1
|
Ratnapriya S, Yadav AK, Sahasrabuddhe AA, Dube A. Immunotherapeutic efficacy of Th1 stimulatory epitopes derived from Leishmania donovani enolase and triose phosphatase isomerase in experimental visceral leishmaniasis. Mol Immunol 2025; 183:104-114. [PMID: 40347781 DOI: 10.1016/j.molimm.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/20/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
Visceral leishmaniasis is a lethal systemic disease which cannot be controlled by drugs alone given asymptomatic reservoirs and must include alternatives to modulate immune responses of the endemic contacts. Our previous studies have yielded potential immunomodulatory proteins by proteomic approach and their peptides showed significant immunomodulation and prophylactic efficacies. The current study has been intended to evaluate immunotherapeutic potential of the lead peptides (P10, P14 and P15) and their combinations with BCG as adjuvant. Results indicated P10 + 14 as well as P10 + P15 to be the most potential therapeutic peptides cocktails as both these combinations reduced parasite burden by > 65 % eliciting remarkable cellular response. High level of IFN-γ, TNF-α and IL-12 with decreased TGF-β expression suggested skewing of immune response towards Th1 type. These results were further supported by strong DTH and lymphoproliferative responses indicating therapeutic potential of the peptide combinations and possibilities in Kala-azar elimination program.
Collapse
Affiliation(s)
| | - Alok K Yadav
- Biochemistry and Structural Biology Division, India
| | | | - Anuradha Dube
- CSIR-Central Drug Research Institute, Jankipuram Extension, Sector 10, Sitapur Road, Lucknow 226031, India; Molecular Microbiology and Immunology Division, Jankipuram Extension, Sector 10, Sitapur Road, Lucknow 226031, India.
| |
Collapse
|
2
|
Montgomery L, Larbi A. Monitoring Immune Responses to Vaccination: A Focus on Single-Cell Analysis and Associated Challenges. Vaccines (Basel) 2025; 13:420. [PMID: 40333304 PMCID: PMC12030821 DOI: 10.3390/vaccines13040420] [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: 03/17/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Monitoring the immune response to vaccination encompasses both significant challenges and promising opportunities for scientific advancement. The primary challenge lies in the inherent complexity and interindividual variability of immune responses, influenced by factors including age, genetic background, and prior immunological history. This variability necessitates the development of sophisticated, highly sensitive assays capable of accurately quantifying immune parameters such as antibody titers, T-cell responses, and cytokine profiles. Furthermore, the temporal dynamics of the immune response require comprehensive longitudinal studies to elucidate the durability and quality of vaccine-induced immunity. Challenges of this magnitude pave the way for immunological research advancements and diagnostic methodologies. Cutting-edge monitoring techniques, such as high-throughput sequencing and advanced flow cytometry, enable deeper insights into the mechanistic underpinnings of vaccine efficacy and contribute to the iterative design of more effective vaccines. Additionally, the integration of analytical tools holds the potential to predict immune responses and tailor personalized vaccination strategies. This will be addressed in this review to provide insight for enhancing public health outcomes and fortifying preparedness against future infectious disease threats.
Collapse
Affiliation(s)
- LaToya Montgomery
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, Brea, CA 92821, USA;
| | - Anis Larbi
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, Brea, CA 92821, USA;
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| |
Collapse
|
3
|
Liu W, Huang X. Very virulent infectious bursal disease virus infection triggered microscopic changes, apoptosis, and inflammatory cytokines imbalance in chicken spleen and thymus. Avian Pathol 2025; 54:62-75. [PMID: 38995197 DOI: 10.1080/03079457.2024.2380420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/22/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024]
Abstract
Infectious bursal disease virus (IBDV) can cause a highly contagious disease, resulting in severe damage to the immune system that causes immunosuppression in young chickens. Both spleen and thymus are important immune organs, which play a key role in eliciting protective immune responses. However, the effects of very virulent IBDV (vvIBDV) strain LJ-5 infection on chicken spleen and thymus are still unknown. In the present study, 3-week-old specific pathogen-free chickens were infected with vvIBDV for 1-5 days. The vvIBDV infection significantly increased the spleen index and decreased the thymus index. Microscopic analysis indicated necrosis, depletion of the lymphoid cells, and complete loss of structural integrity in spleen and thymus. Ultrastructural analysis displayed mitochondrial and nuclear damage, including mitochondrial cristae breaks, and deformation of nuclear membrane in vvIBDV-infected spleen and thymus tissues. Cytokine levels increased in the spleen and thymus after IBDV infection, promoting inflammation and causing an inflammatory imbalance. Moreover, the mRNA expression of apoptosis-related genes was significantly upregulated in the vvIBDV-infected group compared to the control group. Meanwhile, the mRNA expression of mitochondrial dynamics was altered in the spleen and thymus of vvIBDV-infected chickens. These results suggested that vvIBDV infection triggers an imbalance of inflammatory cytokines, and apoptosis in the spleen and thymus, resulting in immune injury in chickens. This study provides basic data for the further study of vvIBDV pathogenesis.
Collapse
Affiliation(s)
- Weiye Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Xuewei Huang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, People's Republic of China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
4
|
Mabrouk SG, El-Nokrashy AM, Ebied NA, Abdella BH, Zayed MM, Aboleila SM, Mohamed RA. A Blend of natural phytobiotics enhances growth performance, feed efficiency, and the immuno-health status of fingerlings of Nile tilapia ( Oreochromis niloticus). Open Vet J 2025; 15:746-764. [PMID: 40201808 PMCID: PMC11974310 DOI: 10.5455/ovj.2025.v15.i2.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/19/2025] [Indexed: 04/10/2025] Open
Abstract
Background Numerous natural phytobiotic products are used as feed additives to enhance fish performance, quality, and immunity. Aim This study evaluated the effect of a natural phytobiotics mixture [Syrena Boost (SB)] on growth performance, intestine health, immune-oxidative status, and hemato-biochemical parameters of fingerlings (Oreochromis niloticus). Methods Fish (n = 4,800, average initial weight = 1.247 ± 0.047 g) were randomly distributed in 12 aquatic compartments with a total water volume of 2 m3 (2 × 1 × 1 m) for each one, representing four groups, in triplicate: control group (CG), SB1, SB2, and SB3, in which fish received a basal diet (30/6, protein/lipid ratio) containing 0, 0.1, 0.2, 0.4 g kg-1 SB, respectively, for 60 days. Results The results showed a significant (p < 0.05) improvement in fish performance (survival rate, specific growth rate, weight gain, final weight, and feed conversion ratio) and intestine histomorphology (increased goblet cell number, intestinal villi length, but inter-villi space, decreased) in fish that received SB1, SB2, and SB3 compared with CG (p < 0.05) with the most favorable outcomes observed in treatment supplemented with SB2. Dietary inclusion of SB at different doses (SB1, SB2, and SB3) improved certain blood hematological parameters, including globulin and total protein, while decreasing aspartate aminotransferase, alanine aminotransferase, cholesterol, triglycerides, glucose, and albumin compared with CG (p < 0.05), with the most favorable outcomes observed in SB2. The fish showed improvements in digestive enzymes (lipase and amylase), antioxidant enzymes (increased catalase, superoxide dismutase), as well as a reduction in malondialdehyde, as well as showed improvement in the immunity health indicators (phagocytic index, phagocytic activity, and lysozyme activity), and gene expression (glutathione peroxidase, and catalase, tumor necrosis factor-alpha, interleukin-1β, insulin-like growth factor, interleukin-8, and growth hormone receptor) when they received SB1, SB2, and SB3 supplemented diets, with the most favorable outcomes observed in SB2 compared with CG (p < 0.05). Thus, the SB at 0.2 g kg-1 diet (SB2) can be used effectively in Tilapia diets to improve growth, intestinal health, blood health, oxidative status, and immune-related gene expression. Conclusion Dietary supplementation with a natural phytobiotic mixture (SB®) improved fish performance, intestine health, and the immune-physiological status of Nile tilapia fingerlings. The best findings in this experiment were observed in fish that received SB2.
Collapse
Affiliation(s)
- Shreifa G. Mabrouk
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Asmaa M. El-Nokrashy
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Nahla A. Ebied
- Food Hygiene Unit, Animal Health Research Institute, Provisional Lab Kafr El-Sheikh, Agricultural Research Center (ARC), Kafr El-Sheikh, Egypt
| | - Bahaa H. Abdella
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Mohamed M. Zayed
- Department of Chemistry, Rabigh College of Sciences and Arts, King Abdulaziz University, Jeddah, Saudi Arabia
- Environmental and Occupational Medicine Department, National Research Centre, Cairo, Egypt
| | - Sayed M. Aboleila
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Radi A. Mohamed
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| |
Collapse
|
5
|
Shapiro JR, Corrado M, Perry J, Watts TH, Bolotin S. The contributions of T cell-mediated immunity to protection from vaccine-preventable diseases: A primer. Hum Vaccin Immunother 2024; 20:2395679. [PMID: 39205626 PMCID: PMC11364080 DOI: 10.1080/21645515.2024.2395679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
In the face of the ever-present burden of emerging and reemerging infectious diseases, there is a growing need to comprehensively assess individual- and population-level immunity to vaccine-preventable diseases (VPDs). Many of these efforts, however, focus exclusively on antibody-mediated immunity, ignoring the role of T cells. Aimed at clinicians, public health practioners, and others who play central roles in human vaccine research but do not have formal training in immunology, we review how vaccines against infectious diseases elicit T cell responses, what types of vaccines elicit T cell responses, and how T cell responses are measured. We then use examples to demonstrate six ways that T cells contribute to protection from VPD, including directly mediating protection, enabling antibody responses, reducing disease severity, increasing cross-reactivity, improving durability, and protecting special populations. We conclude with a discussion of challenges and solutions to more widespread consideration of T cell responses in clinical vaccinology.
Collapse
Affiliation(s)
- Janna R. Shapiro
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Mario Corrado
- Division of General Internal Medicine, University of Toronto, Toronto, ON, Canada
| | - Julie Perry
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tania H. Watts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Shelly Bolotin
- Center for Vaccine Preventable Diseases, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Health Protection, Public Health Ontario, Toronto, ON, Canada
| |
Collapse
|
6
|
Wu YY, Wu FH, Chen IC, Liao TL, Munir M, Liu HJ. Oncolytic avian reovirus-sensitized tumor infiltrating CD8 + T cells triggering immunogenic apoptosis in gastric cancer. Cell Commun Signal 2024; 22:514. [PMID: 39434159 PMCID: PMC11494775 DOI: 10.1186/s12964-024-01888-0] [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: 07/08/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a leading malignant disease in numerous countries, including Taiwan with limited therapeutic options. Animal viruses including oncolytic avian reovirus (ARV) have the possibility to avoid pre-existing immunity in humans, while being safe and immunostimulatory. Here, we provide a novel insight into oncolytic ARV and UV-ARV-sensitized patient's peripheral blood mononuclear cells (P-PBMCs) and tumor infiltrating lymphocytes (TILs) killing primary GC (PGC) cells through the surface TLR3 and TRAIL/DR4/DR5 immunogenic apoptosis pathway. METHODS We conducted a comprehensive study to reveal whether ARV- or UV-inactivated ARV (UV-ARV)-modulated P-PBMCs or TILs killing ARV- and UV-ARV-sensitized AGS cells and PGC cells derived from clinical patients and to investigate the regulation of surface TLR3 receptor and upstream signaling pathways. Apoptosis analysis by flow cytometry and Western blot, suppression of signal pathway by specific inhibitors, in situ proximity ligation assay (PLA), time-resolved flurometry and lactate dehydrogenase (LDH) cytotoxicity assays, and an in vitro co-culture model were established to study the interplay between ARV- and UV-ARV-sensitized P-PBMCs and TILs to kill PGC cells and their upstream pathways. RESULTS Our results reveal that increased levels of DR4 and DR5 were observed in ARV and UV-ARV sensitized PGC cells through the TLR3/p38/p53 signaling pathway. Importantly, we found that the σC protein of ARV or UV-ARV interacted with surface TLR3 of CD8+ TILs, thereby triggering the TLR3/NF-κB/IFN-γ/TRAIL signaling pathway which induces immunogenic apoptosis of PGC cells. This study sheds further light on the molecular basis behind ARV oncolysis and facilitates the ARV or UV-ARV as a cancer therapeutic. CONCLUSIONS The study provides novel insights into ARV- or UV-ARV-sensitized P-PBMCs and CD8+ TILs to kill PGC cells through the immunogenic apoptosis pathway. We conclude that P-PBMCs can easily be obtained from GC patients and provide a rich source as TILs to kill PGC cells.
Collapse
Affiliation(s)
- Yi-Ying Wu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Feng-Hsu Wu
- Department of Critical Care, Taichung Veterans General Hospital, Taichung, Taiwan
- Division of General Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Nursing, Hung Kuang University, Taichung, Taiwan
| | - I-Chun Chen
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan
- Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Hung-Jen Liu
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 402, Taiwan.
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
7
|
Calvo JR, Maldonado MD. Immunoregulatory properties of melatonin in the humoral immune system: A narrative review. Immunol Lett 2024; 269:106901. [PMID: 39032910 DOI: 10.1016/j.imlet.2024.106901] [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: 06/18/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Melatonin is the major product both synthesized and secreted by the pineal gland during the night period and it is the principal chronobiotic hormone that regulates the circadian rhythms and seasonal changes in vertebrate biology. Moreover, melatonin shows both a broad distribution along the phylogenetically distant organisms and a high functional versatility. At the present time, a significant amount of experimental evidence has been reported in scientific literature and has clearly shown a functional relationship between the endocrine, nervous, and immune systems. The biochemistry basis of the functional communication between these systems is the utilization of a common chemicals signals. In this framework, at present melatonin is considered to be a relevant member of the so-called neuro-endocrine-immunological network. Thus, both in vivo and in vitro investigations conducted in both experimental animals and humans, have clearly documented that melatonin has an important immunomodulatory role. However, most of the published results refer to information on T lymphocytes, i.e., cell-mediated immunity. On the contrary, fewer studies have been carried out on B lymphocytes, the cells responsible for the so-called humoral immunity. In this review, we have focused on the biological role of melatonin in the humoral immunity. More precisely, we report the actions of melatonin on B lymphocytes biology and on the production of different types of antibodies.
Collapse
Affiliation(s)
- Juan R Calvo
- Department Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Spain.
| | - María D Maldonado
- Department Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Spain
| |
Collapse
|
8
|
Obeagu EI, Okoroiwu GI, Ubosi NI, Obeagu GU, Onohuean H, Muhammad T, Adias TC. Revolution in malaria detection: unveiling current breakthroughs and tomorrow's possibilities in biomarker innovation. Ann Med Surg (Lond) 2024; 86:5859-5876. [PMID: 39359838 PMCID: PMC11444567 DOI: 10.1097/ms9.0000000000002383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/06/2024] [Indexed: 10/04/2024] Open
Abstract
The ongoing battle against malaria has seen significant advancements in diagnostic methodologies, particularly through the discovery and application of novel biomarkers. Traditional diagnostic techniques, such as microscopy and rapid diagnostic tests, have their limitations in terms of sensitivity, specificity, and the ability to detect low-level infections. Recent breakthroughs in biomarker research promise to overcome these challenges, providing more accurate, rapid, and non-invasive detection methods. These advancements are critical in enhancing early detection, guiding effective treatment, and ultimately reducing the global malaria burden. Innovative approaches in biomarker detection are leveraging cutting-edge technologies like next-generation sequencing, proteomics, and metabolomics. These techniques have led to the identification of new biomarkers that can be detected in blood, saliva, or urine, offering less invasive and more scalable options for widespread screening. For instance, the discovery of specific volatile organic compounds in the breath of infected individuals presents a revolutionary non-invasive diagnostic tool. Additionally, the integration of machine learning algorithms with biomarker data is enhancing the precision and predictive power of malaria diagnostics, making it possible to distinguish between different stages of infection and identify drug-resistant strains. Looking ahead, the future of malaria detection lies in the continued exploration of multi-biomarker panels and the development of portable, point-of-care diagnostic devices. The incorporation of smartphone-based technologies and wearable biosensors promises to bring real-time monitoring and remote diagnostics to even the most resource-limited settings.
Collapse
Affiliation(s)
| | - G. I.A. Okoroiwu
- Department of Public Health Science, Faculty of Health Sciences, National Open University of Nigeria, Jabi, Abuja
| | - N. I. Ubosi
- Department of Public Health Science, Faculty of Health Sciences, National Open University of Nigeria, Jabi, Abuja
| | | | - Hope Onohuean
- Biopharmaceutics Unit, Department of Pharmacology and Toxicology, School of Pharmacy, Kampala International University, Kampala
- Biomolecules, Metagenomics, Endocrine and Tropical Disease Research Group (BMETDREG), Kampala International University, Western Campus, Ishaka-Bushenyi, Uganda
| | - Tukur Muhammad
- Department of Science Education & Educational Foundations, Faculty of Education Kampala International University Western Campus
| | - Teddy C. Adias
- Department of Haematology and Blood Transfusion Science, Faculty of Medical Laboratory Science, Federal University Otuoke, Bayelsa State, Nigeria
| |
Collapse
|
9
|
Kayesh MEH, Hashem MA, Tsukiyama-Kohara K. Epidemiology, Transmission Mode, and Pathogenesis of Chlamydia pecorum Infection in Koalas ( Phascolarctos cinereus): An Overview. Animals (Basel) 2024; 14:2686. [PMID: 39335275 PMCID: PMC11440102 DOI: 10.3390/ani14182686] [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: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Chlamydial infections pose a significant threat to koala populations. Chlamydia pecorum (C. pecorum) remains the major chlamydial species affecting koala health, both in the wild and in captivity, and chlamydial infections are considered important factors affecting the long-term survival of koalas. A clear understanding of chlamydial infections, including the epidemiology, transmission mode, pathogenesis, immune response, control, and prevention thereof, is essential for improving the management of chlamydial infections in koalas. In this study, we discuss the important advances made in our understanding of C. pecorum infection in koalas, focusing on the epidemiology of chlamydial infections, and the transmission, pathogenesis, immune response, and control strategies for chlamydial infection, with the aim of improving koala health and achieving effective conservation strategies.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Md Abul Hashem
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
| |
Collapse
|
10
|
Kuhlman KR, Radin A, Cole SW, Bower JE. Psychosocial predictors of the innate immune response to influenza vaccination. Psychoneuroendocrinology 2024; 163:106989. [PMID: 38354453 DOI: 10.1016/j.psyneuen.2024.106989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Experimental activation of the innate immune system has contributed significantly to both our understanding of how psychological factors influence immune function as well as how immune activity influences the brain and behavior. The annual influenza vaccine can be used to interrogate the effects of mild immune stimulation on day-to-day changes in psychological processes in human subjects that range across the lifespan and in both clinical and non-clinical populations. Yet, the immune response to the influenza vaccine in the days immediately following its administration are not well characterized. The present study describes changes in inflammatory and antiviral gene expression within circulating immune cells, plasma cytokines, and C-reactive protein (CRP) following receipt of the flu vaccine, and further reports the association between several common behavioral health factors and the acute immune response. Participants were 65 adults (mean age 18.81 ± 1.03 years; 66.2% female) who provided a blood sample immediately before and then 24 h after receiving the vaccine. A subsample also provided additional blood samples at 48 and 72 h. Plasma was assayed for CRP, IL-6, IL-10, IL-8, TNF-α, and IFN-γ, and peripheral blood mononuclear cell RNA was sequenced for evidence of change in expression of an a priori set of type 1 interferon (IFN) and inflammatory response genes (INFLAM). Plasma cytokines, CRP, and IFN response genes increased 24 h after vaccination, all ps < .001. The increase in IFN gene expression correlated with the observed increase in plasma cytokines and CRP, p < .0001. The immune response to influenza vaccination at 24-hours was moderated by anxiety symptoms, BMI, being female, sleep, and history of influenza vaccination. These factors and their associations with common immune challenges may be useful in studies interrogating the origins of immune dysregulation. The annual influenza vaccine is an accessible and reliable exogenous activator of both circulating and transcriptional markers of innate immune reactivity, with sensitivity to behavioral health factors relevant for psychoneuroimmunology research.
Collapse
Affiliation(s)
- Kate Ryan Kuhlman
- Department of Psychological Science, School of Social Ecology, University of California Irvine, Irvine, CA, USA; Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Arielle Radin
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| | - Steve W Cole
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Julienne E Bower
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA; Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
11
|
Debbag R, Torres JR, Falleiros-Arlant LH, Avila-Aguero ML, Brea-del Castillo J, Gentile A, Saez-Llorens X, Mascarenas A, Munoz FM, Torres JP, Vazquez L, Safadi MA, Espinal C, Ulloa-Gutierrez R, Pujadas M, Lopez P, López-Medina E, Ramilo O. Are the first 1,000 days of life a neglected vital period to prevent the impact on maternal and infant morbimortality of infectious diseases in Latin America? Proceedings of a workshop of experts from the Latin American Pediatric Infectious Diseases Society, SLIPE. Front Pediatr 2023; 11:1297177. [PMID: 38098643 PMCID: PMC10720332 DOI: 10.3389/fped.2023.1297177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023] Open
Abstract
While the first 1,000 days of life are a critical period in child's development, limited information on the main determinants affecting this period in the Latin America and the Caribbean (LAC) region is available. Therefore, the Latin American Pediatric Infectious Diseases Society (SLIPE) held an ad hoc workshop in May 2022 with an expert panel designed to analyze the main factors impacting the development of childhood in the region during this period and the main causes of maternal infant morbimortality. The aim was to identify priorities, generate recommendations, and advise practical actions to improve this situation. Considerations were made about the challenges involved in bridging the gap that separates the region from more developed countries regarding an optimal early childhood and maternal care. Extensive discussion was conducted to reach consensus recommendations on general strategies intended to reduce maternal and infant mortality associated with infections and immune-preventable diseases during the first 1,000 days of life in LAC.
Collapse
Affiliation(s)
- Roberto Debbag
- President of Sociedad Latinoamericana de Infectología Pediátrica, SLIPE, Buenos Aires, Argentina
| | - Jaime R. Torres
- Infectious Diseases Section, Tropical Medicine Institute, Universidad Central De Venezuela, Caracas, Venezuela
| | - Luiza H. Falleiros-Arlant
- Department of Children’s Health, Faculdade De Medicina, Universidade Metropolitana De Santos, Santos, Brazil
| | - Maria L. Avila-Aguero
- Infectious Diseases Service, Hospital Nacional De Niños “Dr. Carlos Sáenz Herrera”, Caja Costarricense De Seguro Social (CCSS), San José, Costa Rica
- Affiliated Researcher Center for Infectious Disease Modeling and Analysis (CIDMA) at Yale University, New Haven, CT, United States
| | - Jose Brea-del Castillo
- Associated Researcher, Investigador Asociado Hospital Dr. Hugo Mendoza, Santo Domingo, Republic Dominicana
| | - Angela Gentile
- Epidemiology Department, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires University, Buenos Aires, Argentina
| | - Xavier Saez-Llorens
- Head of Infectious Diseases and Director of Clinical Research, Hospital del Niño “Dr. José Renán Esquivel”, Panama City, Panama
| | - Abiel Mascarenas
- Department of Pediatric Infectious Diseases, Hospital Universitario “José E. Gonzalez”, Universidad Autónoma De Nuevo León, Nuevo Leon, México
| | - Flor M. Munoz
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Juan P. Torres
- Department of Pediatrics and Children Surgery, Universidad de Chile, Santiago, Chile
| | - Liliana Vazquez
- Pediatric Infectious Diseases, Clinica y Maternidad Suizo Argentina, Sanatorio Finochietto, Buenos Aires, Argentina
| | - Marco A. Safadi
- Department of Pediatrics, Faculda de de Ciências Médicas da Santa Casa de São Paulo, Sao Paulo, Brazil
| | - Carlos Espinal
- Global Health Consortium, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL, United States
| | - Rolando Ulloa-Gutierrez
- Infectious Diseases Service, Hospital Nacional De Niños “Dr. Carlos Sáenz Herrera”, Caja Costarricense De Seguro Social (CCSS), San José, Costa Rica
| | - Monica Pujadas
- Department of Epidemiology and Pediatric Infectious Diseases, Centro Hospitalario Pereira Rossell, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Pio Lopez
- Department of Pediatrics, Hospital Universitario del Valle, Cali, Colombia
| | - Eduardo López-Medina
- Centro de Estudios en Infectología Pediátrica CEIP, Department of Pediatrics, Universidad del Valle, Clinica Imbanaco Grupo Quironsalud, Cali, Colombia
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
12
|
Gygi JP, Konstorum A, Pawar S, Aron E, Kleinstein SH, Guan L. A supervised Bayesian factor model for the identification of multi-omics signatures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525545. [PMID: 36747790 PMCID: PMC9900835 DOI: 10.1101/2023.01.25.525545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
MOTIVATION Predictive biological signatures provide utility as biomarkers for disease diagnosis and prognosis, as well as prediction of responses to vaccination or therapy. These signatures are iden-tified from high-throughput profiling assays through a combination of dimensionality reduction and machine learning techniques. The genes, proteins, metabolites, and other biological analytes that compose signatures also generate hypotheses on the underlying mechanisms driving biological responses, thus improving biological understanding. Dimensionality reduction is a critical step in signature discovery to address the large number of analytes in omics datasets, especially for multi-omics profiling studies with tens of thousands of measurements. Latent factor models, which can account for the structural heterogeneity across diverse assays, effectively integrate multi-omics data and reduce dimensionality to a small number of factors that capture correlations and associations among measurements. These factors provide biologically interpretable features for predictive model-ing. However, multi-omics integration and predictive modeling are generally performed independent-ly in sequential steps, leading to suboptimal factor construction. Combining these steps can yield better multi-omics signatures that are more predictive while still being biologically meaningful. RESULTS We developed a supervised variational Bayesian factor model that extracts multi-omics signatures from high-throughput profiling datasets that can span multiple data types. Signature-based multiPle-omics intEgration via lAtent factoRs (SPEAR) adaptively determines factor rank, emphasis on factor structure, data relevance and feature sparsity. The method improves the recon-struction of underlying factors in synthetic examples and prediction accuracy of COVID-19 severity and breast cancer tumor subtypes. AVAILABILITY SPEAR is a publicly available R-package hosted at https://bitbucket.org/kleinstein/SPEAR.
Collapse
|
13
|
Gygi JP, Kleinstein SH, Guan L. Predictive overfitting in immunological applications: Pitfalls and solutions. Hum Vaccin Immunother 2023; 19:2251830. [PMID: 37697867 PMCID: PMC10498807 DOI: 10.1080/21645515.2023.2251830] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/27/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
Overfitting describes the phenomenon where a highly predictive model on the training data generalizes poorly to future observations. It is a common concern when applying machine learning techniques to contemporary medical applications, such as predicting vaccination response and disease status in infectious disease or cancer studies. This review examines the causes of overfitting and offers strategies to counteract it, focusing on model complexity reduction, reliable model evaluation, and harnessing data diversity. Through discussion of the underlying mathematical models and illustrative examples using both synthetic data and published real datasets, our objective is to equip analysts and bioinformaticians with the knowledge and tools necessary to detect and mitigate overfitting in their research.
Collapse
Affiliation(s)
- Jeremy P. Gygi
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
| | - Steven H. Kleinstein
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Leying Guan
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
14
|
Affaticati F, Bartholomeus E, Mullan K, Damme PV, Beutels P, Ogunjimi B, Laukens K, Meysman P. Multi-View Learning to Unravel the Different Levels Underlying Hepatitis B Vaccine Response. Vaccines (Basel) 2023; 11:1236. [PMID: 37515051 PMCID: PMC10384938 DOI: 10.3390/vaccines11071236] [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: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The immune system acts as an intricate apparatus that is dedicated to mounting a defense and ensures host survival from microbial threats. To engage this faceted immune response and provide protection against infectious diseases, vaccinations are a critical tool to be developed. However, vaccine responses are governed by levels that, when interrogated, separately only explain a fraction of the immune reaction. To address this knowledge gap, we conducted a feasibility study to determine if multi-view modeling could aid in gaining actionable insights on response markers shared across populations, capture the immune system's diversity, and disentangle confounders. We thus sought to assess this multi-view modeling capacity on the responsiveness to the Hepatitis B virus (HBV) vaccination. Seroconversion to vaccine-induced antibodies against the HBV surface antigen (anti-HBs) in early converters (n = 21; <2 months) and late converters (n = 9; <6 months) and was defined based on the anti-HBs titers (>10IU/L). The multi-view data encompassed bulk RNA-seq, CD4+ T-cell parameters (including T-cell receptor data), flow cytometry data, and clinical metadata (including age and gender). The modeling included testing single-view and multi-view joint dimensionality reductions. Multi-view joint dimensionality reduction outperformed single-view methods in terms of the area under the curve and balanced accuracy, confirming the increase in predictive power to be gained. The interpretation of these findings showed that age, gender, inflammation-related gene sets, and pre-existing vaccine-specific T-cells could be associated with vaccination responsiveness. This multi-view dimensionality reduction approach complements clinical seroconversion and all single modalities. Importantly, this modeling could identify what features could predict HBV vaccine response. This methodology could be extended to other vaccination trials to identify the key features regulating responsiveness.
Collapse
Affiliation(s)
- Fabio Affaticati
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Esther Bartholomeus
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp (VAXINFECTIO), 2610 Antwerp, Belgium
| | - Kerry Mullan
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Pierre Van Damme
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for the Evaluation of Vaccination (CEV), Vaccine and Infectious Disease Institute, University of Antwerp, 2610 Antwerp, Belgium
| | - Philippe Beutels
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2610 Antwerp, Belgium
| | - Benson Ogunjimi
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp (VAXINFECTIO), 2610 Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| |
Collapse
|
15
|
Pasin C, Consiglio CR, Huisman J, de Lange AMG, Peckham H, Vallejo-Yagüe E, Abela IA, Islander U, Neuner-Jehle N, Pujantell M, Roth O, Schirmer M, Tepekule B, Zeeb M, Hachfeld A, Aebi-Popp K, Kouyos RD, Bonhoeffer S. Sex and gender in infection and immunity: addressing the bottlenecks from basic science to public health and clinical applications. ROYAL SOCIETY OPEN SCIENCE 2023; 10:221628. [PMID: 37416827 PMCID: PMC10320357 DOI: 10.1098/rsos.221628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/14/2023] [Indexed: 07/08/2023]
Abstract
Although sex and gender are recognized as major determinants of health and immunity, their role is rarely considered in clinical practice and public health. We identified six bottlenecks preventing the inclusion of sex and gender considerations from basic science to clinical practice, precision medicine and public health policies. (i) A terminology-related bottleneck, linked to the definitions of sex and gender themselves, and the lack of consensus on how to evaluate gender. (ii) A data-related bottleneck, due to gaps in sex-disaggregated data, data on trans/non-binary people and gender identity. (iii) A translational bottleneck, limited by animal models and the underrepresentation of gender minorities in biomedical studies. (iv) A statistical bottleneck, with inappropriate statistical analyses and results interpretation. (v) An ethical bottleneck posed by the underrepresentation of pregnant people and gender minorities in clinical studies. (vi) A structural bottleneck, as systemic bias and discriminations affect not only academic research but also decision makers. We specify guidelines for researchers, scientific journals, funding agencies and academic institutions to address these bottlenecks. Following such guidelines will support the development of more efficient and equitable care strategies for all.
Collapse
Affiliation(s)
- Chloé Pasin
- Collegium Helveticum, 8092 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Camila R. Consiglio
- Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Jana S. Huisman
- Institute of Integrative Biology, ETH Zurich, 8092 Zurich, Switzerland
- Physics of Living Systems, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ann-Marie G. de Lange
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
- Department of Psychology, University of Oslo, 0373 Oslo, Norway
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH and GOSH, London WC1E 6JF, UK
| | | | - Irene A. Abela
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Ulrika Islander
- Department of Rheumatology and Inflammation Research, University of Gothenburg, 40530 Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Nadia Neuner-Jehle
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Maria Pujantell
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Olivia Roth
- Marine Evolutionary Biology, Zoological Institute, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Melanie Schirmer
- Emmy Noether Group for Computational Microbiome Research, ZIEL – Institute for Food and Health, Technical University of Munich, 85354 Freising, Germany
| | - Burcu Tepekule
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Marius Zeeb
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Anna Hachfeld
- Department of Infectious Diseases, University Hospital and University of Bern, 3012 Bern, Switzerland
| | - Karoline Aebi-Popp
- Department of Infectious Diseases, University Hospital and University of Bern, 3012 Bern, Switzerland
- Department of Obstetrics and Gynecology, Lindenhofspital, 3012 Bern, Switzerland
| | - Roger D. Kouyos
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Sebastian Bonhoeffer
- Collegium Helveticum, 8092 Zurich, Switzerland
- Institute of Integrative Biology, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
16
|
Distinct immunological and molecular signatures underpinning influenza vaccine responsiveness in the elderly. Nat Commun 2022; 13:6894. [PMID: 36371426 PMCID: PMC9653450 DOI: 10.1038/s41467-022-34487-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Seasonal influenza outbreaks, especially in high-risk groups such as the elderly, represent an important public health problem. Prevailing inadequate efficacy of seasonal vaccines is a crucial bottleneck. Understanding the immunological and molecular mechanisms underpinning differential influenza vaccine responsiveness is essential to improve vaccination strategies. Here we show comprehensive characterization of the immune response of randomly selected elderly participants (≥ 65 years), immunized with the adjuvanted influenza vaccine Fluad. In-depth analyses by serology, multi-parametric flow cytometry, multiplex and transcriptome analysis, coupled to bioinformatics and mathematical modelling, reveal distinguishing immunological and molecular features between responders and non-responders defined by vaccine-induced seroconversion. Non-responders are specifically characterized by multiple suppressive immune mechanisms. The generated comprehensive high dimensional dataset enables the identification of putative mechanisms and nodes responsible for vaccine non-responsiveness independently of confounding age-related effects, with the potential to facilitate development of tailored vaccination strategies for the elderly.
Collapse
|
17
|
Huda MN, Nurunnabi M. Potential Application of Exosomes in Vaccine Development and Delivery. Pharm Res 2022; 39:2635-2671. [PMID: 35028802 PMCID: PMC8757927 DOI: 10.1007/s11095-021-03143-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023]
Abstract
Exosomes are cell-derived components composed of proteins, lipid, genetic information, cytokines, and growth factors. They play a vital role in immune modulation, cell-cell communication, and response to inflammation. Immune modulation has downstream effects on the regeneration of damaged tissue, promoting survival and repair of damaged resident cells, and promoting the tumor microenvironment via growth factors, antigens, and signaling molecules. On top of carrying biological messengers like mRNAs, miRNAs, fragmented DNA, disease antigens, and proteins, exosomes modulate internal cell environments that promote downstream cell signaling pathways to facilitate different disease progression and induce anti-tumoral effects. In this review, we have summarized how vaccines modulate our immune response in the context of cancer and infectious diseases and the potential of exosomes as vaccine delivery vehicles. Both pre-clinical and clinical studies show that exosomes play a decisive role in processes like angiogenesis, prognosis, tumor growth metastasis, stromal cell activation, intercellular communication, maintaining cellular and systematic homeostasis, and antigen-specific T- and B cell responses. This critical review summarizes the advancement of exosome based vaccine development and delivery, and this comprehensive review can be used as a valuable reference for the broader delivery science community.
Collapse
Affiliation(s)
- Md Nurul Huda
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
18
|
Asmani F, Khavari-Nejad RA, Salmanian AH, Amani J. Immunological evaluation of recombinant chimeric construct from Enterotoxigenic E. coli expressed in hairy roots. Mol Immunol 2022; 147:81-89. [DOI: 10.1016/j.molimm.2022.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/26/2022] [Accepted: 02/09/2022] [Indexed: 01/05/2023]
|
19
|
Silveira CGT, Magnani DM, Costa PR, Avelino-Silva VI, Ricciardi MJ, Timenetsky MDCST, Goulart R, Correia CA, Marmorato MP, Ferrari L, Nakagawa ZB, Tomiyama C, Tomiyama H, Kalil J, Palacios R, Precioso AR, Watkins DI, Kallás EG. Plasmablast Expansion Following the Tetravalent, Live-Attenuated Dengue Vaccine Butantan-DV in DENV-Naïve and DENV-Exposed Individuals in a Brazilian Cohort. Front Immunol 2022; 13:908398. [PMID: 35837409 PMCID: PMC9274664 DOI: 10.3389/fimmu.2022.908398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
An effective vaccine against the dengue virus (DENV) should induce a balanced, long-lasting antibody (Ab) response against all four viral serotypes. The burst of plasmablasts in the peripheral blood after vaccination may reflect enriched vaccine-specific Ab secreting cells. Here we characterize the acute plasmablast responses from naïve and DENV-exposed individuals following immunization with the live attenuated tetravalent (LAT) Butantan DENV vaccine (Butantan-DV). The frequency of circulating plasmablasts was determined by flow cytometric analysis of fresh whole blood specimens collected from 40 participants enrolled in the Phase II Butantan-DV clinical trial (NCT01696422) before and after (days 6, 12, 15 and 22) vaccination. We observed a peak in the number of circulating plasmablast at day 15 after vaccination in both the DENV naïve and the DENV-exposed vaccinees. DENV-exposed vaccinees experienced a significantly higher plasmablast expansion. In the DENV-naïve vaccinees, plasmablasts persisted for approximately three weeks longer than among DENV-exposed volunteers. Our findings indicate that the Butantan-DV can induce plasmablast responses in both DENV-naïve and DENV-exposed individuals and demonstrate the influence of pre-existing DENV immunity on Butantan DV-induced B-cell responses.
Collapse
Affiliation(s)
- Cássia G. T. Silveira
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Diogo M. Magnani
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Priscilla R. Costa
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Vivian I. Avelino-Silva
- Department of Infectious and Parasitic Diseases, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Michael J. Ricciardi
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Raphaella Goulart
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Carolina A. Correia
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mariana P. Marmorato
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Lilian Ferrari
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Zelinda B. Nakagawa
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Claudia Tomiyama
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Helena Tomiyama
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Ricardo Palacios
- Division of Clinical Trials and Pharmacovigilance, Instituto Butantan, São Paulo, Brazil
| | - Alexander R. Precioso
- Division of Clinical Trials and Pharmacovigilance, Instituto Butantan, São Paulo, Brazil
- Pediatrics Department, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - David I. Watkins
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Esper G. Kallás
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Infectious and Parasitic Diseases, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Tabor JB, McCrea MA, Meier TB, Emery CA, Debert CT. Hiding in Plain Sight: Factors Influencing the Neuroinflammatory Response to Sport-Related Concussion. Neurotrauma Rep 2022; 3:200-206. [PMID: 35734393 PMCID: PMC9153987 DOI: 10.1089/neur.2021.0081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Sport-related concussion (SRC) is a major concern among athletes and clinicians around the world. Research into fluid biomarkers of SRC has made significant progress in understanding the complex underlying pathophysiology of concussion. However, little headway has been made toward clinically validating any biomarkers to improve the clinical management of SRC. A major obstacle toward clinical translation of any fluid biomarker is the heterogeneity of SRC overlapping with multiple physiological systems involved in pathology and recovery. Neuroinflammation post-SRC is one such system that may confound fluid biomarker data on many fronts. Neuroinflammatory processes consist of cell mediators, both within the central nervous system and the periphery, that play vital roles in regulating the response to brain injury. Further, neuroinflammation is influenced by many biopsychosocial variables present in most athletic populations. In this commentary, we propose that future fluid biomarker research should take a systems biology approach in the context of the neuroinflammatory response to SRC. We highlight how biological variables, such as age, sex, immune challenges, and hypothalamic-pituitary-adrenal (HPA)-axis responses to stress, may alter neuroinflammation. Further, we underscore the importance of accounting for health and lifestyle variables, such as diet, exercise, sleep, and pre-morbid medical factors, when measuring inflammatory markers of SRC. To successfully move toward clinical translation, fluid biomarker research should take a more holistic approach in study design and data interpretation, collecting information on hidden variables that may be influencing the neuroinflammatory response to SRC.
Collapse
Affiliation(s)
- Jason B. Tabor
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Michael A. McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Carolyn A. Emery
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Chantel T. Debert
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
21
|
Lu M, Su M, Liu N, Zhang J. Effects of environmental salinity on the immune response of the coastal fish Scatophagus argus during bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2022; 124:401-410. [PMID: 35472400 DOI: 10.1016/j.fsi.2022.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
The coastal aquaculture is characterized with environmental salinity fluctuation, and the effects of salinity stress on the immunity of cultured fish are needed to be further explored. Scatophagus argus is an important species in the wild fisheries and aquaculture industry, it would be of great value to reveal the impact of salinity change on the immune response in this species. Understanding the effects of salinity stress on immune response can provide valuable insights into salinity management in the aquacultural process. The head kidney, which is an organ unique for teleost fish, functions not only as a central immune organ but also as a crucial role in the stress response during which the secretion of immunoregulatory molecules i.e. cytokines is facilitated. In the present study, Individuals of S. argus acclimated to 3 different salinities [0‰ (FW), 10‰ (BW), and 25‰ (SW)] were injected intraperitoneally with A. hydrophila, and then monitored throughout one week. The effects of environmental salinity on the immune response in S. argus stimulated by A. hydrophila infection were investigated. mRNA expression profiles of cytokine genes IL-1β, IL-6, IL-10 and TNF-α in different salinity groups was quite different. mRNA expression of cytokine genes in BW group and SW group rose more quickly and significantly higher than FW group (p < 0.05) at early stages (6-24 hpi) after bacterial injection, and before 96 hpi, the highest value of cytokine expression at each time point was recorded in SW group. Immune parameters such as lysozyme level, complement C3 activity and IgM content in BW and FW groups were lower than SW group at each time point from 24 to 144 hpi after bacterial injection. In addition, leukocyte profiles in the head kidney and blood were also investigated. Although hypoosmotic acclimation could temporarily stimulate monocyte and neutrophil proliferation, it was observed that the number of monocytes, neutrophils and lymphocytes of the head kidney and blood in SW group increased more quickly than BW and FW groups after bacterial infection. Our results indicate that hypoosmotic stress due to the decrease of environmental salinity has suppressive immunoregulatory effects on the immune response of S. argus.
Collapse
Affiliation(s)
- Mengying Lu
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Maoliang Su
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Nanxi Liu
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Junbin Zhang
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
22
|
Otoo JA, Schlappi TS. REASSURED Multiplex Diagnostics: A Critical Review and Forecast. BIOSENSORS 2022; 12:bios12020124. [PMID: 35200384 PMCID: PMC8869588 DOI: 10.3390/bios12020124] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 05/05/2023]
Abstract
The diagnosis of infectious diseases is ineffective when the diagnostic test does not meet one or more of the necessary standards of affordability, accessibility, and accuracy. The World Health Organization further clarifies these standards with a set of criteria that has the acronym ASSURED (Affordable, Sensitive, Specific, User-friendly, Rapid and robust, Equipment-free and Deliverable to end-users). The advancement of the digital age has led to a revision of the ASSURED criteria to REASSURED: Real-time connectivity, Ease of specimen collection, Affordable, Sensitive, Specific, User-friendly, Rapid and robust, Equipment-free or simple, and Deliverable to end-users. Many diagnostic tests have been developed that aim to satisfy the REASSURED criteria; however, most of them only detect a single target. With the progression of syndromic infections, coinfections and the current antimicrobial resistance challenges, the need for multiplexed diagnostics is now more important than ever. This review summarizes current diagnostic technologies for multiplexed detection and forecasts which methods have promise for detecting multiple targets and meeting all REASSURED criteria.
Collapse
|
23
|
Seiffert P, Konka A, Kasperczyk J, Kawa J, Lejawa M, Maślanka-Seiffert B, Zembala-John J, Bugdol M, Romanik M, Bułdak R, Marcisz C, Derejczyk J, Religa D. Immunogenicity of the BNT162b2 mRNA COVID-19 vaccine in older residents of a long-term care facility: relation with age, frailty and prior infection status. Biogerontology 2022; 23:53-64. [PMID: 34923608 PMCID: PMC8684786 DOI: 10.1007/s10522-021-09944-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 11/11/2021] [Indexed: 11/29/2022]
Abstract
Clinical and biological assessment of the COVID-19 vaccine efficacy in the frail population is of crucial importance. The study focuses on measuring the levels of anti-SARS-CoV-2 IgG antibodies before and after BNT162b2 mRNA COVID-19 vaccination among long-term care facility (LTCF) elderly residents. We conducted a prospective, single-center, observational study among LTCF residents. The study protocol was based on three blood sample acquisitions: first taken at baseline-5 days before the first dose of the vaccine, second-20 days after the first dose, and third-12 days after the second shot of the vaccine. The comparison was made for two cohorts: patients with and without prior COVID-19 infection. The data was collected from January to March 2021. A total number of 78 LTCF residents (55 women and 23 men) aged 62-104, 85.72 ± 7.59 years (mean ± SD), were enrolled in the study. All study participants were investigated for the presence of SARS-CoV-2 anti-spike (S) protein IgG, using a chemiluminescent immunoassay. Frailty was assessed with the Clinical Frailty Scale. Among elderly COVID-19 survivors in LTCF, a single dose of vaccine significantly increased anti-SARS-CoV-2 IgG antibody levels. IgG concentration after a single and double dose was comparable, which may suggest that elderly COVID-19 survivors do not require a second dose of vaccine. For residents without a previous history of COVID-19, two doses are needed to achieve an effective serological response. The level of anti-SARS-CoV-2 IgG antibodies after vaccination with BNT162b2 mRNA COVID-19 did not correlate with the frailty and age of the studied individuals.
Collapse
Affiliation(s)
- Piotr Seiffert
- Department of Gerontology and Geriatric Nursing, School of Health Sciences, Medical University of Silesia, Katowice, Poland
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Rheumatology Ward, Murcki Hospital, Katowice, Poland
| | - Adam Konka
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Janusz Kasperczyk
- Chair and Department of Medicine and Environmental Epidemiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Jacek Kawa
- Faculty of Biomedical Engineering, Silesian University of Technology, Zabrze, Poland
| | - Mateusz Lejawa
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | | | - Joanna Zembala-John
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Chair and Department of Medicine and Environmental Epidemiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
- Silesian Center for Heart Diseases, Zabrze, Poland
| | - Monika Bugdol
- Faculty of Biomedical Engineering, Silesian University of Technology, Zabrze, Poland
| | - Małgorzata Romanik
- Department of Medical Microbiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Rafał Bułdak
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Opole, Poland
| | - Czesław Marcisz
- Department of Gerontology and Geriatric Nursing, School of Health Sciences, Medical University of Silesia, Katowice, Poland
| | | | - Dorota Religa
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
24
|
Antanasijevic A, Bowman CA, Kirchdoerfer RN, Cottrell CA, Ozorowski G, Upadhyay AA, Cirelli KM, Carnathan DG, Enemuo CA, Sewall LM, Nogal B, Zhao F, Groschel B, Schief WR, Sok D, Silvestri G, Crotty S, Bosinger SE, Ward AB. From structure to sequence: Antibody discovery using cryoEM. SCIENCE ADVANCES 2022; 8:eabk2039. [PMID: 35044813 PMCID: PMC8769551 DOI: 10.1126/sciadv.abk2039] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/23/2021] [Indexed: 06/02/2023]
Abstract
One of the rate-limiting steps in analyzing immune responses to vaccines or infections is the isolation and characterization of monoclonal antibodies. Here, we present a hybrid structural and bioinformatic approach to directly assign the heavy and light chains, identify complementarity-determining regions, and discover sequences from cryoEM density maps of serum-derived polyclonal antibodies bound to an antigen. When combined with next-generation sequencing of immune repertoires, we were able to specifically identify clonal family members, synthesize the monoclonal antibodies, and confirm that they interact with the antigen in a manner equivalent to the corresponding polyclonal antibodies. This structure-based approach for identification of monoclonal antibodies from polyclonal sera opens new avenues for analysis of immune responses and iterative vaccine design.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Charles A. Bowman
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robert N. Kirchdoerfer
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Christopher A. Cottrell
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Amit A. Upadhyay
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30329, USA
- Yerkes Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Yerkes Nonhuman Primate Genomics Core, Emory University, Atlanta, GA 30329, USA
| | - Kimberly M. Cirelli
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Diane G. Carnathan
- Yerkes Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Yerkes Nonhuman Primate Genomics Core, Emory University, Atlanta, GA 30329, USA
| | - Chiamaka A. Enemuo
- Yerkes Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Yerkes Nonhuman Primate Genomics Core, Emory University, Atlanta, GA 30329, USA
| | - Leigh M. Sewall
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bartek Nogal
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Fangzhu Zhao
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- International AIDS Vaccine Initiative–Neutralizing Antibody Center (IAVI-NAC), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bettina Groschel
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - William R. Schief
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Devin Sok
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- International AIDS Vaccine Initiative–Neutralizing Antibody Center (IAVI-NAC), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Guido Silvestri
- Yerkes Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Yerkes Nonhuman Primate Genomics Core, Emory University, Atlanta, GA 30329, USA
| | - Shane Crotty
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Steven E. Bosinger
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30329, USA
- Yerkes Division of Microbiology and Immunology, Yerkes National Primate Research Center, and Yerkes Nonhuman Primate Genomics Core, Emory University, Atlanta, GA 30329, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
25
|
Analysis of TCR Repertoire by High-Throughput Sequencing Indicates the Feature of T Cell Immune Response after SARS-CoV-2 Infection. Cells 2021; 11:cells11010068. [PMID: 35011632 PMCID: PMC8750083 DOI: 10.3390/cells11010068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/13/2021] [Accepted: 12/24/2021] [Indexed: 01/10/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global infectious disease caused by the SARS-CoV-2 coronavirus. T cells play an essential role in the body’s fighting against the virus invasion, and the T cell receptor (TCR) is crucial in T cell-mediated virus recognition and clearance. However, little has been known about the features of T cell response in convalescent COVID-19 patients. In this study, using 5′RACE technology and PacBio sequencing, we analyzed the TCR repertoire of COVID-19 patients after recovery for 2 weeks and 6 months compared with the healthy donors. The TCR clustering and CDR3 annotation were exploited to discover groups of patient-specific TCR clonotypes with potential SARS-CoV-2 antigen specificities. We first identified CD4+ and CD8+ T cell clones with certain clonal expansion after infection, and then observed the preferential recombination usage of V(D) J gene segments in CD4+ and CD8+ T cells of COVID-19 patients with different convalescent stages. More important, the TRBV6-5-TRBD2-TRBJ2-7 combination with high frequency was shared between CD4+ T and CD8+ T cells of different COVID-19 patients. Finally, we found the dominant characteristic motifs of the CDR3 sequence between recovered COVID-19 and healthy control. Our study provides novel insights on TCR in COVID-19 with different convalescent phases, contributing to our understanding of the immune response induced by SARS-CoV-2.
Collapse
|
26
|
Garcia-Pintos C, Riet-Correa F, Menchaca A. Effect of Foot-and-Mouth Disease Vaccine on Pregnancy Failure in Beef Cows. Front Vet Sci 2021; 8:761304. [PMID: 34869734 PMCID: PMC8633299 DOI: 10.3389/fvets.2021.761304] [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: 08/19/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
This study evaluates whether the foot-and-mouth disease (FMD) vaccination increases pregnancy failures in Bos taurus beef cows. A total of 3,379 cows were assigned to two experimental groups to receive (n = 1,722) or not receive (n = 1,657) a FMD vaccine (commercial preparation containing FMD virus, O1 Campos and A24 Cruzeiro) at different gestational age. Pregnancy diagnosis was performed by ultrasonography at vaccination time (Day 0), and the cows were classified by days of pregnancy as follows: (a) <29 days after mating (presumed pregnant cows, n = 778), (b) between 30 and 44 days of pregnancy (n = 1,100), (c) 45 and 59 days of pregnancy (n = 553), and (d) between 60 and 90 days of pregnancy (n = 948). Pregnancy failure was determined 30 days after vaccination by a second ultrasound examination. Cows that were vaccinated within 29 days after mating had a 7.8% greater pregnancy failure rate than non-vaccinated cows (44.1%, 163/370 vs. 36.3%, 148/408, respectively; P <0.05). Cows vaccinated between 30 and 44 days of gestation had a pregnancy failure rate greater than non-vaccinated cows (4.9%, 28/576 vs. 2.5%, 13/524, respectively; P <0.05). When cows received the vaccine between days 45 and 90 of gestation no differences in pregnancy failure were observed (0.8%, 6/776 vs. 1.2%, 9/725, respectively; P = NS). Body temperature and local adverse reactions to vaccine inoculation were recorded in a subset of 152 multiparous cows. Hyperthermia (>39.5°C) was detected on Day 1 or 2 in 28.0% (21/75) of vaccinated vs. 7.8% (6/77) of non-vaccinated cows (P <0.01). Local adverse reaction to the FMD vaccine inoculation increased from 0.0% (0/75) on Day 0, to 15.7% (11/75) on Day 4, and 38.7% (29/75) on Day 10 (P <0.01). On Day 30 local reaction was detected in 10.5% (34/323) and fell to 2.2% on Day 60 (7/323) post vaccination (P <0.01). In conclusion, FMD vaccine increases pregnancy failure when it is administered before 45 days of gestation, an effect that was associated with hyperthermia and local adverse reaction. No effect on pregnancy failure was found when vaccination was performed after 45 days of gestation.
Collapse
Affiliation(s)
- Camila Garcia-Pintos
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Montevideo, Uruguay.,Plataforma de Salud Animal, Instituto Nacional de Investigación Agropecuaria, Montevideo, Uruguay
| | - Franklin Riet-Correa
- Plataforma de Salud Animal, Instituto Nacional de Investigación Agropecuaria, Montevideo, Uruguay.,Programa de Pós-graduação em Ciência Animal nos Trópicos, Escola de Medicina Veterinária e Zootecnia, Universidade Federal da Bahia, Salvador, Brazil
| | - Alejo Menchaca
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Montevideo, Uruguay.,Plataforma de Salud Animal, Instituto Nacional de Investigación Agropecuaria, Montevideo, Uruguay
| |
Collapse
|
27
|
Long Term Immune Response Produced by the SputnikV Vaccine. Int J Mol Sci 2021; 22:ijms222011211. [PMID: 34681885 PMCID: PMC8537212 DOI: 10.3390/ijms222011211] [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] [Received: 08/31/2021] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023] Open
Abstract
SputnikV is a vaccine against SARS-CoV-2 developed by the Gamaleya National Research Centre for Epidemiology and Microbiology. The vaccine has been shown to induce both humoral and cellular immune responses, yet the mechanisms remain largely unknown. Forty SputnikV vaccinated individuals were included in this study which aimed to demonstrate the location of immunogenic domains of the SARS-CoV-2 S protein using an overlapping peptide library. Additionally, cytokines in the serum of vaccinated and convalescent COVID-19 patients were analyzed. We have found antibodies from both vaccinated and convalescent sera bind to immunogenic regions located in multiple domains of SARS-CoV-2 S protein, including Receptor Binding Domain (RBD), N-terminal Domain (NTD), Fusion Protein (FP) and Heptad Repeats (HRs). Interestingly, many peptides were recognized by immunized and convalescent serum antibodies and correspond to conserved regions in circulating variants of SARS-CoV-2. This breadth of reactivity was still evident 90 days after the first dose of the vaccine, showing that the vaccine has induced a prolonged response. As evidenced by the activation of T cells, cellular immunity strongly suggests the high potency of the SputnikV vaccine against SARS-CoV-2 infection.
Collapse
|
28
|
Rose J, McCullough PA. WITHDRAWN: A Report on Myocarditis Adverse Events in the U.S. Vaccine Adverse Events Reporting System (VAERS) in Association with COVID-19 Injectable Biological Products. Curr Probl Cardiol 2021:101011. [PMID: 34601006 PMCID: PMC8483988 DOI: 10.1016/j.cpcardiol.2021.101011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022]
Abstract
The Publisher regrets that this article has been temporarily removed. A replacement will appear as soon as possible in which the reason for the removal of the article will be specified, or the article will be reinstated.
Collapse
Affiliation(s)
- Jessica Rose
- Institute of Pure and Applied Knowledge, Public Health Policy Initiative (PHPI), Texas A & M College of Medicine, Baylor Dallas Campus, Dallas TX USA.
| | - Peter A McCullough
- Institute of Pure and Applied Knowledge, Public Health Policy Initiative (PHPI), Texas A & M College of Medicine, Baylor Dallas Campus, Dallas TX USA
| |
Collapse
|
29
|
Motta F, Selmi C, De Santis M. 5 reasons to encourage anti-SARS-CoV-2 vaccination in patients with rheumatic diseases. Expert Rev Clin Immunol 2021; 17:1241-1244. [PMID: 34499006 DOI: 10.1080/1744666x.2021.1978289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Francesca Motta
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center- IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center- IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center- IRCCS, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
30
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
31
|
Van Tilbeurgh M, Lemdani K, Beignon AS, Chapon C, Tchitchek N, Cheraitia L, Marcos Lopez E, Pascal Q, Le Grand R, Maisonnasse P, Manet C. Predictive Markers of Immunogenicity and Efficacy for Human Vaccines. Vaccines (Basel) 2021; 9:579. [PMID: 34205932 PMCID: PMC8226531 DOI: 10.3390/vaccines9060579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines represent one of the major advances of modern medicine. Despite the many successes of vaccination, continuous efforts to design new vaccines are needed to fight "old" pandemics, such as tuberculosis and malaria, as well as emerging pathogens, such as Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination aims at reaching sterilizing immunity, however assessing vaccine efficacy is still challenging and underscores the need for a better understanding of immune protective responses. Identifying reliable predictive markers of immunogenicity can help to select and develop promising vaccine candidates during early preclinical studies and can lead to improved, personalized, vaccination strategies. A systems biology approach is increasingly being adopted to address these major challenges using multiple high-dimensional technologies combined with in silico models. Although the goal is to develop predictive models of vaccine efficacy in humans, applying this approach to animal models empowers basic and translational vaccine research. In this review, we provide an overview of vaccine immune signatures in preclinical models, as well as in target human populations. We also discuss high-throughput technologies used to probe vaccine-induced responses, along with data analysis and computational methodologies applied to the predictive modeling of vaccine efficacy.
Collapse
Affiliation(s)
- Matthieu Van Tilbeurgh
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Katia Lemdani
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Nicolas Tchitchek
- Unité de Recherche i3, Inserm UMR-S 959, Bâtiment CERVI, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Lina Cheraitia
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Ernesto Marcos Lopez
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Quentin Pascal
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Pauline Maisonnasse
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Caroline Manet
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| |
Collapse
|
32
|
Adam L, Rosenbaum P, Bonduelle O, Combadière B. Strategies for Immunomonitoring after Vaccination and during Infection. Vaccines (Basel) 2021; 9:365. [PMID: 33918841 PMCID: PMC8070333 DOI: 10.3390/vaccines9040365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
Immunomonitoring is the study of an individual's immune responses over the course of vaccination or infection. In the infectious context, exploring the innate and adaptive immune responses will help to investigate their contribution to viral control or toxicity. After vaccination, immunomonitoring of the correlate(s) and surrogate(s) of protection is a major asset for measuring vaccine immune efficacy. Conventional immunomonitoring methods include antibody-based technologies that are easy to use. However, promising sensitive high-throughput technologies allowed the emergence of holistic approaches. This raises the question of data integration methods and tools. These approaches allow us to increase our knowledge on immune mechanisms as well as the identification of key effectors of the immune response. However, the depiction of relevant findings requires a well-rounded consideration beforehand about the hypotheses, conception, organization and objectives of the immunomonitoring. Therefore, well-standardized and comprehensive studies fuel insight to design more efficient, rationale-based vaccines and therapeutics to fight against infectious diseases. Hence, we will illustrate this review with examples of the immunomonitoring approaches used during vaccination and the COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | - Behazine Combadière
- Inserm, Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, 75013 Paris, France; (L.A.); (P.R.); (O.B.)
| |
Collapse
|
33
|
Zhou Y, Yu Z, Wang X, Chen W, Liu Y, Zhang Y, Yin J, Han S. Exosomal circRNAs contribute to intestinal development via the VEGF signalling pathway in human term and preterm colostrum. Aging (Albany NY) 2021; 13:11218-11233. [PMID: 33820870 PMCID: PMC8109075 DOI: 10.18632/aging.202806] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 02/08/2021] [Indexed: 12/21/2022]
Abstract
Human breast milk (HBM) provides essential nutrients for newborn growth and development, and contains a variety of biologically active ingredients that can affect gastrointestinal tract and immune system development in breastfed infants. HBM also contains mRNAs, microRNAs and lncRNAs, most of which are encapsulated in milk-derived exosomes and exhibit various important infant development related biological functions. While previous studies have shown that exosomal circRNAs are involved in the intestinal epithelial cells’ proliferation and repair. However, the effect of HBM exosomal circRNAs on intestinal development is not clear. In this study, we identified 6756 circRNAs both in preterm colostrum (PC) and term colostrum (TC), of which 66 were upregulated, and 42 were downregulated (|fold change>2|, p < 0.05) in PC. Pathway analysis showed that the VEGF signalling pathway was involved, and network analysis revealed that the differentially expressed circRNAs bound various miRNAs. Further analyses showed that has_circRNA_405708 and has_circRNA_104707 were involved in the VEGF signalling pathway, and that they all bound various mirRNAs. Exosomes found in preterm colostrum (PC) and term colostrum (TC) promoted VEGF protein expression and induced the proliferation and migration of small intestinal epithelial cells (FHCs). Exosomal circRNAs found in human colostrum (HC) binding to related miRNAs may regulate VEGF signalling, and intestinal development.
Collapse
Affiliation(s)
- Yahui Zhou
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China.,Department of Pediatrics, Fourth Clinical Medicine College, Nanjing Medical University, Nanjing 210029, China
| | - Zhangbin Yu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Xingyun Wang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Wenjuan Chen
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yiwen Liu
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Yinghui Zhang
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Jing Yin
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Shuping Han
- Department of Pediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| |
Collapse
|
34
|
Aldakak L, Huber VM, Rühli F, Bender N. Sex difference in the immunogenicity of the quadrivalent Human Papilloma Virus vaccine: Systematic review and meta-analysis. Vaccine 2021; 39:1680-1686. [PMID: 33637386 DOI: 10.1016/j.vaccine.2021.02.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 12/02/2020] [Accepted: 02/09/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Immunological differences between males and females in response to viral vaccines are well known. This the first review to examine them for the Human Papilloma Virus. METHODS We conducted a systematic review and meta-analysis of the immunogenicity of the Quadrivalent Human Papilloma Virus Vaccine qHPVV. We searched Medline, Embase, and CENTRAL for trials published until September 17, 2019. Inclusion criteria were 3-doses and reporting geometric mean titers (GMTs). We performed random-effects meta-analyses and meta-regression separated by age group and sex. RESULTS Our search yielded 1809 unique studies. 334 full texts were screened and data from 18 studies were extracted. Females had higher pooled geometric mean titers than males in all age groups. Log transformed GMTs in male children (<16) years were: against HPV6: 6·62 (95% CI 6·29-6·94; I2 = 86·0%), against HPV11: 7·07 (95% CI 6·90-7·23; I2 = 63.1%), against HPV16: 8·53 (95% CI 8·28-8·78; I2 = 73·0%), and against HPV18 7·21 (95% CI 7·08-7·34; I2 = 26·4%). In females: against HPV6 7·10 (95% CI 6·79-7·41; I2 = 96·6%), HPV11: 7·32 (95% CI 7·15-7·50; I2 = 90·6%), HPV16: 8·71 (95% CI 8·52-8·91; I2 = 90·2%), and HPV18 7·35 (95% CI 7·11-7·58; I2 = 92·7%). In the meta-regression, the sexual difference was significant for HPV6 (p = 0·022) with a similar tendency for HPV11 (p = 0·066) and HPV18 (p = 0·079). Immunogenicity was significantly higher in children (<16) than in adults (p < 0·001). CONCLUSION Females have higher antibody titers against HPV after receiving the qHPVV than do males. The difference is bigger in low-risk HPV strains. Adjusting the doses and schedules for each sex should be explored further.
Collapse
Affiliation(s)
- Lafi Aldakak
- Institute of Evolutionary Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Vera Maria Huber
- Institute of Evolutionary Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Frank Rühli
- Institute of Evolutionary Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Nicole Bender
- Institute of Evolutionary Medicine, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
35
|
De Marco Castro E, Calder PC, Roche HM. β-1,3/1,6-Glucans and Immunity: State of the Art and Future Directions. Mol Nutr Food Res 2021; 65:e1901071. [PMID: 32223047 PMCID: PMC7816268 DOI: 10.1002/mnfr.201901071] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/28/2020] [Indexed: 12/16/2022]
Abstract
The innate immune system responds in a rapid and non-specific manner against immunologic threats; inflammation is part of this response. This is followed by a slower but targeted and specific response termed the adaptive or acquired immune response. There is emerging evidence that dietary components, including yeast-derived β-glucans, can aid host defense against pathogens by modulating inflammatory and antimicrobial activity of neutrophils and macrophages. Innate immune training refers to a newly recognized phenomenon wherein compounds may "train" innate immune cells, such that monocyte and macrophage precursor biology is altered to mount a more effective immunological response. Although various human studies have been carried out, much uncertainty still exists and further studies are required to fully elucidate the relationship between β-glucan supplementation and human immune function. This review offers an up-to-date report on yeast-derived β-glucans as immunomodulators, including a brief overview of the current paradigm regarding the interaction of β-glucans with the immune system. The recent pre-clinical work that has partly decrypted mode of action and the newest evidence from human trials are also reviewed. According to pre-clinical studies, β-1,3/1,6-glucan derived from baker's yeast may offer increased immuno-surveillance, although the human evidence is weaker than that gained from pre-clinical studies.
Collapse
Affiliation(s)
- Elena De Marco Castro
- Nutrigenomics Research GroupSchool of Public Health, Physiotherapy, and Sports ScienceConway Institute, and Institute of Food and HealthUniversity College DublinDublin 4D04 V1W8Ireland
- Diabetes Complications Research CentreConway InstituteUniversity College DublinDublin 4D04 V1W8Ireland
| | - Philip C. Calder
- Faculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- NIHR Southampton Biomedical Research CentreUniversity Hospital Southampton NHS Foundation TrustUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Helen M. Roche
- Nutrigenomics Research GroupSchool of Public Health, Physiotherapy, and Sports ScienceConway Institute, and Institute of Food and HealthUniversity College DublinDublin 4D04 V1W8Ireland
- Diabetes Complications Research CentreConway InstituteUniversity College DublinDublin 4D04 V1W8Ireland
- Institute for Global Food SecurityQueens University BelfastBelfastNorthern IrelandBT9 5DLUK
| |
Collapse
|
36
|
Domachowske J. The Immune Response to Infection. Vaccines (Basel) 2021. [DOI: 10.1007/978-3-030-58414-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
37
|
Cao Y, Qiu Y, Tu G, Yang C. Single-cell RNA Sequencing in Immunology. Curr Genomics 2020; 21:564-575. [PMID: 33414678 PMCID: PMC7770633 DOI: 10.2174/1389202921999201020203249] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
The complex immune system is involved in multiple pathological processes. Single-cell RNA sequencing (scRNA-seq) is able to analyze complex cell mixtures correct to a single cell and single molecule, thus is qualified to analyze immune reactions in several diseases. In recent years, scRNA-seq has been applied in many researching fields and has presented many innovative results. In this review, we intend to provide an overview of single-cell RNA sequencing applications in immunology and a prospect of future directions.
Collapse
Affiliation(s)
| | | | - Guowei Tu
- Address correspondence to these authors at the Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Tel: +86-21-64041990; E-mails: ; and Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Tel: +86-21-64041990;, E-mail:
| | - Cheng Yang
- Address correspondence to these authors at the Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Tel: +86-21-64041990; E-mails: ; and Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Tel: +86-21-64041990;, E-mail:
| |
Collapse
|
38
|
Dimond A, Van de Pette M, Fisher AG. Illuminating Epigenetics and Inheritance in the Immune System with Bioluminescence. Trends Immunol 2020; 41:994-1005. [PMID: 33036908 DOI: 10.1016/j.it.2020.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022]
Abstract
The remarkable process of light emission by living organisms has fascinated mankind for thousands of years. A recent expansion in the repertoire of catalytic luciferase enzymes, coupled with the discovery of the genes and pathways that encode different luciferin substrates, means that bioluminescence imaging (BLI) is set to revolutionize longitudinal and dynamic studies of gene control within biomedicine, including the regulation of immune responses. In this review article, we summarize recent advances in bioluminescence-based imaging approaches that promise to enlighten our understanding of in vivo gene and epigenetic control within the immune system.
Collapse
Affiliation(s)
- Andrew Dimond
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mathew Van de Pette
- Epigenetic Mechanisms of Toxicity, MRC Toxicology Unit, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Amanda G Fisher
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
39
|
Jacobsen-Pereira CH, Cardoso CC, Gehlen TC, Regina Dos Santos C, Santos-Silva MC. Immune response of Brazilian farmers exposed to multiple pesticides. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 202:110912. [PMID: 32800247 DOI: 10.1016/j.ecoenv.2020.110912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 06/11/2023]
Abstract
Occupational exposure to pesticides has been identified as a factor that predisposes to disorders of the immune system. Immunosuppression, autoimmunity, cancer of various organs and other diseases in people who apply these products have been reported by the studies. This study aimed to investigate the relationship between occupational exposure to pesticides and the immunological profile in 43 farmers exposed to mixtures of pesticides for at least 15 years. A control group composed of 30 individuals without a history of occupational exposure to pesticides was also evaluated. Peripheral blood samples were processed by flow cytometry and cells were labelled with an 8-color monoclonal antibody panel. Plasma cytokines were also measured. Significant increase in classical monocytes (p < 0.001) and dendritic cells (p < 0.001) in the exposed group was observed as well in total T cells (p = 0.04), central memory CD8 T cells (p = 0.02) and effector memory CD8 T cells (p = 0.01). On the other hand, the activation markers of T cells as the expression of CD57, HLA-DR, CD25 and CD28 were evaluated and no difference was found between groups. When the B cells were analyzed, a significant decrease in total B cells (p = 0.01), regulatory B cells (p < 0.001) and plasmablasts (p < 0.001) in the exposed group, compared to healthy controls, was observed. Pro-inflammatory IL-6 was significantly elevated (p = 0.04) in the plasma of farmers compared to that of controls. The constant antigenic stimulus that occurs during exposure to pesticides can favor the recruitment of dendritic cells and macrophages (APCs) presents in the skin and respiratory tract. In the secondary lymphoid organs, the CD4 T and B cells that process such antigens are possibly undergoing proliferative exhaustion, with the consequent depletion of all mature B subpopulations. The resulting drop in humoral immunity may be offset by an increase in the number of circulating CD8 T lymphocytes due to their cytotoxic action.
Collapse
Affiliation(s)
| | - Chandra Chiappin Cardoso
- Postgraduate Program in Pharmacy of the Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil; Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil
| | - Tais Cristina Gehlen
- Laboratory of Toxicology, University Hospital of the Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil
| | - Claudia Regina Dos Santos
- Laboratory of Toxicology, University Hospital of the Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil; Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil
| | - Maria Claudia Santos-Silva
- Division of Clinical Analysis, Flow Cytometry Service, University Hospital of the Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil; Clinical Analysis Department, Health Sciences Center, Federal University of Santa Catarina (UFSC), Florianopolis, SC, 88040-900, Brazil.
| |
Collapse
|
40
|
Johnson AS, Fatemi R, Winlow W. SARS-CoV-2 Bound Human Serum Albumin and Systemic Septic Shock. Front Cardiovasc Med 2020; 7:153. [PMID: 33088822 PMCID: PMC7498713 DOI: 10.3389/fcvm.2020.00153] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022] Open
Abstract
The emergence of the COVID-19 virus and the subsequent pandemic have driven a great deal of research activity. The effects of COVID-19 are caused by the severe respiratory syndrome coronavirus 2 (SARS-CoV-2) and it is the underlying actions of SARs-CoV-2 virions on the endothelial glycocalyx that we consider here. One of the key factors in COVID-19 infection is its almost unique age-related profile, with a doubling in mortality every 10 years after the age of 50. The endothelial glycocalyx layer is essential in maintaining normal fluid homeostasis, but is fragile and prone to pathophysiological damage. It is physiologically significant in capillary microcirculation and in fluid distribution to the tissues. Human serum albumin (HSA), the most abundant protein in plasma, is created in the liver which also maintains its concentration, but this reduces by 10-15% after 50 years of age. HSA transports hormones, free fatty acids and maintains oncotic pressure, but SARS-CoV-2 virions bind competitively to HSA diminishing its normal transport function. Furthermore, hypoalbuminemia is frequently observed in patients with such conditions as diabetes, hypertension, and chronic heart failure, i.e., those most vulnerable to SARS-CoV-2 infection. Hypoalbuminemia, coagulopathy, and vascular disease have been linked in COVID-19 and have been shown to predict outcome independent of age and morbidity. Hypoalbuminemia is also known factor in sepsis and Acute respiratory distress syndrome (ARDS) occurs when fluids build-up in the alveoli and it is associated with sepsis, whose mechanism is systemic, being associated with the fluid and logistic mechanisms of the circulation. Glycocalyx damage is associated with changes plasma protein concentration, particularly HSA and blockage of albumin transport can produce the systemic symptoms seen in SARS-CoV-2 infection and sepsis. We therefore conclude that albumin binding to SARS-CoV-2 virions may inhibit the formation of the endothelial glycocalyx by inhibition of albumin transport binding sites. We postulate that albumin therapy to replace bound albumin might alleviate some of the symptoms leading to sepsis and that clinical trials to test this postulation should be initiated as a matter of urgency.
Collapse
Affiliation(s)
- Andrew S. Johnson
- Dipartimento di Biologia, Università Degli Studi di Naples, Federico II, Naples, Italy
| | - Rouholah Fatemi
- Physiology Research Center (PRC), School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - William Winlow
- Dipartimento di Biologia, Università Degli Studi di Naples, Federico II, Naples, Italy
- Institute of Ageing and Chronic Diseases, The Apex Building, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
41
|
Abstract
Active surveillance of invasive group A Streptococcus (iGAS) disease indicates that its incidence in the US general population is low, but limited studies show rates for American Indians and Alaska Natives (AI/AN) are severalfold higher. Major disparities in rates of iGAS exist between Indigenous and non-Indigenous populations of Australia, New Zealand, and Canada, but much less is understood about iGAS among AI/AN in the United States. Although complex host-pathogen interactions influence the rates of iGAS, including strain variation and virulence, the number and type of concurrent conditions, and socioeconomic status, the relative contribution of each remains unclear. We highlight the poor correlation between the substantial effect of iGAS among Indigenous persons in industrialized countries and the current understanding of factors that influence iGAS disease in these populations. Prospective, large-scale, population-based studies of iGAS are needed that include AI/AN as a necessary first step to understanding the effects of iGAS.
Collapse
|
42
|
Franco J, Rajwa B, Ferreira CR, Sundberg JP, HogenEsch H. Lipidomic Profiling of the Epidermis in a Mouse Model of Dermatitis Reveals Sexual Dimorphism and Changes in Lipid Composition before the Onset of Clinical Disease. Metabolites 2020; 10:metabo10070299. [PMID: 32708296 PMCID: PMC7408197 DOI: 10.3390/metabo10070299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023] Open
Abstract
Atopic dermatitis (AD) is a multifactorial disease associated with alterations in lipid composition and organization in the epidermis. Multiple variants of AD exist with different outcomes in response to therapies. The evaluation of disease progression and response to treatment are observational assessments with poor inter-observer agreement highlighting the need for molecular markers. SHARPIN-deficient mice (Sharpincpdm) spontaneously develop chronic proliferative dermatitis with features similar to AD in humans. To study the changes in the epidermal lipid-content during disease progression, we tested 72 epidermis samples from three groups (5-, 7-, and 10-weeks old) of cpdm mice and their WT littermates. An agnostic mass-spectrometry strategy for biomarker discovery termed multiple-reaction monitoring (MRM)-profiling was used to detect and monitor 1,030 lipid ions present in the epidermis samples. In order to select the most relevant ions, we utilized a two-tiered filter/wrapper feature-selection strategy. Lipid categories were compressed, and an elastic-net classifier was used to rank and identify the most predictive lipid categories for sex, phenotype, and disease stages of cpdm mice. The model accurately classified the samples based on phospholipids, cholesteryl esters, acylcarnitines, and sphingolipids, demonstrating that disease progression cannot be defined by one single lipid or lipid category.
Collapse
Affiliation(s)
- Jackeline Franco
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA;
| | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: (B.R.); (H.H.)
| | - Christina R. Ferreira
- Metabolite Profiling Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA;
| | | | - Harm HogenEsch
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute of Inflammation, Immunology and Infectious Diseases, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: (B.R.); (H.H.)
| |
Collapse
|
43
|
Kennedy RB, Ovsyannikova IG, Palese P, Poland GA. Current Challenges in Vaccinology. Front Immunol 2020; 11:1181. [PMID: 32670279 PMCID: PMC7329983 DOI: 10.3389/fimmu.2020.01181] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
The development of vaccines, which prime the immune system to respond to future infections, has led to global declines in morbidity and mortality from dreadful infectious communicable diseases. However, many pathogens of public health importance are highly complex and/or rapidly evolving, posing unique challenges to vaccine development. Several of these challenges include an incomplete understanding of how immunity develops, host and pathogen genetic variability, and an increased societal skepticism regarding vaccine safety. In particular, new high-dimensional omics technologies, aided by bioinformatics, are driving new vaccine development (vaccinomics). Informed by recent insights into pathogen biology, host genetic diversity, and immunology, the increasing use of genomic approaches is leading to new models and understanding of host immune system responses that may provide solutions in the rapid development of novel vaccine candidates.
Collapse
Affiliation(s)
- Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| | - Peter Palese
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
44
|
Bannister S, Messina NL, Novakovic B, Curtis N. The emerging role of epigenetics in the immune response to vaccination and infection: a systematic review. Epigenetics 2020; 15:555-593. [PMID: 31914857 PMCID: PMC7574386 DOI: 10.1080/15592294.2020.1712814] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
Extensive research has highlighted the role of infection-induced epigenetic events in the development of cancer. More recently, attention has focused on the ability of non-carcinogenic infections, as well as vaccines, to modify the human epigenome and modulate the immune response. This review explores this rapidly evolving area of investigation and outlines the many and varied ways in which vaccination and natural infection can influence the human epigenome from modulation of the innate and adaptive immune response, to biological ageing and modification of disease risk. The implications of these epigenetic changes on immune regulation and their potential application to the diagnosis and treatment of chronic infection and vaccine development are also discussed.
Collapse
Affiliation(s)
- Samantha Bannister
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Infectious Diseases Unit, Royal Children’s Hospital Melbourne, Parkville, Australia
| | - Nicole L. Messina
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
| | - Boris Novakovic
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Epigenetics Research Group, Murdoch Children’s Research Institute, Parkville, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Infectious Diseases Unit, Royal Children’s Hospital Melbourne, Parkville, Australia
| |
Collapse
|
45
|
Singh G, Pritam M, Banerjee M, Singh AK, Singh SP. Designing of precise vaccine construct against visceral leishmaniasis through predicted epitope ensemble: A contemporary approach. Comput Biol Chem 2020; 86:107259. [PMID: 32339913 DOI: 10.1016/j.compbiolchem.2020.107259] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/25/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022]
Abstract
Visceral leishmaniasis (VL) caused by Leishmania donovani is a fatal parasitic disease affecting primarily the poor population in endemic countries. Increasing number of deaths as well as resistant to existing drugs necessitates the development of an effective vaccine for successful treatment of VL. The present study employed a combinatorial approach for designing monomer vaccine construct against L. donovani by applying forecasted B- and T- cell epitopes from 4 genome derived antigenic proteins having secretory signal peptides and glycophosphatidylinositol (GPI) anchors with ≤ 1 transmembrane helix. The forecasted population coverage of chosen T cell epitope ensemble (combined HLA class I and II) cover 99.14 % of world-wide human population. The predicted 3D structure of vaccine constructs (VC1/VC2) were modeled using homology modeling approach and docked to innate immune receptors TLR-2 and TLR-4 with respective docking energies -1231.4/-910.3 and -1119.4/-1476 kcal/mol. Overall, the aforementioned designed vaccine constructs were found appropriate for including in self-assembly protein nanoparticles (SAPN) for further study in developing cutting-edge precision vaccine against VL in short duration with cost-effective manner.
Collapse
Affiliation(s)
- Garima Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India.
| | - Manisha Pritam
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India.
| | - Monisha Banerjee
- Molecular and Human Genetics Lab, Department of Zoology, University of Lucknow, Lucknow, 226007, India.
| | - Akhilesh Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India; Department of Biotechnology, Mahatma Gandhi Central University, Bihar, 845401, India.
| | - Satarudra Prakash Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India; Department of Biotechnology, Mahatma Gandhi Central University, Bihar, 845401, India.
| |
Collapse
|
46
|
Elicitation of integrated immunity in mice by a novel pneumococcal polysaccharide vaccine conjugated with HBV surface antigen. Sci Rep 2020; 10:6470. [PMID: 32286332 PMCID: PMC7156719 DOI: 10.1038/s41598-020-62185-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/10/2020] [Indexed: 12/25/2022] Open
Abstract
The conjugation of polysaccharides with an effective carrier protein is critical for the development of effective bacterial polysaccharide vaccines. Therefore, the identification and optimization of carrier proteins to induce an effective immune response is necessary for developing a combined vaccine. In the current study, we utilized hepatitis B virus surface antigen (HBsAg) as a novel carrier protein combined with a capsular polysaccharide molecule to develop a new pneumococcal conjugated vaccine. The specific antibodies and T cell immune response against the capsular polysaccharide and HBsAg in the mice immunized with this conjugated vaccine were evaluated. In addition, the unique gene profiles of immune cells induced by this conjugated vaccine in the immunized mice were analyzed. Our results demonstrated that the vaccine consisting of pneumonia type 33 F capsular polysaccharide (Pn33Fps) conjugated with HBsAg can induce strong specific immune responses against both antigens in vivo in immunized mice. Furthermore, the conjugated vaccine induced higher expression of genes related to the activation of immunity and higher antibody titers against Pn33Fps and HBsAg in mice than those obtained via vaccination with a single antigen. Analyses of the dynamic expression changes in immunity-related genes in mice immunized with Pn33Fps_HBs, Pn33Fps, or HBsAg indicated the potent immunogenicity of the conjugated vaccine. In addition, a pathological evaluation of the organs from immunized mice further suggested that the conjugated vaccine is safe. Together, these results indicate that a conjugated vaccine consisting of Pn33Fps with HBsAg is a novel and effective vaccine.
Collapse
|
47
|
Dawood MAO, Zommara M, Eweedah NM, Helal AI, Aboel-Darag MA. The potential role of nano-selenium and vitamin C on the performances of Nile tilapia (Oreochromis niloticus). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:9843-9852. [PMID: 31925699 DOI: 10.1007/s11356-020-07651-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/07/2020] [Indexed: 05/21/2023]
Abstract
Functional trace elements and vitamins can boost immunity and antioxidative response in aquatic animals without creating environmental hazards. While nano-selenium (Nano-Se) and vitamin C (VC) have been used as immunomodulators and antioxidants in animal and poultry feed, there is little data on Nano-Se and/or VC supplementation in aquatic animals. Thus, the current study evaluated the impact of adding Nano-Se and VC to the diets of Nile tilapia for 8 weeks. Four diets were formulated and offered to the fish: no supplementation (control), 1 mg Nano-Se/kg, 500 mg VC/kg, and 1 mg Nano-Se + 500 mg VC/kg of food. Growth-related parameters (final body weight, weight gain, and specific growth rate) were significantly increased in tilapia fed Nano-Se and VC, with a reduced feed conversion ratio (P < 0.05). Intestinal villus length and width as well as the number of goblet cells were increased in tilapia fed Nano-Se and/or VC (P < 0.05). Additionally, dietary Nano-Se and/or VC significantly increased nitro-blue tetrazolium (NBT) level, superoxide dismutase, glutathione peroxidase, catalase, the phagocytic index, and lysozyme and phagocytic activities (P < 0.05). However, significantly reduced levels of malonaldehyde were observed in fish fed Nano-Se and/or VC (P < 0.05). TNF-α and IL-1β gene expressions in the liver and spleen of the fish were significantly upregulated by Nano-Se and/or VC (P < 0.05). The results revealed the potential role of Nano-Se and/or VC in enhancing growth, intestinal morphometry, and immune and antioxidative responses in Nile tilapia.
Collapse
Affiliation(s)
- Mahmoud A O Dawood
- Department of Animal Production, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| | - Mohsen Zommara
- Department of Dairy Science, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Nabil M Eweedah
- Department of Animal Production, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Azmy I Helal
- Department of Animal Production, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Mohamed A Aboel-Darag
- Department of Animal Production, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| |
Collapse
|
48
|
Oli AN, Obialor WO, Ifeanyichukwu MO, Odimegwu DC, Okoyeh JN, Emechebe GO, Adejumo SA, Ibeanu GC. Immunoinformatics and Vaccine Development: An Overview. Immunotargets Ther 2020; 9:13-30. [PMID: 32161726 PMCID: PMC7049754 DOI: 10.2147/itt.s241064] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/25/2020] [Indexed: 12/11/2022] Open
Abstract
The use of vaccines have resulted in a remarkable improvement in global health. It has saved several lives, reduced treatment costs and raised the quality of animal and human lives. Current traditional vaccines came empirically with either vague or completely no knowledge of how they modulate our immune system. Even at the face of potential vaccine design advance, immune-related concerns (as seen with specific vulnerable populations, cases of emerging/re-emerging infectious disease, pathogens with complex lifecycle and antigenic variability, need for personalized vaccinations, and concerns for vaccines' immunological safety -specifically vaccine likelihood to trigger non-antigen-specific responses that may cause autoimmunity and vaccine allergy) are being raised. And these concerns have driven immunologists toward research for a better approach to vaccine design that will consider these challenges. Currently, immunoinformatics has paved the way for a better understanding of some infectious disease pathogenesis, diagnosis, immune system response and computational vaccinology. The importance of this immunoinformatics in the study of infectious diseases is diverse in terms of computational approaches used, but is united by common qualities related to host–pathogen relationship. Bioinformatics methods are also used to assign functions to uncharacterized genes which can be targeted as a candidate in vaccine design and can be a better approach toward the inclusion of women that are pregnant into vaccine trials and programs. The essence of this review is to give insight into the need to focus on novel computational, experimental and computation-driven experimental approaches for studying of host–pathogen interactions and thus making a case for its use in vaccine development.
Collapse
Affiliation(s)
- Angus Nnamdi Oli
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Wilson Okechukwu Obialor
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Martins Ositadimma Ifeanyichukwu
- Department of Immunology, College of Health Sciences, Faculty of Medicine, Nnamdi Azikiwe University, Anambra, Nigeria.,Department of Medical Laboratory Science,Faculty of Health Science and Technology, College of Health Sciences, Nnamdi Azikiwe University,Nnewi Campus, Nnewi, Nigeria
| | - Damian Chukwu Odimegwu
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, University of Nigeria Nsukka, Enugu, Nigeria
| | - Jude Nnaemeka Okoyeh
- Department of Biology and Clinical Laboratory Science, Division of Arts and Sciences, Neumann University, Aston, PA 19014-1298, USA
| | - George Ogonna Emechebe
- Department of Pediatrics, Faculty of Clinical Medicine, Chukwuemeka Odumegwu Ojukwu University, Awka, Nigeria
| | - Samson Adedeji Adejumo
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Gordon C Ibeanu
- Department of Pharmaceutical Science, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
49
|
Okon E, Kukula-Koch W, Jarzab A, Halasa M, Stepulak A, Wawruszak A. Advances in Chemistry and Bioactivity of Magnoflorine and Magnoflorine-Containing Extracts. Int J Mol Sci 2020; 21:ijms21041330. [PMID: 32079131 PMCID: PMC7072879 DOI: 10.3390/ijms21041330] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/09/2023] Open
Abstract
The review collects together some recent information on the identity and pharmacological properties of magnoflorine, a quaternary aporphine alkaloid, that is widely distributed within the representatives of several botanical families like Berberidaceae, Magnoliaceae, Papaveraceae, or Menispermaceae. Several findings published in the scientific publications mention its application in the treatment of a wide spectrum of diseases including inflammatory ones, allergies, hypertension, osteoporosis, bacterial, viral and fungal infections, and some civilization diseases like cancer, obesity, diabetes, dementia, or depression. The pharmacokinetics and perspectives on its introduction to therapeutic strategies will also be discussed.
Collapse
Affiliation(s)
- Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland
- Correspondence: (W.K.-K.); (A.W.); Tel.: +48-81448-6350 (W.K.-K.); +48-81448-7087 (A.W.)
| | - Agata Jarzab
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
- Correspondence: (W.K.-K.); (A.W.); Tel.: +48-81448-6350 (W.K.-K.); +48-81448-7087 (A.W.)
| |
Collapse
|
50
|
da Costa A, Nascimento ND, Galisteo AJ, Dias Dos Passos AB, de Andrade HF. Radiation effects on Toxoplasma antigens: different immune responses of irradiated intact tachyzoites or soluble antigens in experimental mice models. Int J Radiat Biol 2020; 96:697-706. [PMID: 31855098 DOI: 10.1080/09553002.2020.1704298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Purpose: Purpose: Protein irradiation causes aggregation, chain breakage, and oxidation, enhancing its uptake by antigen-presenting cells. To evaluate if irradiated proteins participate on the protection, we studied the immune response induced in mice immunized with irradiated soluble extracts of T. gondii tachyzoites (STag) or irradiated intact T. gondii RH tachyzoites (RH0.25 kGy).Material and Methods: Soluble extracts of Toxoplasma gondii tachyzoites (STag) were irradiated at different dose by Cobalt-60 source. By polyacrylamide gel electrophoresis (SDS-Page) we evaluated the effects on primary structures of protein STags induced by irradiation. By Enzyme-linked Immunosorbent Assay (ELISA) we evaluated the difference between humoral immune response induced by irradiated STag or RH tachyzoites in immunized mice from the detection of specific immunoglobulin G (IgG) antibodies in the serum of immunized mice. From challenge with viable RH strain of T. gondii we evaluated the protection induced in the immunized animals. By cytometry we performed the phenotyping of T and B lymphocytes in the peripheral blood of the immunized animals.Results: Irradiation dose of 1.5 kGy induced minimal changes in most proteins, without affecting their antigenicity or immunogenicity. Immunization showed saturation at the dose of 10 µg/mice, with worst response at higher doses. STag irradiated at 1.5 kGy (STag1.5 kGy) induced higher survival and protection similar to T. gondii RH strain irradiated at 0.25 kGy (RH0.25 kGy), with higher serum levels of high affinity IgG compared to STag native. Blood immune memory cells of mice immunized with STag1.5 kGy had higher proportions of CD19+ (cluster of differentiation 19) and CD4+ (cluster of differentiation 14) cells, whereas mice RH0.25 kGy had high proportion of memory CD8+ (cluster of differentiation 8) cells.Conclusions: Our data suggest that major histocompatibility complex type I (MHCI) pathway, appears seem to be used by RH0.25 kGy to generate cytotoxic cells while STag1.5 kGy uses a major histocompatibility complex type II (MHCII) pathway for B-cell memory, but both induce sufficient immune response for protection in mice without any adjuvant. Irradiation of soluble protein extracts enhances their immune response, allowing similar protection against T. gondii in mice as compared to irradiated intact parasites.
Collapse
Affiliation(s)
- Andrea da Costa
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brasil
| | - Nanci do Nascimento
- Instituto de Pesquisas Energéticas e Nucleares, Centro de Biotecnologia, São Paulo, Brasil
| | - Andrés Jimenez Galisteo
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brasil.,Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brasil
| | - Aline Bastos Dias Dos Passos
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brasil
| | - Heitor Franco de Andrade
- Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brasil
| |
Collapse
|