IAI and DME pathophysiological processes. We also undertook an enrichment analysis to explore the processes modulated by IAI, visualized the effectors' predicted protein activity, and specifically evaluated the role of VEGFR1 pathway inhibition on DME treatment. The models simulated the potential pathophysiology of DME and the likely IAI's MoA by inhibiting VEGFR1 and VEGFR2 signaling. The action of IAI through both signaling pathways modulated the identified pathophysiological processes associated with DME, with the strongest effects in angiogenesis, blood-retinal barrier alteration and permeability, and inflammation. VEGFR1 inhibition was essential to modulate inflammatory protein effectors. Given the role of VEGFR1 signaling on the modulation of inflammatory-related pathways, IAI may offer therapeutic advantages for DME through sustained VEGFR1 pathway inhibition.
Collapse
Affiliation(s)
- Morgane Blanot
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| | - Ricardo Pedro Casaroli-Marano
- Department of Surgery (FMCS), Universitat de Barcelona, 08007 Barcelona, Spain
- Hospital Clínic de Barcelona (IDIBAPS), Universitat de Barcelona, 08007 Barcelona, Spain
| | | | - Thaïs Sallén
- Bayer Hispania S.L., 08970 Sant Joan Despí, Spain; (J.M.-M.); (T.S.)
| | - Esther Ramírez
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| | - Cristina Segú-Vergés
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
- Research Programme on Biomedical Informatics (GRIB), Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Laura Artigas
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| |
Collapse
|
7
|
Nehar-Belaid D, Sokolowski M, Ravichandran S, Banchereau J, Chaussabel D, Ucar D. Baseline immune states (BIS) associated with vaccine responsiveness and factors that shape the BIS. Semin Immunol 2023; 70:101842. [PMID: 37717525 DOI: 10.1016/j.smim.2023.101842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
Vaccines are among the greatest inventions in medicine, leading to the elimination or control of numerous diseases, including smallpox, polio, measles, rubella, and, most recently, COVID-19. Yet, the effectiveness of vaccines varies among individuals. In fact, while some recipients mount a robust response to vaccination that protects them from the disease, others fail to respond. Multiple clinical and epidemiological factors contribute to this heterogeneity in responsiveness. Systems immunology studies fueled by advances in single-cell biology have been instrumental in uncovering pre-vaccination immune cell types and genomic features (i.e., the baseline immune state, BIS) that have been associated with vaccine responsiveness. Here, we review clinical factors that shape the BIS, and the characteristics of the BIS associated with responsiveness to frequently studied vaccines (i.e., influenza, COVID-19, bacterial pneumonia, malaria). Finally, we discuss potential strategies to enhance vaccine responsiveness in high-risk groups, focusing specifically on older adults.
Collapse
Affiliation(s)
| | - Mark Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | | | - Damien Chaussabel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Institute for Systems Genomics, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
8
|
Cretin J, Adjemout M, Dieppois C, Gallardo F, Torres M, Merard Z, Sawadogo SA, Picard C, Rihet P, Paul P. A Non-Coding Fc Gamma Receptor Cis-Regulatory Variant within the 1q23 Gene Cluster Is Associated with Plasmodium falciparum Infection in Children Residing in Burkina Faso. Int J Mol Sci 2023; 24:15711. [PMID: 37958695 PMCID: PMC10650193 DOI: 10.3390/ijms242115711] [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: 08/07/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 11/15/2023] Open
Abstract
Antibodies play a crucial role in activating protective immunity against malaria by interacting with Fc-gamma receptors (FcγRs). Genetic variations in genes encoding FcγRs can affect immune cell responses to the parasite. In this study, our aim was to investigate whether non-coding variants that regulate FcγR expression could influence the prevalence of Plasmodium falciparum infection. Through bioinformatics approaches, we selected expression quantitative trait loci (eQTL) for FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B genes encoding FcγRs (FCGR), in whole blood. We prioritized two regulatory variants, rs2099684 and rs1771575, located in open genomic regions. These variants were identified using RegVar, ImmuNexUT, and transcription factor annotations specific to immune cells. In addition to these, we genotyped the coding variants FCGR2A/rs1801274 and FCGR2B/rs1050501 in 234 individuals from a malaria-endemic area in Burkina Faso. We conducted age and family-based analyses to evaluate associations with the prevalence of malarial infection in both children and adults. The analysis revealed that the regulatory rs1771575-CC genotype was predicted to influence FCGR2B/FCGR2C/FCGR3A transcripts in immune cells and was the sole variant associated with a higher prevalence of malarial infection in children. In conclusion, this study identifies the rs1771575 cis-regulatory variant affecting several FcγRs in myeloid and neutrophil cells and associates it with the inter-individual capacity of children living in Burkina Faso to control malarial infection.
Collapse
Affiliation(s)
- Jules Cretin
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
- Institut MarMaRa, 13288 Marseille, France
| | - Mathieu Adjemout
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
- Institut MarMaRa, 13288 Marseille, France
| | - Christelle Dieppois
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Frederic Gallardo
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Magali Torres
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Zachary Merard
- ADES UMR, Aix Marseille University, 13288 Marseille, France (C.P.)
| | - Serge Aimé Sawadogo
- Unité de Formation en Sciences de la Santé (UFR/SDS), Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso;
- Centre PrïmO-Nelson Mandela, 84 rue Sao Tomé et Principe, Ouagadougou 09 BP 706, Burkina Faso
| | - Christophe Picard
- ADES UMR, Aix Marseille University, 13288 Marseille, France (C.P.)
- Immunogenetics Laboratory, Etablissement Français du Sang PACA-Corse, 13001 Marseille, France
| | - Pascal Rihet
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Pascale Paul
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| |
Collapse
|
9
|
Gómez J, Artigas L, Valls R, Gervas-Arruga J. An in silico approach to identify early damage biomarker candidates in metachromatic leukodystrophy. Mol Genet Metab Rep 2023; 35:100974. [PMID: 37275681 PMCID: PMC10233284 DOI: 10.1016/j.ymgmr.2023.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 06/07/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a rare, autosomal recessive lysosomal storage disease. Deficient activity of arylsulfatase A causes sulfatides to accumulate in cells of different tissues, including those in the central and peripheral nervous systems, leading to progressive demyelination and neurodegeneration. Although there is some association between specific arylsulfatase A alleles and disease severity, genotype-phenotype correlations are not fully understood. We aimed to identify biomarker candidates of early tissue damage in MLD using a modeling approach based on systems biology. A review of the literature was performed in an initial disease characterization step, allowing identification of pathophysiological processes involved in MLD and proteins relating to these processes. Three mathematical models were generated to simulate different stages of MLD at the molecular level: an early pro-inflammatory stage model (including only processes considered to be active in the early stages of disease), a pre-demyelination stage model (including additional processes that are active after some disease progression), and a demyelination stage model (in which all pathophysiological processes are active). The models evaluated 3457 proteins of interest, individually and by pairs through data mining techniques, applying five filters to prioritize biomarkers that could differentiate between the models. Sixteen potential biomarkers were identified, including effectors relating to mitochondrial dysfunction, remyelination, and neurodegeneration. The findings were corroborated in a gene expression data set from T lymphocytes of patients with MLD; all candidates formed combinations that were able to distinguish patients with MLD from controls, and all but one candidate distinguished late-infantile MLD from juvenile MLD as part of a combinatorial biomarker pair. In particular, pro-neuregulin-1 appeared as differential on all comparisons (patients with MLD vs controls and within clinical subtypes); casein kinase II subunit alpha was detected as a potential individual marker within clinical subtypes. These findings provide a panel of biomarker candidates suitable for experimental validation and highlight the utility of mathematical models to identify biomarker candidates of early tissue damage in MLD with a high degree of accuracy and sensitivity.
Collapse
|
10
|
Hirota M, Tamai M, Yukawa S, Taira N, Matthews MM, Toma T, Seto Y, Yoshida M, Toguchi S, Miyagi M, Mori T, Tomori H, Tamai O, Kina M, Sakihara E, Yamashiro C, Miyagi M, Tamaki K, Wolf M, Collins MK, Kitano H, Ishikawa H. Human immune and gut microbial parameters associated with inter-individual variations in COVID-19 mRNA vaccine-induced immunity. Commun Biol 2023; 6:368. [PMID: 37081096 PMCID: PMC10119155 DOI: 10.1038/s42003-023-04755-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
COVID-19 mRNA vaccines induce protective adaptive immunity against SARS-CoV-2 in most individuals, but there is wide variation in levels of vaccine-induced antibody and T-cell responses. However, the mechanisms underlying this inter-individual variation remain unclear. Here, using a systems biology approach based on multi-omics analyses of human blood and stool samples, we identified several factors that are associated with COVID-19 vaccine-induced adaptive immune responses. BNT162b2-induced T cell response is positively associated with late monocyte responses and inversely associated with baseline mRNA expression of activation protein 1 (AP-1) transcription factors. Interestingly, the gut microbial fucose/rhamnose degradation pathway is positively correlated with mRNA expression of AP-1, as well as a gene encoding an enzyme producing prostaglandin E2 (PGE2), which promotes AP-1 expression, and inversely correlated with BNT162b2-induced T-cell responses. These results suggest that baseline AP-1 expression, which is affected by commensal microbial activity, is a negative correlate of BNT162b2-induced T-cell responses.
Collapse
Affiliation(s)
- Masato Hirota
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Miho Tamai
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Sachie Yukawa
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
- Integrated Open Systems Unit, OIST, Onna-son, Okinawa, Japan
| | - Naoyuki Taira
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | | | - Takeshi Toma
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Yu Seto
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Makiko Yoshida
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Sakura Toguchi
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Mio Miyagi
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan
| | - Tomoari Mori
- Research Support Division, Occupational Health and Safety, OIST, Onna-son, Okinawa, Japan
| | | | | | | | - Eishin Sakihara
- Health Care Center of the Naha Medical Association, Naha-city, Okinawa, Japan
| | - Chiaki Yamashiro
- Yamashiro Orthopedic Surgery Ophthalmology Clinic, Naha-city, Okinawa, Japan
| | | | - Kentaro Tamaki
- Naha-Nishi Clinic, Department of Breast Surgery, Naha-city, Okinawa, Japan
| | - Matthias Wolf
- Molecular Cryo-Electron Microscopy Unit, OIST, Onna-son, Okinawa, Japan
| | - Mary K Collins
- Research Support Division, Office of the Provost, OIST, Onna-son, Okinawa, Japan
| | - Hiroaki Kitano
- Integrated Open Systems Unit, OIST, Onna-son, Okinawa, Japan
| | - Hiroki Ishikawa
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa, Japan.
| |
Collapse
|
11
|
Chandley P, Ranjan R, Kumar S, Rohatgi S. Host-parasite interactions during Plasmodium infection: Implications for immunotherapies. Front Immunol 2023; 13:1091961. [PMID: 36685595 PMCID: PMC9845897 DOI: 10.3389/fimmu.2022.1091961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Malaria is a global infectious disease that remains a leading cause of morbidity and mortality in the developing world. Multiple environmental and host and parasite factors govern the clinical outcomes of malaria. The host immune response against the Plasmodium parasite is heterogenous and stage-specific both in the human host and mosquito vector. The Plasmodium parasite virulence is predominantly associated with its ability to evade the host's immune response. Despite the availability of drug-based therapies, Plasmodium parasites can acquire drug resistance due to high antigenic variations and allelic polymorphisms. The lack of licensed vaccines against Plasmodium infection necessitates the development of effective, safe and successful therapeutics. To design an effective vaccine, it is important to study the immune evasion strategies and stage-specific Plasmodium proteins, which are targets of the host immune response. This review provides an overview of the host immune defense mechanisms and parasite immune evasion strategies during Plasmodium infection. Furthermore, we also summarize and discuss the current progress in various anti-malarial vaccine approaches, along with antibody-based therapy involving monoclonal antibodies, and research advancements in host-directed therapy, which can together open new avenues for developing novel immunotherapies against malaria infection and transmission.
Collapse
Affiliation(s)
- Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Ravikant Ranjan
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India,*Correspondence: Soma Rohatgi,
| |
Collapse
|
12
|
Daubenberger CA, Moncunill G. Next-generation malaria subunit vaccines to reduce disease burden in African children. THE LANCET. INFECTIOUS DISEASES 2022; 22:1655-1656. [PMID: 36087589 DOI: 10.1016/s1473-3099(22)00523-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Claudia A Daubenberger
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; University of Basel, Switzerland, Basel, Switzerland.
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
13
|
Beeson JG, Kurtovic L, Valim C, Asante KP, Boyle MJ, Mathanga D, Dobano C, Moncunill G. The RTS,S malaria vaccine: Current impact and foundation for the future. Sci Transl Med 2022; 14:eabo6646. [DOI: 10.1126/scitranslmed.abo6646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The RTS,S vaccine has recently been recommended for implementation as a childhood vaccine in regions with moderate-to-high malaria transmission. We discuss mechanisms of vaccine protection and longevity, implementation considerations, and future research needed to increase the vaccine’s health impact, including vaccine modifications for higher efficacy and longevity of protection.
Collapse
Affiliation(s)
- James G. Beeson
- Burnet Institute, Melbourne 3004, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, Victoria, Australia
- Monash University, Central Clinical School and Department of Microbiology, Victoria, Australia
| | - Liriye Kurtovic
- Burnet Institute, Melbourne 3004, Victoria, Australia
- Monash University, Central Clinical School and Department of Microbiology, Victoria, Australia
| | - Clarissa Valim
- Department of Global Health, Boston University School of Public Health, Boston, MA, USA
| | - Kwaku Poku Asante
- Kintampo Health Research Centre, Kintampo North Municipality, Bono East Region, Ghana
| | - Michelle J. Boyle
- QIMR Berghofer Institute, Herston, Queensland, Australia
- University of Queensland, School of Biomedical Sciences, St Lucia, Queensland, Australia
- Griffith University, Brisbane, Queensland, Australia
| | - Don Mathanga
- Malaria Alert Centre, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Carlota Dobano
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Catalonia, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
14
|
Dey S, Kaur H, Mazumder M, Brodsky E. Analysis of gene expression profiles to study malaria vaccine dose efficacy and immune response modulation. Genomics Inform 2022; 20:e32. [PMID: 36239109 PMCID: PMC9576474 DOI: 10.5808/gi.22049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2002] [Accepted: 09/04/2022] [Indexed: 11/20/2022] Open
Abstract
Malaria is a life-threatening disease, and Africa is still one of the most affected endemic regions despite years of policy to limit infection and transmission rates. Further, studies into the variable efficacy of the vaccine are needed to provide a better understanding of protective immunity. Thus, the current study is designed to delineate the effect of each dose of vaccine on the transcriptional profiles of subjects to determine its efficacy and understand the molecular mechanisms underlying the protection this vaccine provides. Here, we used gene expression profiles of pre and post-vaccination patients after various doses of RTS,S based on samples collected from the Gene Expression Omnibus datasets. Subsequently, differential gene expression analysis using edgeR revealed the significantly (false discovery rate < 0.005) 158 downregulated and 61 upregulated genes between control vs. controlled human malaria infection samples. Further, enrichment analysis of significant genes delineated the involvement of CCL8, CXCL10, CXCL11, XCR1, CSF3, IFNB1, IFNE, IL12B, IL22, IL6, IL27, etc., genes which found to be upregulated after earlier doses but downregulated after the 3rd dose in cytokine-chemokine pathways. Notably, we identified 13 cytokine genes whose expression significantly varied during three doses. Eventually, these findings give insight into the dual role of cytokine responses in malaria pathogenesis. The variations in their expression patterns after various doses of vaccination are linked to the protection as it decreases the severe inflammatory effects in malaria patients. This study will be helpful in designing a better vaccine against malaria and understanding the functions of cytokine response as well.
Collapse
Affiliation(s)
- Supantha Dey
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
- Pine Biotech, New Orleans, LA 70112, USA
- Corresponding author: ,
| | | | | | | |
Collapse
|
15
|
Nunes-Cabaço H, Moita D, Prudêncio M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front Immunol 2022; 13:977472. [PMID: 36159849 PMCID: PMC9493004 DOI: 10.3389/fimmu.2022.977472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In 1967, pioneering work by Ruth Nussenzweig demonstrated for the first time that irradiated sporozoites of the rodent malaria parasite Plasmodium berghei protected mice against a challenge with infectious parasites of the same species. This remarkable finding opened up entirely new prospects of effective vaccination against malaria using attenuated sporozoites as immunization agents. The potential for whole-sporozoite-based immunization in humans was established in a clinical study in 1973, when a volunteer exposed to X-irradiated P. falciparum sporozoites was found to be protected against malaria following challenge with a homologous strain of this parasite. Nearly five decades later, much has been achieved in the field of whole-sporozoite malaria vaccination, and multiple reports on the clinical evaluation of such candidates have emerged. However, this process has known different paces before and after the turn of the century. While only a few clinical studies were published in the 1970’s, 1980’s and 1990’s, remarkable progress was made in the 2000’s and beyond. This article reviews the history of the clinical assessment of whole-sporozoite malaria vaccines over the last forty-nine years, highlighting the impressive achievements made over the last few years, and discussing some of the challenges ahead.
Collapse
|
16
|
Kiecolt-Glaser JK, Renna M, Peng J, Sheridan J, Lustberg M, Ramaswamy B, Wesolowski R, VanDeusen JB, Williams NO, Sardesai SD, Noonan AM, Reinbolt RE, Stover DG, Cherian MA, Malarkey WB, Andridge R. Breast cancer survivors' typhoid vaccine responses: Chemotherapy, obesity, and fitness make a difference. Brain Behav Immun 2022; 103:1-9. [PMID: 35378230 PMCID: PMC9149127 DOI: 10.1016/j.bbi.2022.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
PURPOSE To investigate breast cancer survivors' inflammatory responses to typhoid vaccine as a window into their innate immune response to novel pathogens. METHODS This double-blind crossover trial randomized 158 breast cancer survivors to either the vaccine/saline placebo or the placebo/vaccine sequence. The relative contributions of age, cardiorespiratory fitness (VO2peak), type of cancer treatment, central obesity, and depression to interleukin (IL)-6, IL-1 receptor antagonist (IL-1Ra), and WBC vaccine responses were assessed pre-injection and 1.5, 3, 4.5, 6, and 7.5 h post-injection. RESULTS The vaccine produced larger IL-6, IL-1Ra, and WBC responses than placebo, ps < 0.0001. Prior chemotherapy, higher central obesity, and lower VO2peak were associated with smaller vaccine responses after controlling for baseline inflammation. Vaccine response was summarized by the percent increase in area under the curve (IL-6, WBC) or average post-injection mean (IL-1Ra) for vaccine relative to placebo. Women who received chemotherapy had smaller vaccine responses than women who did not for both IL-6 (44% vs 78%, p <.001) and WBC (26% vs 40%, p <.001); IL-1ra response was not significantly moderated by chemotherapy. Women whose central adiposity was one standard deviation above the mean had smaller vaccine responses than women with average adiposity for IL-6 (33% vs 54%, p <.001), WBC (20% vs 30%, p <.001), and IL-1Ra (2.0% vs 3.2%, p <.001). Women with an average level of VO2peak had smaller vaccine responses than women whose VO2peak was one standard deviation above the mean for IL-6 (54% vs 73%, p <.001), WBC (30% vs 40%, p <.001), and IL-1Ra (3.2% vs. 4.1%, p = 0.01). Age and depression did not significantly moderate vaccine responses. CONCLUSIONS This study provided novel data on chemotherapy's longer-term adverse immune consequences. The data also have an important public health message: even relatively low levels of fitness can benefit the innate immune response to a vaccine.
Collapse
Affiliation(s)
- Janice K. Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH,Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, Columbus, OH
| | - Megan Renna
- School of Psychology, University of Southern Mississippi, Hattiesburg, MS
| | - Juan Peng
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, College of Medicine, Columbus, OH
| | - John Sheridan
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH,Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH
| | | | - Bhuvaneswari Ramaswamy
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Robert Wesolowski
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Jeffrey B. VanDeusen
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Nicole O. Williams
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Sagar D. Sardesai
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Anne M. Noonan
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Raquel E. Reinbolt
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Daniel G. Stover
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - Mathew A. Cherian
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH,Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH
| | - William B. Malarkey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH,Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH
| | - Rebecca Andridge
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH
| |
Collapse
|
17
|
Moncunill G, Carnes J, Chad Young W, Carpp L, De Rosa S, Campo JJ, Nhabomba A, Mpina M, Jairoce C, Finak G, Haas P, Muriel C, Van P, Sanz H, Dutta S, Mordmüller B, Agnandji ST, Díez-Padrisa N, Williams NA, Aponte JJ, Valim C, Neafsey DE, Daubenberger C, McElrath MJ, Dobaño C, Stuart K, Gottardo R. Transcriptional correlates of malaria in RTS,S/AS01-vaccinated African children: a matched case–control study. eLife 2022; 11:70393. [PMID: 35060479 PMCID: PMC8782572 DOI: 10.7554/elife.70393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background: In a phase 3 trial in African infants and children, the RTS,S/AS01 vaccine (GSK) showed moderate efficacy against clinical malaria. We sought to further understand RTS,S/AS01-induced immune responses associated with vaccine protection. Methods: Applying the blood transcriptional module (BTM) framework, we characterized the transcriptomic response to RTS,S/AS01 vaccination in antigen-stimulated (and vehicle control) peripheral blood mononuclear cells sampled from a subset of trial participants at baseline and month 3 (1-month post-third dose). Using a matched case–control study design, we evaluated which of these ‘RTS,S/AS01 signature BTMs’ associated with malaria case status in RTS,S/AS01 vaccinees. Antigen-specific T-cell responses were analyzed by flow cytometry. We also performed a cross-study correlates analysis where we assessed the generalizability of our findings across three controlled human malaria infection studies of healthy, malaria-naive adult RTS,S/AS01 recipients. Results: RTS,S/AS01 vaccination was associated with downregulation of B-cell and monocyte-related BTMs and upregulation of T-cell-related BTMs, as well as higher month 3 (vs. baseline) circumsporozoite protein-specific CD4+ T-cell responses. There were few RTS,S/AS01-associated BTMs whose month 3 levels correlated with malaria risk. In contrast, baseline levels of BTMs associated with dendritic cells and with monocytes (among others) correlated with malaria risk. The baseline dendritic cell- and monocyte-related BTM correlations with malaria risk appeared to generalize to healthy, malaria-naive adults. Conclusions: A prevaccination transcriptomic signature associates with malaria in RTS,S/AS01-vaccinated African children, and elements of this signature may be broadly generalizable. The consistent presence of monocyte-related modules suggests that certain monocyte subsets may inhibit protective RTS,S/AS01-induced responses. Funding: Funding was obtained from the NIH-NIAID (R01AI095789), NIH-NIAID (U19AI128914), PATH Malaria Vaccine Initiative (MVI), and Ministerio de Economía y Competitividad (Instituto de Salud Carlos III, PI11/00423 and PI14/01422). The RNA-seq project has been funded in whole or in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under grant number U19AI110818 to the Broad Institute. This study was also supported by the Vaccine Statistical Support (Bill and Melinda Gates Foundation award INV-008576/OPP1154739 to R.G.). C.D. was the recipient of a Ramon y Cajal Contract from the Ministerio de Economía y Competitividad (RYC-2008-02631). G.M. was the recipient of a Sara Borrell–ISCIII fellowship (CD010/00156) and work was performed with the support of Department of Health, Catalan Government grant (SLT006/17/00109). This research is part of the ISGlobal’s Program on the Molecular Mechanisms of Malaria which is partially supported by the Fundación Ramón Areces and we acknowledge support from the Spanish Ministry of Science and Innovation through the ‘Centro de Excelencia Severo Ochoa 2019–2023’ Program (CEX2018-000806-S), and support from the Generalitat de Catalunya through the CERCA Program.
Collapse
Affiliation(s)
- Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona
- CIBER de Enfermedades Infecciosas
| | - Jason Carnes
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
| | - William Chad Young
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Lindsay Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Stephen De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | | | - Augusto Nhabomba
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça
| | | | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Paige Haas
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
| | - Carl Muriel
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Phu Van
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - Héctor Sanz
- ISGlobal, Hospital Clínic - Universitat de Barcelona
| | | | - Benjamin Mordmüller
- CIBER de Enfermedades Infecciosas
- Institute of Tropical Medicine and German Center for Infection Research
| | - Selidji T Agnandji
- Institute of Tropical Medicine and German Center for Infection Research
- Centre de Recherches Médicales de Lambaréné (CERMEL), BP 242
| | | | | | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona
| | - Clarissa Valim
- Department of Global Health, Boston University School of Public Health
| | - Daniel E Neafsey
- Broad Institute of Massachusetts Institute of Technology and Harvard
- Harvard T.H. Chan School of Public Health
| | | | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Departments of Laboratory Medicine and Medicine, University of Washington
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona
- CIBER de Enfermedades Infecciosas
| | - Ken Stuart
- Center for Global Infectious Disease Research, Seattle Children's Research Institute
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Department of Pediatrics, University of Washington
- Department of Global Health, University of Washington
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- University of Lausanne and Centre Hospitalier Universitaire Vaudois
| |
Collapse
|
18
|
Preimmunization correlates of protection shared across malaria vaccine trials in adults. NPJ Vaccines 2022; 7:5. [PMID: 35031601 PMCID: PMC8760258 DOI: 10.1038/s41541-021-00425-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/02/2021] [Indexed: 12/15/2022] Open
Abstract
Identifying preimmunization biological characteristics that promote an effective vaccine response offers opportunities for illuminating the critical immunological mechanisms that confer vaccine-induced protection, for developing adjuvant strategies, and for tailoring vaccination regimens to individuals or groups. In the context of malaria vaccine research, studying preimmunization correlates of protection can help address the need for a widely effective malaria vaccine, which remains elusive. In this study, common preimmunization correlates of protection were identified using transcriptomic data from four independent, heterogeneous malaria vaccine trials in adults. Systems-based analyses showed that a moderately elevated inflammatory state prior to immunization was associated with protection against malaria challenge. Functional profiling of protection-associated genes revealed the importance of several inflammatory pathways, including TLR signaling. These findings, which echo previous studies that associated enhanced preimmunization inflammation with protection, illuminate common baseline characteristics that set the stage for an effective vaccine response across diverse malaria vaccine strategies in adults.
Collapse
|
19
|
Parreira KS, Scarpelli P, Rezende Lima W, Garcia RS. Contribution of Transcriptome to Elucidate the Biology of Plasmodium spp. Curr Top Med Chem 2022; 22:169-187. [PMID: 35021974 DOI: 10.2174/1568026622666220111140803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 11/22/2022]
Abstract
In the present review, we discuss some of the new technologies that have been applied to elucidate how Plasmodium spp escape from the immune system and subvert the host physiology to orchestrate the regulation of its biological pathways. Our manuscript describes how techniques such as microarray approaches, RNA-Seq and single-cell RNA sequencing have contributed to the discovery of transcripts and changed the concept of gene expression regulation in closely related malaria parasite species. Moreover, the text highlights the contributions of high-throughput RNA sequencing for the current knowledge of malaria parasite biology, physiology, vaccine target and the revelation of new players in parasite signaling.
Collapse
Affiliation(s)
| | - Pedro Scarpelli
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo - USP, São Paulo, Brazil
| | - Wânia Rezende Lima
- Departamento de Medicina, Instituto de Biotecnologia-Universidade Federal de Catalão
| | - R S Garcia
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo - USP, São Paulo, Brazil
| |
Collapse
|
20
|
Gomez MA, Belew AT, Navas A, Rosales-Chilama M, Murillo J, Dillon LAL, Alexander TA, Martinez-Valencia A, El-Sayed NM. Early Leukocyte Responses in Ex-Vivo Models of Healing and Non-Healing Human Leishmania (Viannia) panamensis Infections. Front Cell Infect Microbiol 2021; 11:687607. [PMID: 34557423 PMCID: PMC8453012 DOI: 10.3389/fcimb.2021.687607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/16/2021] [Indexed: 11/21/2022] Open
Abstract
Early host-pathogen interactions drive the host response and shape the outcome of natural infections caused by intracellular microorganisms. These interactions involve a number of immune and non-immune cells and tissues, along with an assortment of host and pathogen-derived molecules. Our current knowledge has been predominantly derived from research on the relationships between the pathogens and the invaded host cell(s), limiting our understanding of how microbes elicit and modulate immunological responses at the organismal level. In this study, we explored the early host determinants of healing and non-healing responses in human cutaneous leishmaniasis (CL) caused by Leishmania (Viannia) panamensis. We performed a comparative transcriptomic profiling of peripheral blood mononuclear cells from healthy donors (PBMCs, n=3) exposed to promastigotes isolated from patients with chronic (CHR, n=3) or self-healing (SH, n=3) CL, and compared these to human macrophage responses. Transcriptomes of L. V. panamensis-infected PBMCs showed enrichment of functional gene categories derived from innate as well as adaptive immune cells signatures, demonstrating that Leishmania modulates adaptive immune cell functions as early as after 24h post interaction with PBMCs from previously unexposed healthy individuals. Among differentially expressed PBMC genes, four broad categories were commonly modulated by SH and CHR strains: cell cycle/proliferation/differentiation, metabolism of macromolecules, immune signaling and vesicle trafficking/transport; the first two were predominantly downregulated, and the latter upregulated in SH and CHR as compared to uninfected samples. Type I IFN signaling genes were uniquely up-regulated in PBMCs infected with CHR strains, while genes involved in the immunological synapse were uniquely downregulated in SH infections. Similarly, pro-inflammatory response genes were upregulated in isolated macrophages infected with CHR strains. Our data demonstrate that early responses during Leishmania infection extend beyond innate cell and/or phagocytic host cell functions, opening new frontiers in our understanding of the triggers and drivers of human CL.
Collapse
Affiliation(s)
- Maria Adelaida Gomez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad IcesiI, Cali, Colombia
| | - Ashton Trey Belew
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | - Adriana Navas
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Mariana Rosales-Chilama
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad IcesiI, Cali, Colombia
| | - Julieth Murillo
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Pontificia Universidad Javeriana, Cali, Colombia
| | - Laura A. L. Dillon
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| | - Theresa A. Alexander
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| | | | - Najib M. El-Sayed
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| |
Collapse
|
21
|
Seaton KE, Spreng RL, Abraha M, Reichartz M, Rojas M, Feely F, Huntwork RHC, Dutta S, Mudrak SV, Alam SM, Gregory S, Jongert E, Coccia M, Ulloa-Montoya F, Wille-Reece U, Tomaras GD, Dennison SM. Subclass and avidity of circumsporozoite protein specific antibodies associate with protection status against malaria infection. NPJ Vaccines 2021; 6:110. [PMID: 34462438 PMCID: PMC8405700 DOI: 10.1038/s41541-021-00372-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022] Open
Abstract
RTS,S/AS01 is an advanced pre-erythrocytic malaria vaccine candidate with demonstrated vaccine efficacy up to 86.7% in controlled human malaria infection (CHMI) studies; however, reproducible immune correlates of protection (CoP) are elusive. To identify candidates of humoral correlates of vaccine mediated protection, we measured antibody magnitude, subclass, and avidity for Plasmodium falciparum (Pf) circumsporozoite protein (CSP) by multiplex assays in two CHMI studies with varying RTS,S/AS01B vaccine dose and timing regimens. Central repeat (NANP6) IgG1 magnitude correlated best with protection status in univariate analyses and was the most predictive for protection in a multivariate model. NANP6 IgG3 magnitude, CSP IgG1 magnitude, and total serum antibody dissociation phase area-under-the-curve for NANP6, CSP, NPNA3, and N-interface binding were also associated with protection status in the regimen adjusted univariate analysis. Identification of multiple immune response features that associate with protection status, such as antibody subclasses, fine specificity and avidity reported here may accelerate development of highly efficacious vaccines against P. falciparum.
Collapse
Grants
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- OPP1151372, OPP12109388 Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation)
- United States Department of Defense | United States Army | Army Medical Command | Walter Reed Army Institute of Research (WRAIR)
- PATH Malaria Vaccine Initiative
- GlaxoSmithKline (GlaxoSmithKline plc.)
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
| | - Rachel L Spreng
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
| | - Milite Abraha
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Matthew Reichartz
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | | | - Frederick Feely
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Richard H C Huntwork
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - Sheetij Dutta
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sarah V Mudrak
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke Center for Human Systems Immunology, Durham, NC, USA
- Duke University Department of Surgery, Durham, NC, USA
| | - S Munir Alam
- Duke Human Vaccine Institute, Durham, NC, USA
- Duke University Department of Pathology, Durham, NC, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
| | | | | | | | - Ulrike Wille-Reece
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
- GSK, Rockville, MD, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
- Duke University Department of Immunology, Durham, NC, USA.
- Duke University Department of Molecular Genetics and Microbiology, Durham, NC, USA.
| | - S Moses Dennison
- Duke Human Vaccine Institute, Durham, NC, USA.
- Duke Center for Human Systems Immunology, Durham, NC, USA.
- Duke University Department of Surgery, Durham, NC, USA.
| |
Collapse
|
22
|
Experience counts in the malaria response. Nat Immunol 2021; 22:537-539. [PMID: 33888897 DOI: 10.1038/s41590-021-00917-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Matorras R, Valls R, Azkargorta M, Burgos J, Rabanal A, Elortza F, Mas JM, Sardon T. Proteomics based drug repositioning applied to improve in vitro fertilization implantation: an artificial intelligence model. Syst Biol Reprod Med 2021; 67:281-297. [PMID: 34126818 DOI: 10.1080/19396368.2021.1928792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Embryo implantation is one of the most inefficient steps in assisted reproduction, so the identifying drugs with a potential clinical application to improve it has a strong interest. This work applies artificial intelligence and systems biology-based mathematical modeling strategies to unveil potential treatments by computationally analyzing and integrating available molecular and clinical data from patients. The mathematical models of embryo implantation computationally generated here simulate the molecular networks underneath this biological process. Once generated, these models were analyzed in order to identify potential repositioned drugs (drugs already used for other indications) able to improve embryo implantation by modulating the molecular pathways involved. Interestingly, the repositioning analysis has identified drugs considering two endpoints: (1) drugs able to modulate the activity of proteins whose role in embryo implantation is already bibliographically acknowledged, and (2) drugs that modulate key proteins in embryo implantation previously predicted through a mechanistic analysis of the mathematical models. This second approach increases the scope open for examination and potential novelty of the repositioning strategy. As a result, a list of 23 drug candidates to improve embryo implantation after IVF was identified by the mathematical models. This list includes many of the compounds already tested for this purpose, which reinforces the predictive capacity of our approach, together with novel repositioned candidates (e.g., Infliximab, Polaprezinc, and Amrinone). In conclusion, the present study exploits existing molecular and clinical information to offer new hypotheses regarding molecular mechanisms in embryo implantation and therapeutic candidates to improve it. This information will be very useful to guide future research.Abbreviations: IVF: in vitro fertilization; EI: Embryo implantation; TPMS: Therapeutic Performance Mapping System; MM: mathematical models; ANN: Artificial Neuronal Networks; TNFα: tumour necrosis factor factor-alpha; HSPs: heat shock proteins; VEGF: vascular endothelial growth factor; PPARA: peroxisome proliferator activated receptor-α PXR: pregnane X receptor; TTR: transthyretin; BED: Biological Effectors Database; MLP: multilayer perceptron.
Collapse
Affiliation(s)
- Roberto Matorras
- Department of Obstetrics and Gynecology, University of the Basque Country, Bilbao, Spain.,IVIRMA Bilbao, Bilbao, Spain
| | | | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, Derio, Spain
| | - Jorge Burgos
- Biocruces Bizkaia Health Research Institute. Osakidetza. Cruces University Hospital, University of the Basque Country, Bilbao, Spain
| | - Aintzane Rabanal
- Department of Obstetrics and Gynecology, University of the Basque Country, Bilbao, Spain
| | - Felix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, Derio, Spain
| | | | | |
Collapse
|
24
|
Moita D, Nunes-Cabaço H, Mendes AM, Prudêncio M. A guide to investigating immune responses elicited by whole-sporozoite pre-erythrocytic vaccines against malaria. FEBS J 2021; 289:3335-3359. [PMID: 33993649 DOI: 10.1111/febs.16016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/19/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Abstract
In the last few decades, considerable efforts have been made toward the development of efficient vaccines against malaria. Whole-sporozoite (Wsp) vaccines, which induce efficient immune responses against the pre-erythrocytic (PE) stages (sporozoites and liver forms) of Plasmodium parasites, the causative agents of malaria, are among the most promising immunization strategies tested until present. Several Wsp PE vaccination approaches are currently under evaluation in the clinic, including radiation- or genetically-attenuated Plasmodium sporozoites, live parasites combined with chemoprophylaxis, or genetically modified rodent Plasmodium parasites. In addition to the assessment of their protective efficacy, clinical trials of Wsp PE vaccine candidates inevitably involve the thorough investigation of the immune responses elicited by vaccination, as well as the identification of correlates of protection. Here, we review the main methodologies employed to dissect the humoral and cellular immune responses observed in the context of Wsp PE vaccine clinical trials and discuss future strategies to further deepen the knowledge generated by these studies, providing a toolbox for the in-depth analysis of vaccine-induced immunogenicity.
Collapse
Affiliation(s)
- Diana Moita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - António M Mendes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| |
Collapse
|
25
|
Abstract
Introduction: An effective vaccine against malaria forms a global health priority. Both naturally acquired immunity and sterile protection induced by irradiated sporozoite immunization were described decades ago. Still no vaccine exists that sufficiently protects children in endemic areas. Identifying immunological correlates of vaccine efficacy can inform rational vaccine design and potentially accelerate clinical development.Areas covered: We discuss recent research on immunological correlates of malaria vaccine efficacy, including: insights from state-of-the-art omics platforms and systems vaccinology analyses; functional anti-parasitic assays; pre-immunization predictors of vaccine efficacy; and comparison of correlates of vaccine efficacy against controlled human malaria infections (CHMI) and against naturally acquired infections.Expert Opinion: Effective vaccination may be achievable without necessarily understanding immunological correlates, but the relatively disappointing efficacy of malaria vaccine candidates in target populations is concerning. Hypothesis-generating omics and systems vaccinology analyses, alongside assessment of pre-immunization correlates, have the potential to bring about paradigm-shifts in malaria vaccinology. Functional assays may represent in vivo effector mechanisms, but have scarcely been formally assessed as correlates. Crucially, evidence is still meager that correlates of vaccine efficacy against CHMI correspond with those against naturally acquired infections in target populations. Finally, the diversity of immunological assays and efficacy endpoints across malaria vaccine trials remains a major confounder.
Collapse
Affiliation(s)
| | - Matthew B B McCall
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| |
Collapse
|
26
|
Dumonteil E, Herrera C. The Case for the Development of a Chagas Disease Vaccine: Why? How? When? Trop Med Infect Dis 2021; 6:tropicalmed6010016. [PMID: 33530605 PMCID: PMC7851737 DOI: 10.3390/tropicalmed6010016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/06/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Abstract
Chagas disease is a major neglected tropical disease, transmitted predominantly by triatomine insect vectors, but also through congenital and oral routes. While endemic in the Americas, it has turned into a global disease. Because of the current drug treatment limitations, a vaccine would represent a major advancement for better control of the disease. Here, we review some of the rationale, advances, and challenges for the ongoing development of a vaccine against Chagas disease. Recent pre-clinical studies in murine models have further expanded (i) the range of vaccine platforms and formulations tested; (ii) our understanding of the immune correlates for protection; and (iii) the extent of vaccine effects on cardiac function, beyond survival and parasite burden. We further discuss outstanding issues and opportunities to move Chagas disease development forward in the near future.
Collapse
|
27
|
Nyarko PB, Claessens A. Understanding Host-Pathogen-Vector Interactions with Chronic Asymptomatic Malaria Infections. Trends Parasitol 2020; 37:195-204. [PMID: 33127332 DOI: 10.1016/j.pt.2020.09.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 01/06/2023]
Abstract
The last malaria parasite standing will display effective adaptations to selective forces. While substantial progress has been made in reducing malaria mortality, eradication will require elimination of all Plasmodium parasites, including those in asymptomatic infections. These typically chronic, low-density infections are difficult to detect, yet can persist for months. We argue that asymptomatic infection is the parasite's best asset for survival but it can be exploited if studied as a new model for host-pathogen-vector interactions. Regular sampling from cohorts of asymptomatic individuals can provide a means to investigate continuous parasite development within its natural host. State-of-the-art techniques can now be applied to such infections. This approach may reveal key molecular drivers of chronic infections - a critical step for malaria eradication.
Collapse
Affiliation(s)
- Prince B Nyarko
- Laboratory of Pathogen-Host Interaction (LPHI), CNRS, University of Montpellier, France
| | | |
Collapse
|
28
|
McCall MBB, Yap XZ, Bousema T. Optimizing RTS,S Vaccination Strategies: Give It Your Best Parting Shot. J Infect Dis 2020; 222:1581-1584. [DOI: 10.1093/infdis/jiaa423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Matthew B B McCall
- Department of Medical Microbiology, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
- Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| | - Xi Zen Yap
- Department of Medical Microbiology, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
29
|
Coban C. The host targeting effect of chloroquine in malaria. Curr Opin Immunol 2020; 66:98-107. [PMID: 32823144 PMCID: PMC7431399 DOI: 10.1016/j.coi.2020.07.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 01/07/2023]
Abstract
Due to the rapid onset and spread of the COVID-19 pandemic, the treatment of COVID-19 patients by hydroxychloroquine alone or in combination with other drugs has captured a great deal of attention and triggered considerable debate. Historically, the worldwide use of quinoline based-drugs has led to a spectacular reduction in death from malaria. Unfortunately, scientists have been forced to seek alternative drugs to treat malaria due to the emergence of chloroquine-resistant parasites in the 1960s. The repurposing of hydroxychloroquine against viral infections, various types of cancer and autoimmune diseases has been ongoing for more than 70 years, with no clear understanding of its mechanism of action (MOA). Here, we closely examine the MOA of this old but influential drug in and beyond malaria. Better insights into how chloroquine targets the host's cellular and immune responses may help to develop applications against to new pathogens and diseases, and perhaps even restore the clinical utility of chloroquine against malaria.
Collapse
Affiliation(s)
- Cevayir Coban
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo, Japan; Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.
| |
Collapse
|