101
|
Intrathymic differentiation of natural antibody-producing plasma cells in human neonates. Nat Commun 2021; 12:5761. [PMID: 34599177 PMCID: PMC8486820 DOI: 10.1038/s41467-021-26069-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
The thymus is a central lymphoid organ primarily responsible for the development of T cells. A small proportion of B cells, however, also reside in the thymus to assist negative selection of self-reactive T cells. Here we show that the thymus of human neonates contains a consistent contingent of CD138+ plasma cells, producing all classes and subclasses of immunoglobulins with the exception of IgD. These antibody-secreting cells are part of a larger subset of B cells that share the expression of signature genes defining mouse B1 cells, yet lack the expression of complement receptors CD21 and CD35. Data from single-cell transcriptomic, clonal correspondence and in vitro differentiation assays support the notion of intrathymic CD138+ plasma cell differentiation, alongside other B cell subsets with distinctive molecular phenotypes. Lastly, neonatal thymic plasma cells also include clones reactive to commensal and pathogenic bacteria that commonly infect children born with antibody deficiency. Thus, our findings point to the thymus as a source of innate humoral immunity in human neonates.
Collapse
|
102
|
Domínguez-Avila JA, Villa-Rodriguez JA, Montiel-Herrera M, Pacheco-Ordaz R, Roopchand DE, Venema K, González-Aguilar GA. Phenolic Compounds Promote Diversity of Gut Microbiota and Maintain Colonic Health. Dig Dis Sci 2021; 66:3270-3289. [PMID: 33111173 DOI: 10.1007/s10620-020-06676-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/13/2020] [Indexed: 02/08/2023]
Abstract
The role of non-energy-yielding nutrients on health has been meticulously studied, and the evidence shows that a compound can exert significant effects on health even if not strictly required by the organism. Phenolic compounds are among the most widely studied molecules that fit this description; they are found in plants as secondary metabolites and are not required by humans for growth or development, but they can influence a wide array of processes that modulate health across multiple organs and systems. The lower gastrointestinal tract is a prime site of action of phenolic compounds, namely, by their effects on gut microbiota and colonic health. As with humans, phenolic compounds are not required by most bacteria but can be substrates of others; in fact, some phenolic compounds exert antibacterial actions. A diet rich in phenolic compounds can lead to qualitative and quantitative effects on gut microbiota, thereby inducing indirect health effects in mammals through the action of these microorganisms. Moreover, phenolic compounds may be fermented by the gut microbiota, thereby modulating the compounds bioactivity. In the colon, phenolic compounds promote anti-inflammatory, anti-oxidant and antiproliferative actions. The aim of the present review is to highlight the role of phenolic compounds on maintaining or restoring a healthy microbiota and overall colonic health. Mechanisms of action that substantiate the reported evidence will also be discussed.
Collapse
Affiliation(s)
- J Abraham Domínguez-Avila
- Cátedras CONACYT-Centro de Investigación en Alimentación y Desarrollo A. C., Carretera Gustavo Enrique Astiazarán Rosas No. 46, Col. La Victoria, 83304, Hermosillo, Sonora, Mexico.
| | - Jose A Villa-Rodriguez
- Center for Digestive Health, Department of Food Science, Institute for Food Nutrition and Health, Rutgers, The State University of New Jersey, 61 Dudley Road, New Brunswick, NJ, 08901, USA
| | - Marcelino Montiel-Herrera
- Departamento de Medicina y Ciencias de la Salud, Universidad de Sonora, 83000, Hermosillo, Sonora, Mexico
| | - Ramón Pacheco-Ordaz
- Centro de Investigación en Alimentación y Desarrollo A. C., Carretera Gustavo Enrique Astiazarán Rosas No. 46, Col. La Victoria, 83304, Hermosillo, Sonora, Mexico
| | - Diana E Roopchand
- Center for Digestive Health, Department of Food Science, Institute for Food Nutrition and Health, Rutgers, The State University of New Jersey, 61 Dudley Road, New Brunswick, NJ, 08901, USA
| | - Koen Venema
- Centre for Healthy Eating and Food Innovation, Maastricht University - Campus Venlo, St. Jansweg 20, 5928 RC, Venlo, The Netherlands
| | - Gustavo A González-Aguilar
- Centro de Investigación en Alimentación y Desarrollo A. C., Carretera Gustavo Enrique Astiazarán Rosas No. 46, Col. La Victoria, 83304, Hermosillo, Sonora, Mexico
| |
Collapse
|
103
|
Schultze-Florey CR, Chukhno E, Goudeva L, Blasczyk R, Ganser A, Prinz I, Förster R, Koenecke C, Odak I. Distribution of major lymphocyte subsets and memory T-cell subpopulations in healthy adults employing GLP-conforming multicolor flow cytometry. Leukemia 2021; 35:3021-3025. [PMID: 34290358 PMCID: PMC8478656 DOI: 10.1038/s41375-021-01348-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Christian R Schultze-Florey
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Lilia Goudeva
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
104
|
SIGIRR Mutation in Human Necrotizing Enterocolitis (NEC) Disrupts STAT3-Dependent microRNA Expression in Neonatal Gut. Cell Mol Gastroenterol Hepatol 2021; 13:425-440. [PMID: 34563711 PMCID: PMC8688179 DOI: 10.1016/j.jcmgh.2021.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS Single immunoglobulin interleukin-1-related receptor (SIGIRR) is a major inhibitor of Toll-like receptor signaling. Our laboratory identified a novel SIGIRR stop mutation (p.Y168X) in an infant who died of severe necrotizing enterocolitis (NEC). Herein, we investigated the mechanisms by which SIGIRR mutations induce Toll-like receptor hyper-responsiveness in the neonatal gut, disrupting postnatal intestinal adaptation. METHODS Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 was used to generate transgenic mice encoding the SIGIRR p.Y168X mutation. Ileal lysates, mouse intestinal epithelial cell (IEC) lysates, and intestinal sections were used to assess inflammation, signal transducer and activator of transcription 3 (STAT3) phosphorylation, microRNA (miRNA), and interleukin-1-related-associated kinase 1 (IRAK1) expression. Western blot, quantitative reverse-transcription polymerase chain reaction(qRT-PCR), and luciferase assays were performed to investigate SIGIRR-STAT3 signaling in human intestinal epithelial cells (HIEC) expressing wild-type or SIGIRR (p.Y168X) plasmids. RESULTS SigirrTg mice showed increased intestinal inflammation and nuclear factor-κB activation concomitant with decreased IEC expression of miR-146a and miR-155. Mechanistic studies in HIECs showed that although SIGIRR induced STAT3-mediated expression of miR-146a and miR-155, the p.Y168X mutation disrupted SIGIRR-mediated STAT3-dependent miRNA expression. Chromatin immunoprecipitation and luciferase assays showed that SIGIRR activation of STAT3-induced miRNA expression is dependent on IRAK1. Both in HIECs and in the mouse intestine, decreased expression of miR-146a observed with the p.Y168X mutation increased expression of IRAK1, a protein whose down-regulation is important for postnatal gut adaptation. CONCLUSIONS Our results uncover a novel pathway (SIGIRR-STAT3-miRNA-IRAK1 repression) by which SIGIRR regulates postnatal intestine adaptation, which is disrupted by a SIGIRR mutation identified in human NEC. These data provide new insights into how human genetic mutations in SIGIRR identified in NEC result in loss of postnatal intestinal immune tolerance.
Collapse
|
105
|
Preeti K, Sood A, Fernandes V. Metabolic Regulation of Glia and Their Neuroinflammatory Role in Alzheimer's Disease. Cell Mol Neurobiol 2021; 42:2527-2551. [PMID: 34515874 DOI: 10.1007/s10571-021-01147-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disorder. It is characterized clinically by progressive memory loss and impaired cognitive function. Its progression occurs from neuronal synapse loss to amyloid pathology and Tau deposit which eventually leads to the compromised neuronal function. Neurons in central nervous tissue work in a composite and intricate network with the glia and vascular cells. Microglia and astrocytes are becoming the prime focus due to their involvement in various aspects of neurophysiology, such as trophic support to neurons, synaptic modulation, and brain surveillance. AD is also often considered as the sequela of prolonged metabolic dyshomeostasis. The neuron and glia have different metabolic profiles as cytosolic glycolysis and mitochondrial-dependent oxidative phosphorylation (OXPHOS), especially under dyshomeostasis or with aging pertaining to their unique genetic built-up. Various efforts are being put in to decipher the role of mitochondrial dynamics regarding their trafficking, fission/fusion imbalance, and mitophagy spanning over both neurons and glia to improve aging-related brain health. The mitochondrial dysfunction may lead to activation in various signaling mechanisms causing metabolic reprogramming in glia cells, further accelerating AD-related pathogenic events. The glycolytic-dominant astrocytes switch to the neurotoxic phenotype, i.e., disease-associated astrocyte under metabolic stress. The microglia also transform from resting to reactive phenotype, i.e., disease-associated microglia. It may also exist in otherwise a misconception an M1, glycolytic, or M2, an OXPHOS-dependent phenotype. Further, glial transformation plays a vital role in regulating hallmarks of AD pathologies like synapse maintenance, amyloid, and Tau clearance. In this updated review, we have tried to emphasize the metabolic regulation of glial reactivity, mitochondrial quality control mechanisms, and their neuroinflammatory response in Alzheimer's progression.
Collapse
Affiliation(s)
- Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| | - Anika Sood
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Valencia Fernandes
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
106
|
Yao ZA, Xu L, Jin LM, Wang BX, Fu CZ, Bai Y, Wu HG. κ-Carrageenan Oligosaccharides Inhibit the Inflammation of Lipopolysaccharide-Activated Microglia Via TLR4/NF-κB and p38/JNK MAPKs Pathways. Neurochem Res 2021; 47:295-304. [PMID: 34491516 DOI: 10.1007/s11064-021-03443-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
Microglial inflammation plays an essential role in neurodegenerative disease. Our previous studies had shown that κ-carrageenan oligosaccharides (KOS) could inhibit the excessive activation of microglia that induced by LPS, while the interrelated mechanisms were still indistinct. Therefore, we detected the inflammatory signaling pathway on LPS-activated microglia that pretreat by different content of KOS to reveal the mechanism on KOS's inhibition of microglia inflammatory response. ELISA was used to detect the effects of KOS on the secretion of interleukin-1 (IL-1β), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and prostaglandin E2 (PG-2) by LPS-activated microglia, respectively. The production of reactive oxygen species (ROS) and nitric oxide (NO) in microglia cells was detected by flow cytometry, and the protein expression of immunoinflammation-related signaling pathways were detected by Western Blot. The results showed that KOS could significantly protected the microglia from the over-activated inflammatory by inhibiting the release of inflammatory cytokines and the oxidative stress response. And KOS could reduce the expression of the protein that related to the TLR4/NF-κB and p38/JNK MAPKs pathways activated by LPS in microglia. However, there may be no specific target of KOS in cells. Therefore, KOS, a natural algal source oligosaccharide, has immunomodulatory effects and can be used as a potential intervention therapy for inflammatory related neurodegenerative diseases.
Collapse
Affiliation(s)
- Zi-Ang Yao
- College of Life Science, Dalian Minzu University, Dalian, 116600, Liaoning, China
| | - Ling Xu
- Department of Clinical Laboratory, Xinhua Hospital Affiliated to Dalian University, Dalian, 116021, Liaoning, China
| | - Li-Ming Jin
- College of Life Science, Dalian Minzu University, Dalian, 116600, Liaoning, China
| | - Bai-Xiang Wang
- College of Life Science and Technology, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China
| | - Cheng-Zhu Fu
- College of Life Science and Technology, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China
| | - Ying Bai
- Department of Clinical Laboratory, Xinhua Hospital Affiliated to Dalian University, Dalian, 116021, Liaoning, China
| | - Hai-Ge Wu
- College of Life Science and Technology, Dalian University, No. 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China.
| |
Collapse
|
107
|
Yeung SSH, Ho YS, Chang RCC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med 2021; 53:1251-1267. [PMID: 34489558 PMCID: PMC8492689 DOI: 10.1038/s12276-021-00660-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.
Collapse
Affiliation(s)
- Sherry Sin-Hang Yeung
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Yuen-Shan Ho
- grid.16890.360000 0004 1764 6123School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR China
| | - Raymond Chuen-Chung Chang
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| |
Collapse
|
108
|
Cananzi M, Wohler E, Marzollo A, Colavito D, You J, Jing H, Bresolin S, Gaio P, Martin R, Mescoli C, Bade S, Posey JE, Dalle Carbonare M, Tung W, Jhangiani SN, Bosa L, Zhang Y, Filho JS, Gabelli M, Kellermayer R, Kader HA, Oliva-Hemker M, Perilongo G, Lupski JR, Biffi A, Valle D, Leon A, de Macena Sobreira NL, Su HC, Guerrerio AL. IFIH1 loss-of-function variants contribute to very early-onset inflammatory bowel disease. Hum Genet 2021; 140:1299-1312. [PMID: 34185153 PMCID: PMC8423350 DOI: 10.1007/s00439-021-02300-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Genetic defects of innate immunity impairing intestinal bacterial sensing are linked to the development of Inflammatory Bowel Disease (IBD). Although much evidence supports a role of the intestinal virome in gut homeostasis, most studies focus on intestinal viral composition rather than on host intestinal viral sensitivity. To demonstrate the association between the development of Very Early Onset IBD (VEOIBD) and variants in the IFIH1 gene which encodes MDA5, a key cytosolic sensor for viral nucleic acids. Whole exome sequencing (WES) was performed in two independent cohorts of children with VEOIBD enrolled in Italy (n = 18) and USA (n = 24). Luciferase reporter assays were employed to assess MDA5 activity. An enrichment analysis was performed on IFIH1 comparing 42 VEOIBD probands with 1527 unrelated individuals without gastrointestinal or immunological issues. We identified rare, likely loss-of-function (LoF), IFIH1 variants in eight patients with VEOIBD from a combined cohort of 42 children. One subject, carrying a homozygous truncating variant resulting in complete LoF, experienced neonatal-onset, pan-gastrointestinal, IBD-like enteropathy plus multiple infectious episodes. The remaining seven subjects, affected by VEOIBD without immunodeficiency, were carriers of one LoF variant in IFIH1. Among these, two patients also carried a second hypomorphic variant, with partial function apparent when MDA5 was weakly stimulated. Furthermore, IFIH1 variants were significantly enriched in children with VEOIBD as compared to controls (p = 0.007). Complete and partial MDA5 deficiency is associated with VEOIBD with variable penetrance and expressivity, suggesting a role for impaired intestinal viral sensing in IBD pathogenesis.
Collapse
Affiliation(s)
- Mara Cananzi
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child with Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica, Fondazione Città della Speranza, Padova, Italy
| | - Davide Colavito
- Research & Innovation (R&I Genetics) Srl, C.so Stati Uniti 4, Padova, Italy
| | - Jing You
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Huie Jing
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Silvia Bresolin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica, Fondazione Città della Speranza, Padova, Italy
| | - Paola Gaio
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child with Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Renan Martin
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Claudia Mescoli
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University Hospital of Padova, Padova, Italy
| | - Sangeeta Bade
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Wesley Tung
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Luca Bosa
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child with Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Yu Zhang
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joselito Sobreira Filho
- Division of Genetics, Department of Morphology and Genetics, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Maria Gabelli
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Richard Kellermayer
- Section of Pediatric Gastroenterology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Howard A Kader
- Department of Pediatrics, Division of Pediatric Gastroenterology & Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Oliva-Hemker
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giorgio Perilongo
- Unit of Pediatric Gastroenterology, Digestive Endoscopy, Hepatology and Care of the Child with Liver Transplantation, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - David Valle
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Alberta Leon
- Research & Innovation (R&I Genetics) Srl, C.so Stati Uniti 4, Padova, Italy
| | | | - Helen C Su
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anthony L Guerrerio
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
109
|
Louise Benelli J, Basso RP, Rodrigues MDL, Poester VR, Munhoz LS, Aquino VR, Stevens DA, Xavier MO. Coinfection of disseminated cryptococcosis and BK Virus, a casualty of missed diagnosis during the COVID-19 Pandemic: A case report and review of the literature. Curr Med Mycol 2021; 7:44-49. [PMID: 35528625 PMCID: PMC9006733 DOI: 10.18502/cmm.7.2.7802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/14/2021] [Accepted: 10/17/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Purpose The COVID-19 pandemic resulted in an overload of health services and healthcare professionals. The result is a setback in health promotion and prevention, delays in diagnosis, and deaths from other diseases that are currently receiving inadequate attention. This article illustrates the risk of this negligence. Case report This study aimed to report a case of coinfection of disseminated cryptococcosis and BK virus in a patient without a previous diagnosis of human immunodeficiency virus infection and COVID-19 negative in the context of the COVID-19 pandemic. Despite receiving antifungal therapy, the patient died. Conclusion This fatal case is a warning regarding delay of diagnosis and neglect of other serious illnesses owing to the current pandemic, including fungal diseases and neglected diagnoses.
Collapse
Affiliation(s)
- Jéssica Louise Benelli
- Laboratory of Mycology, Faculty of Medicine, Federal University of Rio Grande, Rio Grande (FURG), Brazil
- Postgraduate Program in Health Science, Federal University of Rio Grande (PPGCS-FURG), Rio Grande, Brazil
- Clinical Analysis Laboratory, Dr. Miguel Riet Correa University Hospital, Rio Grande (HU-FURG/EBSERH), Brazil
| | - Rossana Patrícia Basso
- Postgraduate Program in Health Science, Federal University of Rio Grande (PPGCS-FURG), Rio Grande, Brazil
- Specialized Care Service in Infectious Diseases, Dr. Miguel Riet Correa University Hospital, Rio Grande, Brazil
| | - Márcia de Lima Rodrigues
- Specialized Care Service in Infectious Diseases, Dr. Miguel Riet Correa University Hospital, Rio Grande, Brazil
| | - Vanice Rodrigues Poester
- Laboratory of Mycology, Faculty of Medicine, Federal University of Rio Grande, Rio Grande (FURG), Brazil
- Postgraduate Program in Health Science, Federal University of Rio Grande (PPGCS-FURG), Rio Grande, Brazil
| | - Lívia Silveira Munhoz
- Postgraduate Program in Health Science, Federal University of Rio Grande (PPGCS-FURG), Rio Grande, Brazil
| | - Valerio Rodrigues Aquino
- Laboratory Diagnostic Service, Microbiology Unit, Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil
| | - David A Stevens
- California Institute for Medical Research, San Jose, and Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Melissa Orzechowski Xavier
- Laboratory of Mycology, Faculty of Medicine, Federal University of Rio Grande, Rio Grande (FURG), Brazil
- Postgraduate Program in Health Science, Federal University of Rio Grande (PPGCS-FURG), Rio Grande, Brazil
| |
Collapse
|
110
|
Sardesai AU, Tanak AS, Krishnan S, Striegel DA, Schully KL, Clark DV, Muthukumar S, Prasad S. An approach to rapidly assess sepsis through multi-biomarker host response using machine learning algorithm. Sci Rep 2021; 11:16905. [PMID: 34413363 PMCID: PMC8377018 DOI: 10.1038/s41598-021-96081-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 12/02/2022] Open
Abstract
Sepsis is a life-threatening condition and understanding the disease pathophysiology through the use of host immune response biomarkers is critical for patient stratification. Lack of accurate sepsis endotyping impedes clinicians from making timely decisions alongside insufficiencies in appropriate sepsis management. This work aims to demonstrate the potential feasibility of a data-driven validation model for supporting clinical decisions to predict sepsis host-immune response. Herein, we used a machine learning approach to determine the predictive potential of identifying sepsis host immune response for patient stratification by combining multiple biomarker measurements from a single plasma sample. Results were obtained using the following cytokines and chemokines IL-6, IL-8, IL-10, IP-10 and TRAIL where the test dataset was 70%. Supervised machine learning algorithm naïve Bayes and decision tree algorithm showed good accuracy of 96.64% and 94.64%. These promising findings indicate the proposed AI approach could be a valuable testing resource for promoting clinical decision making.
Collapse
Affiliation(s)
- Abha Umesh Sardesai
- Department of Computer Engineering, University of Texas at Dallas, 800 W. Campbell Rd., Richardson, TX, USA
| | - Ambalika Sanjeev Tanak
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd. BSB 11, Richardson, TX, USA
| | - Subramaniam Krishnan
- Austere Environments Consortium for Enhanced Sepsis Outcomes (ACESO), Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Deborah A Striegel
- Austere Environments Consortium for Enhanced Sepsis Outcomes (ACESO), Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Kevin L Schully
- Biological Defense Research Directorate, Naval Medical Research Center-Frederick, Ft. Detrick, MD, 21702, USA
| | - Danielle V Clark
- Austere Environments Consortium for Enhanced Sepsis Outcomes (ACESO), Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Sriram Muthukumar
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd. BSB 11, Richardson, TX, USA.
- EnLiSense LLC, 1813 Audubon Pondway, 1813 Audubon Pond Way, Allen, TX, 75013, USA.
| | - Shalini Prasad
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd. BSB 11, Richardson, TX, USA.
| |
Collapse
|
111
|
Fang Y, Wang Y, Zeng D, Zhi S, Shu T, Huang N, Zheng S, Wu J, Liu Y, Huang G, Xue Y, Bin J, Liao Y, Shi M, Liao W. Comprehensive analyses reveal TKI-induced remodeling of the tumor immune microenvironment in EGFR/ALK-positive non-small-cell lung cancer. Oncoimmunology 2021; 10:1951019. [PMID: 34345533 PMCID: PMC8288040 DOI: 10.1080/2162402x.2021.1951019] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Tyrosine kinase inhibitors (TKI) play a pivotal role in the treatment of non-small-cell lung cancer (NSCLC) with mutations in epidermal growth factor receptor (EGFR) and rearrangements in anaplastic lymphoma kinase (ALK). However, the influences of TKIs on the tumor immune microenvironment (TIM), especially dynamic changes of responders, have not yet been fully elucidated. Therefore, RNA sequencing and whole-exome sequencing were performed on EGFR/ALK-positive NSCLC samples before and after TKI treatment. In combination with neoantigen and mutational-load estimations, xCell and single-sample gene set enrichment analysis (ssGSEA) were used to assess tumor immune-cell infiltration and activity. Furthermore, weighted-gene correlation network analysis and the bottleneck method were used to identify the hub genes that affected treatment-related immune responses. We found that TKI treatment remodeled the TIM in treatment-responsive samples. Profound increases in the rate of anti-tumor cell infiltration and cytotoxicity was observed following TKI treatment, while antigen presentation was limited in ALK-rearranged samples. However, no significant change in anti-tumor cell infiltration or cytotoxicity was found between pre-treatment and post-progression samples. Subsequently, we found that neurofilament heavy (NEFH) mutations were enriched in samples after TKI treatment and were associated with reduced neutrophil infiltration. The cytotoxicity of EGFR-mutant NSCLCs with co-driver TP53 mutation and ALK-rearranged samples with wild-type TP53 seems to be more easily induced by TKI. Finally, the immune-associated score generated by hub genes was positively correlated with immune infiltration, immune activation, and a favorable prognosis. In conclusion, the dynamic changes in the TIM provide clues to drug selection and timing for TKI-immunotherapy combinations.
Collapse
Affiliation(s)
- Yisheng Fang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanyuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shimeng Zhi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tingting Shu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Siting Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yantan Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Genjie Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yichen Xue
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
112
|
Correlation between human health and reactive oxygen species produced in blood: a long-term chemiluminescence and fluorescence analysis. Sci Rep 2021; 11:14545. [PMID: 34267248 PMCID: PMC8282623 DOI: 10.1038/s41598-021-93887-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
The previous slide-glass type system could simultaneously detect reactive and highly reactive oxygen species, i.e., superoxide radicals (O2−·) and hypochlorite ions (OCl−) elicited from leucocytes in sample blood, but had some drawbacks, i.e., signal noise from air-flow stirring, potential biohazard risks, etc. because of open samples placed on a slide glass. We overcame these drawbacks by adopting a fluidic-chip container in a new system, which resulted in higher sensitivity and more stable measurements. Using the new system, we conducted a pilot study on nominally healthy volunteers to find whether or not the monitored activities of leukocytes can distinguish more or less unhealthy conditions from healthy ones. At first, healthy volunteers of both genders and of various ages showed that the fluctuation magnitudes (%) of O2−· and OCl− were nearly similar to each other and to that of the neutrophil count fluctuation. These parameters sometimes exceeded the healthy fluctuation range. By comparing these large fluctuations with the data of an inflammation marker C-reactive protein (CRP), the neutrophil count fluctuation and the timings/symptoms of abnormalities found in questionnaire, we could gain information suggesting the factors causing the large fluctuations. The new system could detect bodily abnormalities earlier than CRP or self-aware symptoms.
Collapse
|
113
|
Santos JRDJ, Jesus CAFD, Damasceno Pinto C. Scientific mapping of stem cells associated with Chagas disease : A bibliometric analysis. COLLNET JOURNAL OF SCIENTOMETRICS AND INFORMATION MANAGEMENT 2021. [DOI: 10.1080/09737766.2021.1977094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | | | - Cláudio Damasceno Pinto
- Technological Innovation Center of Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Salvador, Brazil
| |
Collapse
|
114
|
Dhenni R, Yohan B, Alisjahbana B, Lucanus A, Riswari SF, Megawati D, Haryanto S, Gampamole D, Hayati RF, Sari K, Witari NPD, Myint KSA, Sasmono RT. Comparative cytokine profiling identifies common and unique serum cytokine responses in acute chikungunya and dengue virus infection. BMC Infect Dis 2021; 21:639. [PMID: 34215212 PMCID: PMC8254284 DOI: 10.1186/s12879-021-06339-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 06/18/2021] [Indexed: 01/09/2023] Open
Abstract
Background Infection by chikungunya (CHIKV) and dengue virus (DENV) can cause a wide spectrum of clinical features, many of which are undifferentiated. Cytokines, which broadly also include chemokines and growth factors, have been shown to play a role in protective immunity as well as DENV and CHIKV pathogenesis. However, differences in cytokine response to both viruses remain poorly understood, especially in patients from countries where both viruses are endemic. Our study is therefore aimed to provide a comparative profiling of cytokine response induced by acute DENV and CHIKV infections in patients with similar disease stages and in experimental in vitro infections. Methods By using multiplex immunoassay, we compared host cytokine profiles between acute CHIKV and DENV infections by analysing serum cytokine levels of IL-1α, IL-4, IL-5, IL-8, IL-13, RANTES, MCP-3, eotaxin, PDGF-AB/BB, and FGF-2 from the sera of acute chikungunya and dengue fever patients. We further investigated the cytokine profile responses using experimental in vitro CHIKV and DENV infections of peripheral blood mononuclear cells (PBMCs). Results We found that both CHIKV and DENV-infected patients had an upregulated level of IL-8 and IL-4, with the highest IL-4 level observed in DENV-2 infected patients. Higher IL-8 level was also correlated with lower platelet count in dengue patients. IL-13 and MCP-3 downregulation was observed only in chikungunya patients, while conversely PDGF-AB/BB and FGF-2 downregulation was unique in dengue patients. Age-associated differential expression of IL-13, MCP-3, and IL-5 was also observed, while distinct kinetics of IL-4, IL-8, and FGF-2 expression between CHIKV and DENV-infected patients were identified. Furthermore, the unique pattern of IL-8, IL-13 and MCP-3, but not IL-4 expression was also recapitulated using experimental in vitro infection in PBMCs. Conclusions Taken together, our study identified common cytokine response profile characterized by upregulation of IL-8 and IL-4 between CHIKV and DENV infection. Downregulation of IL-13 and MCP-3 was identified as a unique cytokine response profile of acute CHIKV infection, while distinct downregulation of PDGF-AB/BB and FGF-2 characterized the response from acute DENV infection. Our study provides an important overview of the host cytokine responses between CHIKV and DENV infection, which is important to further understand the mechanism and pathology of these diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06339-6.
Collapse
Affiliation(s)
- Rama Dhenni
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | | | - Anton Lucanus
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia
| | | | - Dewi Megawati
- Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar, Bali, Indonesia
| | | | | | - Rahma F Hayati
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Kartika Sari
- Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar, Bali, Indonesia
| | - Ni Putu Diah Witari
- Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar, Bali, Indonesia
| | | | - R Tedjo Sasmono
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia.
| |
Collapse
|
115
|
Beattie G, Cohan CM, Victorino GP. Predicting Acute Respiratory Distress Syndrome in Severe Blunt Trauma: The Utility of Interleukin-18. Surg Infect (Larchmt) 2021; 22:948-954. [PMID: 33983849 DOI: 10.1089/sur.2021.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: In trauma, direct pulmonary injury and innate immune response activation primes the lungs for acute respiratory distress syndrome (ARDS). The inflammasome-dependent release of interleukin-18 (IL-18) was recently identified as a key mediator in ARDS pathogenesis, leading us to hypothesize that plasma IL-18 is a diagnostic predictor of ARDS in severe blunt trauma. Patients and Methods: Secondary analysis of the Inflammation and Host Response to Injury database was performed on plasma cytokines collected within 12 hours of severe blunt trauma. Trauma-related cytokines, including IL-18, were compared between patients with and without ARDS and were evaluated for association with ARDS using regression analysis. Threshold cytokine concentrations predictive of ARDS were determined using receiver-operating curve (ROC) analysis. Results: Cytokine analysis of patients without ARDS patients (n = 61) compared with patients with ARDS (n = 19) demonstrated elevated plasma IL-18 concentration in ARDS and IL-18 remained correlated with ARDS on logistic regression after confounder adjustment (p = 0.008). Additionally, ROC analysis revealed IL-18 as a strong ARDS predictor (area under the curve [AUC] = 0.83), with a threshold IL-18 value of 170 pg/mL (Youden index, 0.3). Unlike in patients without ARDS, elevated IL-18 persisted in patients with ARDS during the acute injury phase (p ≤ 0.02). Other trauma-related cytokines did not correlate with ARDS. Conclusions: In severe blunt trauma, IL-18 is a robust predictor of ARDS and remains elevated throughout the acute injury phase. These findings support the use of IL-18 as a key ARDS biomarker, promoting early identification of trauma patients at greater risk of developing ARDS. Timely recognition of ARDS and implementation of advantageous supportive care practices may reduce trauma-related ARDS morbidity and costs.
Collapse
Affiliation(s)
- Genna Beattie
- Department of Surgery, University of California San Francisco-East Bay, Oakland, California, USA
| | - Caitlin M Cohan
- Department of Surgery, University of California San Francisco-East Bay, Oakland, California, USA
| | - Gregory P Victorino
- Department of Surgery, University of California San Francisco-East Bay, Oakland, California, USA
| |
Collapse
|
116
|
Yang Y, Lin Y, Zhang Z, Xu R, Yu X, Deng F. Micro/nano-net guides M2-pattern macrophage cytoskeleton distribution via Src-ROCK signalling for enhanced angiogenesis. Biomater Sci 2021; 9:3334-3347. [PMID: 33725044 DOI: 10.1039/d1bm00116g] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Implant surface topography has been proven to determine the fate of adhered macrophage polarization, which is closely related to the cytoskeletal arrangement during adhesion. Our purpose was to establish a topography that is favourable to M2 macrophage switching by regulating macrophage cytoskeleton distribution. Two micro/nano-net structures with different pore sizes were generated by alkali bathing at medium (SAM) or high (SAH) temperature based on the micro-level surface. Their surface characteristics, in vitro macrophage polarization and impact on endothelial cells were analysed. The in vivo macrophage response and osseointegration were also tested. The results showed that the micro/nano-net has high hydrophilicity and moderate roughness. In the SAH and SAM groups, macrophages exhibited an elongated cytoskeleton with tiny protrusions and had a high M2/M1 polarization ratio with enhanced angiogenic ability, and in vivo studies also showed faster angiogenesis and bone formation in these groups. SAH showed even better results than SAM. For cytoskeleton related pathway explanation, ROCK expression was upregulated and Src expression was downregulated at the early or late adhesion stage in both the SAH and SAM groups. These results indicated that the micro/nano-net structure guides elongated macrophage adhesion states via Src-ROCK signalling and switches macrophages towards the M2 phenotype, which provides a cytoskeleton-oriented topography design for an ideal immune response.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Yujing Lin
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Zhengchuan Zhang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Ruogu Xu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Xiaoran Yu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Feilong Deng
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| |
Collapse
|
117
|
Lifelong Endurance Exercise as a Countermeasure Against Age-Related [Formula: see text] Decline: Physiological Overview and Insights from Masters Athletes. Sports Med 2021; 50:703-716. [PMID: 31873927 DOI: 10.1007/s40279-019-01252-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Maximum oxygen consumption ([Formula: see text]) is not only an indicator of endurance performance, but also a strong predictor of cardiovascular disease and mortality. This physiological parameter is known to decrease with aging. In turn, physical exercise might attenuate the rate of aging-related decline in [Formula: see text], which in light of the global population aging is of major clinical relevance, especially at advanced ages. In this narrative review, we summarize the evidence available from masters athletes about the role of lifelong endurance exercise on aging-related [Formula: see text] decline, with examples of the highest [Formula: see text] values reported in the scientific literature for athletes across different ages (e.g., 35 ml·kg-1·min-1 in a centenarian cyclist). These data suggest that a linear decrease in [Formula: see text] might be possible if physical exercise loads are kept consistently high through the entire life span, with [Formula: see text] values remaining higher than those of the general population across all ages. We also summarize the main physiological changes that occur with inactive aging at different system levels-pulmonary and cardiovascular function, blood O2 carrying capacity, skeletal muscle capillary density and oxidative capacity-and negatively influence [Formula: see text], and review how lifelong exercise can attenuate or even prevent most-but apparently not all (e.g., maximum heart rate decline)-of them. In summary, although aging seems to be invariably associated with a progressive decline in [Formula: see text], maintaining high levels of physical exercise along the life span slows the multi-systemic deterioration that is commonly observed in inactive individuals, thereby attenuating age-related [Formula: see text] decline.
Collapse
|
118
|
Fries CN, Curvino EJ, Chen JL, Permar SR, Fouda GG, Collier JH. Advances in nanomaterial vaccine strategies to address infectious diseases impacting global health. NATURE NANOTECHNOLOGY 2021; 16:1-14. [PMID: 32807876 DOI: 10.1038/s41565-020-0739-9] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 06/23/2020] [Indexed: 05/18/2023]
Abstract
Despite the overwhelming success of vaccines in preventing infectious diseases, there remain numerous globally devastating diseases without fully protective vaccines, particularly human immunodeficiency virus (HIV), malaria and tuberculosis. Nanotechnology approaches are being developed both to design new vaccines against these diseases as well as to facilitate their global implementation. The reasons why a given pathogen may present difficulties for vaccine design are unique and tied to the co-evolutionary history of the pathogen and humans, but there are common challenges that nanotechnology is beginning to help address. In each case, a successful vaccine will need to raise immune responses that differ from the immune responses raised by normal infection. Nanomaterials, with their defined compositions, commonly modular construction, and length scales allowing the engagement of key immune pathways, collectively facilitate the iterative design process necessary to identify such protective immune responses and achieve them reliably. Nanomaterials also provide strategies for engineering the trafficking and delivery of vaccine components to key immune cells and lymphoid tissues, and they can be highly multivalent, improving their engagement with the immune system. This Review will discuss these aspects along with recent nanomaterial advances towards vaccines against infectious disease, with a particular emphasis on HIV/AIDS, malaria and tuberculosis.
Collapse
Affiliation(s)
- Chelsea N Fries
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Jui-Lin Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sallie R Permar
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Genevieve G Fouda
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
119
|
Kim J, Park JA, Yim G, Jang H, Kim TH, Sohn H, Lee T. Fabrication of an electrochemical biosensor composed of multi-functional Ag ion intercalated DNA four-way junctions/rhodium nanoplate heterolayer on a micro-gap for C-reactive protein detection in human serum. Analyst 2021; 146:2131-2137. [PMID: 33861260 DOI: 10.1039/d0an02135k] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As inflammation plays a role in the pathogenesis of acute coronary syndromes, C-reactive protein (CRP) can be used as a biomarker. To detect CRP precisely, the authors prepared a CRP electrochemical biosensor consisting of an eight Ag ion-intercalated multifunctional DNA four-way junction (MF-DNA-4WJ) and a porous rhodium nanoparticle (pRhNP) heterolayer on a micro-gap electrode. To increase conductivity, we used eight Ag+ ion-inserted DNA four-way junctions through a C-C mismatch. Each DNA 4WJ was designed to have the CRP aptamer sequence, an anchoring region (thiol group), and two of four C-C mismatch regions at the end of the fragments. After an annealing step, the MF-DNA-4WJ assembly configuration and selective binding of CRP were confirmed through native TBM-PAGE (Tris-borate-magnesium chloride-polyacrylamide gel electrophoresis). The Au micro-gap electrode was fabricated to load 5 μl of the sample, and this was performed during eight experiments on one chip to establish the accuracy of the data. Then, pRhNPs were immobilized on a Au micro-gap electrode using cysteamine. To confirm the electrochemical properties, cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS) were conducted. The durability of pRhNPs was confirmed through CV. To test the sensing performance of the prepared CRP biosensor, the limit of detection (LOD) and selectivity tests were conducted using EIS. The results indicated that charge transfer resistance (Rct) can be used efficiently to probe these interactions within the variable CRP concentration range, from 1 pM to 100 nM (0.23 ng L-1-23 μg L-1). The LOD of this sensor was 0.349 pM (0.08 ng L-1) (at S/N = 3). As a result of diluting the CRP to the same concentration range in a 20% human serum sample, the LOD was 3.55 fM (0.814 pg L-1) (at S/N = 3).
Collapse
Affiliation(s)
- Jinmyeong Kim
- Department of Chemical Engineering, Kwangwoon University, 20 Kwangwoon-Ro, Nowon-Gu, Seoul 01897, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
120
|
Caruntu A, Scheau C, Tampa M, Georgescu SR, Caruntu C, Tanase C. Complex Interaction Among Immune, Inflammatory, and Carcinogenic Mechanisms in the Head and Neck Squamous Cell Carcinoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1335:11-35. [PMID: 33650087 DOI: 10.1007/5584_2021_626] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammation is deeply involved in the development of most types of cancer. Many studies focus on the interaction between immune-inflammatory mechanisms and tumorigenesis in the head and neck squamous cell carcinoma (HNSCC). In this chapter, we emphasize the complexity of processes underlying this interaction and discuss the mechanisms of carcinogenesis in HNSCC with a special focus on metabolic changes, inflammation, and the immune landscape. Unveiling complex connections between immuno-inflammatory processes and tumor initiation, promotion, and progression will open new directions in the reliable identification of predictive factors and therapeutic targets in HNSCC.
Collapse
Affiliation(s)
- Ana Caruntu
- Department of Oral and Maxillofacial Surgery, "Carol Davila" Central Military Emergency Hospital, Bucharest, Romania.,Faculty of Dental Medicine, "Titu Maiorescu" University, Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Mircea Tampa
- Department of Dermatology, "Victor Babes" Clinical Hospital for Infectious Diseases, Bucharest, Romania. .,Department of Dermatology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.
| | - Simona Roxana Georgescu
- Department of Dermatology, "Victor Babes" Clinical Hospital for Infectious Diseases, Bucharest, Romania.,Department of Dermatology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania. .,Department of Dermatology, "Prof. N.C. Paulescu" National Institute of Diabetes, Nutrition, and Metabolic Diseases, Bucharest, Romania.
| | - Cristiana Tanase
- Faculty of Dental Medicine, "Titu Maiorescu" University, Bucharest, Romania.,Department of Biochemistry-Proteomics, "Victor Babes" National Institute of Pathology, Bucharest, Romania
| |
Collapse
|
121
|
Damakoudi V, Feldner T, Dilji E, Belkin A, Richert C. Hybridization Networks of mRNA and Branched RNA Hybrids. Chembiochem 2021; 22:924-930. [PMID: 33105062 PMCID: PMC7984269 DOI: 10.1002/cbic.202000678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Indexed: 12/28/2022]
Abstract
Messenger RNA (mRNA) is emerging as an attractive biopolymer for therapy and vaccination. To become suitable for vaccination, mRNA is usually converted to a biomaterial, using cationic peptides, polymers or lipids. An alternative form of converting mRNA into a material is demonstrated that uses branched oligoribonucleotide hybrids with the ability to hybridize with one or more regions of the mRNA sequence. Two such hybrids with hexamer arms and adamantane tetraol as branching element were prepared by solution-phase synthesis. When a rabies mRNA was treated with the branched hybrids at 1 M NaCl concentration, biomaterials formed that contained both of the nucleic acids. These results show that branched oligoribonucleotides are an alternative to the often toxic reagents commonly used to formulate mRNA for medical applications.
Collapse
Affiliation(s)
| | - Tobias Feldner
- Institute of Organic ChemistryUniversity of Stuttgart70569StuttgartGermany
| | - Edina Dilji
- Institute of Organic ChemistryUniversity of Stuttgart70569StuttgartGermany
| | - Andrey Belkin
- Institute of Organic ChemistryUniversity of Stuttgart70569StuttgartGermany
| | - Clemens Richert
- Institute of Organic ChemistryUniversity of Stuttgart70569StuttgartGermany
| |
Collapse
|
122
|
Spallanzani RG. Visceral adipose tissue mesenchymal stromal cells in the intersection of immunology and metabolism. Am J Physiol Endocrinol Metab 2021; 320:E512-E519. [PMID: 33427047 DOI: 10.1152/ajpendo.00341.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Visceral adipose tissue (VAT) is now recognized as an endocrine organ that plays a key role in organismal homeostasis by integrating metabolic and immunological aspects. In healthy individuals, this fat depot participates in the storage and release of lipids as per physiological demand, while maintaining a local anti-inflammatory environment. In this regard, recent findings highlight the pivotal role of distinct subtypes of mesenchymal stromal cells (mSCs) as orchestrators of metabolic homeostasis by engendering adipocytes to sustain adequate lipid storage as well as immune regulators via cross-talk with specialized tissue-resident immunocytes, especially regulatory T cells (Tregs) and group 2 innate lymphoid cells (ILC2s) to prevent the development of local inflammation. In addition, these stromal-immunocyte interactions are influenced by a number of physiological conditions such as aging and sex hormones. Perturbation of VAT equilibrium occurring during obesity appreciably alters the distribution and phenotype of mSCs, immunocytes, and other cell types, thereby promoting the development of chronic, low-grade inflammation locally and systemically. These alterations impair metabolic signaling and substantially contribute to the onset of disease, including type 2 diabetes. The present mini-review discusses the latest advances in this area, with an emphasis on the newly uncovered heterogeneity of mSCs, how they communicate with Tregs and ILC2s under different physio-pathological circumstances and future challenges to face.
Collapse
|
123
|
Saygili E, Yildiz-Ozturk E, Green MJ, Ghaemmaghami AM, Yesil-Celiktas O. Human lung-on-chips: Advanced systems for respiratory virus models and assessment of immune response. BIOMICROFLUIDICS 2021; 15:021501. [PMID: 33791050 PMCID: PMC7990507 DOI: 10.1063/5.0038924] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/15/2021] [Indexed: 05/06/2023]
Abstract
Respiratory viral infections are leading causes of death worldwide. A number of human respiratory viruses circulate in all age groups and adapt to person-to-person transmission. It is vital to understand how these viruses infect the host and how the host responds to prevent infection and onset of disease. Although animal models have been widely used to study disease states, incisive arguments related to poor prediction of patient responses have led to the development of microfluidic organ-on-chip models, which aim to recapitulate organ-level physiology. Over the past decade, human lung chips have been shown to mimic many aspects of the lung function and its complex microenvironment. In this review, we address immunological responses to viral infections and elaborate on human lung airway and alveolus chips reported to model respiratory viral infections and therapeutic interventions. Advances in the field will expedite the development of therapeutics and vaccines for human welfare.
Collapse
Affiliation(s)
- Ecem Saygili
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
| | - Ece Yildiz-Ozturk
- Translational Pulmonary Research Center, Ege University, 35100 Izmir, Turkey
| | | | | | | |
Collapse
|
124
|
Víctor-Carvalho P, Thome R, Rapôso C. Can tetracyclines ensure help in multiple sclerosis immunotherapy? J Clin Transl Res 2021; 7:22-33. [PMID: 34104806 PMCID: PMC8177043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/15/2020] [Accepted: 10/27/2020] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a disease of the central nervous system where an autoimmune response leads to chronic inflammation. It represents the second leading cause of non-traumatic disability in the world, affecting mainly young adults and with high female to male incidence. At present, the causative agent in MS is unknown, preventing the development of prophylaxis policies and the understanding of how the human system copes with this complex inflammation. Tetracyclines (Tet) have attracted great attention due to their anti-inflammatory effects. Minocycline and doxycycline represent the second-generation Tet that have been largely used to treat acne and to suppress inflammation. In addition, they are safer and cheaper than other drugs currently used to treat MS. AIM This study aims to review recent data involving the Tet minocycline and doxycycline and their therapeutic potential in MS. RELEVANCE FOR PATIENTS Many of the drugs used to treat MS have severe side effects and are costly. Tet, on the other hand, are a safe and inexpensive class of drugs that can modulate the immune response in MS patients.
Collapse
Affiliation(s)
- Pedro Víctor-Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
- Laboratory of Drug Development, Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Rodolfo Thome
- Department of Neurology, Thomas Jefferson University, Philadelphia-PA, USA
| | - Catarina Rapôso
- Laboratory of Drug Development, Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
125
|
Xydia M, Rahbari R, Ruggiero E, Macaulay I, Tarabichi M, Lohmayer R, Wilkening S, Michels T, Brown D, Vanuytven S, Mastitskaya S, Laidlaw S, Grabe N, Pritsch M, Fronza R, Hexel K, Schmitt S, Müller-Steinhardt M, Halama N, Domschke C, Schmidt M, von Kalle C, Schütz F, Voet T, Beckhove P. Common clonal origin of conventional T cells and induced regulatory T cells in breast cancer patients. Nat Commun 2021; 12:1119. [PMID: 33602930 PMCID: PMC7893042 DOI: 10.1038/s41467-021-21297-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Regulatory CD4+ T cells (Treg) prevent tumor clearance by conventional T cells (Tconv) comprising a major obstacle of cancer immune-surveillance. Hitherto, the mechanisms of Treg repertoire formation in human cancers remain largely unclear. Here, we analyze Treg clonal origin in breast cancer patients using T-Cell Receptor and single-cell transcriptome sequencing. While Treg in peripheral blood and breast tumors are clonally distinct, Tconv clones, including tumor-antigen reactive effectors (Teff), are detected in both compartments. Tumor-infiltrating CD4+ cells accumulate into distinct transcriptome clusters, including early activated Tconv, uncommitted Teff, Th1 Teff, suppressive Treg and pro-tumorigenic Treg. Trajectory analysis suggests early activated Tconv differentiation either into Th1 Teff or into suppressive and pro-tumorigenic Treg. Importantly, Tconv, activated Tconv and Treg share highly-expanded clones contributing up to 65% of intratumoral Treg. Here we show that Treg in human breast cancer may considerably stem from antigen-experienced Tconv converting into secondary induced Treg through intratumoral activation.
Collapse
Affiliation(s)
- Maria Xydia
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany.
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany.
| | - Raheleh Rahbari
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
| | - Eliana Ruggiero
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Iain Macaulay
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- Technical Development, Earlham Institute, Norwich, UK
| | - Maxime Tarabichi
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- The Francis Crick Institute, London, UK
| | - Robert Lohmayer
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany
- Institute for Theoretical Physics, University of Regensburg, Regensburg, Germany
| | - Stefan Wilkening
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Tillmann Michels
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany
| | - Daniel Brown
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Sebastiaan Vanuytven
- The Francis Crick Institute, London, UK
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Svetlana Mastitskaya
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Sean Laidlaw
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
| | - Niels Grabe
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Hamamatsu Tissue Imaging and Analysis Centre, BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Maria Pritsch
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany
| | - Raffaele Fronza
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Klaus Hexel
- Flow Cytometry Core Facility, German Cancer Research Centre, Heidelberg, Germany
| | - Steffen Schmitt
- Flow Cytometry Core Facility, German Cancer Research Centre, Heidelberg, Germany
| | - Michael Müller-Steinhardt
- German Red Cross (DRK Blood Donation Service in Baden-Württemberg-Hessen) and Institute for Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Niels Halama
- Medical Oncology Department, National Centre for Tumor Diseases, Heidelberg, Germany
- Hamamatsu Tissue Imaging and Analysis Centre, BIOQUANT, University of Heidelberg, Heidelberg, Germany
| | - Christoph Domschke
- Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Manfred Schmidt
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
| | - Christof von Kalle
- Translational Oncology Department, National Centre for Tumor Diseases and German Cancer Research Centre, Heidelberg, Germany
- Clinical Study Centre, Charité/BIH, Berlin, Germany
| | - Florian Schütz
- Department of Gynecology and Obstetrics, University Hospital of Heidelberg, Heidelberg, Germany
| | - Thierry Voet
- The Cancer, Ageing and Somatic Mutation Program, Wellcome Sanger Institute, Hinxton, UK
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Philipp Beckhove
- RCI Regensburg Centre for Interventional Immunology, University and Department of Hematology/Oncology, University Medical Centre of Regensburg, Regensburg, Germany.
- Translational Immunology Department, German Cancer Research Centre, Heidelberg, Germany.
| |
Collapse
|
126
|
Ulitzka M, Carrara S, Grzeschik J, Kornmann H, Hock B, Kolmar H. Engineering therapeutic antibodies for patient safety: tackling the immunogenicity problem. Protein Eng Des Sel 2021; 33:5944198. [PMID: 33128053 DOI: 10.1093/protein/gzaa025] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Established monoclonal antibodies (mAbs) allow treatment of cancers, autoimmune diseases and other severe illnesses. Side effects either arise due to interaction with the target protein and its biology or result from of the patient's immune system reacting to the foreign protein. This immunogenic reaction against therapeutic antibodies is dependent on various factors. The presence of non-human sequences can trigger immune responses as well as chemical and post-translational modifications of the antibody. However, even fully human antibodies can induce immune response through T cell epitopes or aggregates. In this review, we briefly describe, how therapeutic antibodies can interact with the patient's immune system and summarize recent advancements in protein engineering and in silico methods to reduce immunogenicity of therapeutic monoclonal antibodies.
Collapse
Affiliation(s)
- Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.,Ferring Darmstadt Labs, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Stefania Carrara
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.,Ferring Darmstadt Labs, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Labs, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Henri Kornmann
- Ferring International Center S.A., Chemin de la Vergognausaz 50, CH-1162 Saint-Prex, Switzerland
| | - Björn Hock
- Ferring International Center S.A., Chemin de la Vergognausaz 50, CH-1162 Saint-Prex, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| |
Collapse
|
127
|
Zhang C, Hu S, Zosky GR, Wei X, Shu S, Wang D, Chai X. Paracoxib Alleviates Ventilator-Induced Lung Injury Through Functional Modulation of Lung-Recruited CD11bloLy6Chi Monocytes. Shock 2021; 55:236-243. [PMID: 32590697 DOI: 10.1097/shk.0000000000001591] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Lung-recruited Ly6Chi monocytes had been shown to be involved in ventilator-induced lung injury (VILI). Our present study aimed to investigate whether the cyclooxygenase-2 (COX-2) inhibition modulates the function of lung-recruited Ly6Chi monocytes in a mouse model of VILI. METHODS Mice were exposed to lipopolysaccharide (LPS; 20 ng) intraperitoneally prior to injurious mechanical ventilation (Vt = 30 mL/kg, PEEP = 0 cmH2O). A subgroup of mice was treated with intravenous parecoxib (30 mg/kg), a COX-2 inhibitor, 1 h prior to ventilation. Control mice received saline and were not ventilated. At the end of the experiment, blood gas analysis was performed and lung tissue was collected for histological assessment. Flow cytometry was employed to quantify the different populations of lung monocytes/macrophages and their function. Isolated Ly6Chi cells were used to measure the intracellular concentrations of reactive oxygen species (ROS) and nitric oxide (NO) by fluorescent probes, and cytokine production by cytometric bead array. RESULTS Exposure to LPS and injurious ventilation was associated with severe lung histological damage, oxygenation impairment, and pulmonary edema; all of which were largely attenuated following the treatment of parecoxib. Furthermore, flow cytometry analysis revealed that parecoxib caused a reduction in the number of the lung-recruited CD11bloLy6Chi monocytes while there was no effect on tissue-resident CD64+ alveolar macrophages. In addition, the production of oxidative stress products (ROS, NO), MHC-II expression, and inflammatory cytokines in response to LPS and VILI in CD11bloLy6Chi monocytes was ameliorated by parecoxib. CONCLUSION Parecoxib-induced alleviation of oxidative stress and inflammation in lung-recruited Ly6Chi monocytes may partly explain the beneficial action of COX-2 inhibition in VILI.
Collapse
Affiliation(s)
- Chaofeng Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shanshan Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Graeme R Zosky
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Xin Wei
- Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuhua Shu
- Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Di Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaoqing Chai
- Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
128
|
Ferluga J, Yasmin H, Bhakta S, Kishore U. Vaccination Strategies Against Mycobacterium tuberculosis: BCG and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:217-240. [PMID: 34661897 DOI: 10.1007/978-3-030-67452-6_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Tuberculosis (TB) is a highly contagious disease caused by Mycobacterium tuberculosis (Mtb) and is the major cause of morbidity and mortality across the globe. The clinical outcome of TB infection and susceptibility varies among individuals and even among different populations, contributed by host genetic factors such as polymorphism in the human leukocyte antigen (HLA) alleles as well as in cytokine genes, nutritional differences between populations, immunometabolism, and other environmental factors. Till now, BCG is the only vaccine available to prevent TB but the protection rendered by BCG against pulmonary TB is not uniform. To deliver a vaccine which can give consistent protection against TB is a great challenge with rising burden of drug-resistant TB. Thus, expectations are quite high with new generation vaccines that will improve the efficiency of BCG without showing any discordance for all forms of TB, effective for individual of all ages in all parts of the world. In order to enhance or improve the efficacy of BCG, different strategies are being implemented by considering the immunogenicity of various Mtb virulence factors as well as of the recombinant strains, co-administration with adjuvants and use of appropriate vehicle for delivery. This chapter discusses several such pre-clinical attempts to boost BCG with subunit vaccines tested in murine models and also highlights various recombinant TB vaccines undergoing clinical trials. Promising candidates include new generation of live recombinant BCG (rBCG) vaccines, VPM1002, which are deleted in one or two virulence genes. They encode for the mycobacteria-infected macrophage-inhibitor proteins of host macrophage apoptosis and autophagy, key events in killing and eradication of Mtb. These vaccines are rBCG- ΔureC::hly HMR, and rBCG-ΔureC::hly ΔnuoG. The former vaccine has passed phase IIb in clinical trials involving South African infants and adults. Thus, with an aim of elimination of TB by 2050, all these cumulative efforts to develop a better TB vaccine possibly is new hope for positive outcomes.
Collapse
Affiliation(s)
- Janez Ferluga
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Sanjib Bhakta
- Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
129
|
Macrophage metabolic reprogramming during chronic lung disease. Mucosal Immunol 2021; 14:282-295. [PMID: 33184475 PMCID: PMC7658438 DOI: 10.1038/s41385-020-00356-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 10/24/2020] [Indexed: 02/04/2023]
Abstract
Airway macrophages (AMs) play key roles in the maintenance of lung immune tolerance. Tissue tailored, highly specialised and strategically positioned, AMs are critical sentinels of lung homoeostasis. In the last decade, there has been a revolution in our understanding of how metabolism underlies key macrophage functions. While these initial observations were made during steady state or using in vitro polarised macrophages, recent studies have indicated that during many chronic lung diseases (CLDs), AMs adapt their metabolic profile to fit their local niche. By generating reactive oxygen species (ROS) for pathogen defence, utilising aerobic glycolysis to rapidly generate cytokines, and employing mitochondrial respiration to fuel inflammatory responses, AMs utilise metabolic reprogramming for host defence, although these changes may also support chronic pathology. This review focuses on how metabolic alterations underlie AM phenotype and function during CLDs. Particular emphasis is given to how our new understanding of AM metabolic plasticity may be exploited to develop AM-focused therapies.
Collapse
|
130
|
Lim K, Haider A, Adams C, Sleigh A, Savage DB. Lipodistrophy: a paradigm for understanding the consequences of "overloading" adipose tissue. Physiol Rev 2020; 101:907-993. [PMID: 33356916 DOI: 10.1152/physrev.00032.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lipodystrophies have been recognized since at least the nineteenth century and, despite their rarity, tended to attract considerable medical attention because of the severity and somewhat paradoxical nature of the associated metabolic disease that so closely mimics that of obesity. Within the last 20 yr most of the monogenic subtypes have been characterized, facilitating family genetic screening and earlier disease detection as well as providing important insights into adipocyte biology and the systemic consequences of impaired adipocyte function. Even more recently, compelling genetic studies have suggested that subtle partial lipodystrophy is likely to be a major factor in prevalent insulin-resistant type 2 diabetes mellitus (T2DM), justifying the longstanding interest in these disorders. This progress has also underpinned novel approaches to treatment that, in at least some patients, can be of considerable therapeutic benefit.
Collapse
Affiliation(s)
- Koini Lim
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Afreen Haider
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Claire Adams
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Alison Sleigh
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - David B Savage
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
131
|
Syed Najmuddin SUF, Amin ZM, Tan SW, Yeap SK, Kalyanasundram J, Veerakumarasivam A, Chan SC, Chia SL, Yusoff K, Alitheen NB. Oncolytic effects of the recombinant Newcastle disease virus, rAF-IL12, against colon cancer cells in vitro and in tumor-challenged NCr-Foxn1nu nude mice. PeerJ 2020; 8:e9761. [PMID: 33354412 PMCID: PMC7731658 DOI: 10.7717/peerj.9761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Colon cancer remains one of the main cancers causing death in men and women worldwide as certain colon cancer subtypes are resistant to conventional treatments and the development of new cancer therapies remains elusive. Alternative modalities such as the use of viral-based therapeutic cancer vaccine is still limited, with only the herpes simplex virus (HSV) expressing granulocyte-macrophage colony- stimulating factor (GM-CSF) or talimogene laherparepvec (T-Vec) being approved in the USA and Europe so far. Therefore, it is imperative to continue the search for a new treatment modality. This current study evaluates a combinatorial therapy between the oncolytic Newcastle disease virus (NDV) and interleukin-12 (IL-12) cytokine as a potential therapeutic vaccine to the current anti-cancer drugs. Several in vitro analyses such as MTT assay, Annexin V/FITC flow cytometry, and cell cycle assay were performed to evaluate the cytotoxicity effect of recombinant NDV, rAF-IL12. Meanwhile, serum cytokine, serum biochemical, histopathology of organs and TUNEL assay were carried out to assess the anti-tumoral effects of rAF-IL12 in HT29 tumor-challenged nude mice. The apoptosis mechanism underlying the effect of rAF-IL12 treatment was also investigated using NanoString Gene expression analysis. The recombinant NDV, rAF-IL12 replicated in HT29 colon cancer cells as did its parental virus, AF2240-i. The rAF-IL12 treatment had slightly better cytotoxicity effects towards HT29 cancer cells when compared to the AF2240-i as revealed by the MTT, Annexin V FITC and cell cycle assay. Meanwhile, the 28-day treatment with rAF-IL12 had significantly (p < 0.05) perturbed the growth and progression of HT29 tumor in NCr-Foxn1nu nude mice when compared to the untreated and parental wild-type NDV strain AF2240-i. The rAF-IL12 also modulated the immune system in nude mice by significantly (p < 0.05) increased the level of IL-2, IL-12, and IFN-γ cytokines. Treatment with rAF-IL12 had also significantly (p < 0.05) increased the expression level of apoptosis-related genes such as Fas, caspase-8, BID, BAX, Smad3 and granzyme B in vitro and in vivo. Besides, rAF-IL12 intra-tumoral delivery was considered safe and was not hazardous to the host as evidenced in pathophysiology of the normal tissues and organs of the mice as well as from the serum biochemistry profile of liver and kidney. Therefore, this study proves that rAF-IL12 had better cytotoxicity effects than its parental AF2240-i and could potentially be an ideal treatment for colon cancer in the near future.
Collapse
Affiliation(s)
| | - Zahiah Mohamed Amin
- Universiti Putra Malaysia, Serdang, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sheau Wei Tan
- Universiti Putra Malaysia, Serdang, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | | | | | | | | | - Suet Lin Chia
- Universiti Putra Malaysia, Serdang, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Khatijah Yusoff
- Universiti Putra Malaysia, Serdang, Malaysia.,Malaysian Genome Institute, National Institute of Biotechnology Malaysia, Kajang, Malaysia
| | - Noorjahan Banu Alitheen
- Universiti Putra Malaysia, Serdang, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
132
|
Priyam M, Gupta SK, Sarkar B, Sharma TR, Pattanayak A. Variation in selection constraints on teleost TLRs with emphasis on their repertoire in the Walking catfish, Clarias batrachus. Sci Rep 2020; 10:21394. [PMID: 33288798 PMCID: PMC7721727 DOI: 10.1038/s41598-020-78347-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/24/2020] [Indexed: 11/10/2022] Open
Abstract
The high degree of conservation of toll-like receptors (TLRs), and yet their subtle variations for better adaptation of species in the host–pathogen arms race make them worthy candidates for understanding evolution. We have attempted to track the trend of TLR evolution in the most diverse vertebrate group—teleosts, where Clarias batrachus was given emphasis, considering its traits for terrestrial adaptation. Eleven C. batrachus TLRs (TLR1, 2, 3, 5, 7, 8 9, 13, 22, 25, 26) were identified in this study which clustered in proximity to its Siluriformes relative orthologues in the phylogenetic analysis of 228 TLRs from 25 teleosts. Ten TLRs (TLR1, 2, 3, 5, 7, 8 9, 13, 21, 22) with at least 15 member orthologues for each alignment were processed for selection pressure and coevolutionary analysis. TLR1, 7, 8 and 9 were found to be under positive selection in the alignment-wide test. TLR1 also showed maximum episodic diversification in its clades while the teleost group Eupercaria showed the maximum divergence in their TLR repertoire. Episodic diversification was evident in C. batrachus TLR1 and 7 alignments. These results present a strong evidence of a divergent TLR repertoire in teleosts which may be contributing towards species-specific variation in TLR functions.
Collapse
Affiliation(s)
- Manisha Priyam
- School of Molecular Diagnostics and Prophylactics, ICAR-Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, 834 010, India
| | - Sanjay K Gupta
- School of Molecular Diagnostics and Prophylactics, ICAR-Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, 834 010, India.
| | - Biplab Sarkar
- School of Molecular Diagnostics and Prophylactics, ICAR-Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, 834 010, India
| | - T R Sharma
- School of Molecular Diagnostics and Prophylactics, ICAR-Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, 834 010, India
| | - A Pattanayak
- School of Molecular Diagnostics and Prophylactics, ICAR-Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, 834 010, India
| |
Collapse
|
133
|
Watanabe S, Iwata Y, Fukushima H, Saito K, Tanaka Y, Hasegawa Y, Akiyama M, Sugiura K. Neutrophil extracellular traps are induced in a psoriasis model of interleukin-36 receptor antagonist-deficient mice. Sci Rep 2020; 10:20149. [PMID: 33214582 PMCID: PMC7678853 DOI: 10.1038/s41598-020-76864-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 11/03/2020] [Indexed: 01/15/2023] Open
Abstract
Loss-of-function mutations in the interleukin (IL)-36 gene IL36RN are associated with psoriasis. The importance of neutrophil extracellular traps (NETs), web-like structures composed of neutrophil DNA, in the pathogenesis of psoriasis has been unclear. Here, we aimed to clarify the role of NET signaling in the deficiency of IL36 receptor antagonist (DITRA). We evaluated the severity of psoriasis-like lesions induced by imiquimod cream treatment in Il36rn-/- mice. The mRNA levels of psoriasis-related cytokines were measured via real-time reverse transcription polymerase chain reaction, and the effects of Cl-amidine, a peptidyl arginine deiminase 4 (PAD4) inhibitor, on psoriasis-like lesions were evaluated. PAD4 is a histone-modifying enzyme that is involved in NET formation. Psoriasis area and severity index scores, epidermal thickness, and infiltrated neutrophil counts were significantly increased in Il36rn-/- mice; NET formation was confirmed pathologically. Several cytokines and chemokines were upregulated in the skin lesions of Il36rn-/- mice and Cl-amidine treatment improved these psoriasis-like lesions. These results suggest that NET formation plays an important role in the pathology of psoriasis-like lesions in these mice and might represent a promising therapeutic target for DITRA.
Collapse
Affiliation(s)
- Soichiro Watanabe
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hidehiko Fukushima
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kenta Saito
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yoshihito Tanaka
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yurie Hasegawa
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
134
|
Lenders V, Koutsoumpou X, Sargsian A, Manshian BB. Biomedical nanomaterials for immunological applications: ongoing research and clinical trials. NANOSCALE ADVANCES 2020; 2:5046-5089. [PMID: 36132021 PMCID: PMC9418019 DOI: 10.1039/d0na00478b] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/22/2020] [Indexed: 05/04/2023]
Abstract
Research efforts on nanomaterial-based therapies for the treatment of autoimmune diseases and cancer have spiked and have made rapid progress over the past years. Nanomedicine has been shown to contribute significantly to overcome current therapeutic limitations, exhibiting advantages compared to conventional therapeutics, such as sustained drug release, delayed drug degradation and site-specific drug delivery. Multiple nanodrugs have reached the clinic, but translation is often hampered by either low targeting efficiency or undesired side effects. Nanomaterials, and especially inorganic nanoparticles, have gained criticism due to their potential toxic effects, including immunological alterations. However, many strategies have been attempted to improve the therapeutic efficacy of nanoparticles and exploit their unique properties for the treatment of inflammation and associated diseases. In this review, we elaborate on the immunomodulatory effects of nanomaterials, with a strong focus on the underlying mechanisms that lead to these specific immune responses. Nanomaterials to be discussed include inorganic nanoparticles such as gold, silica and silver, as well as organic nanomaterials such as polymer-, dendrimer-, liposomal- and protein-based nanoparticles. Furthermore, various approaches for tuning nanomaterials in order to enhance their efficacy and attenuate their immune stimulation or suppression, with respect to the therapeutic application, are described. Additionally, we illustrate how the acquired insights have been used to design immunotherapeutic strategies for a variety of diseases. The potential of nanomedicine-based therapeutic strategies in immunotherapy is further illustrated by an up to date overview of current clinical trials. Finally, recent efforts into enhancing immunogenic cell death through the use of nanoparticles are discussed.
Collapse
Affiliation(s)
- Vincent Lenders
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Xanthippi Koutsoumpou
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Ara Sargsian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| | - Bella B Manshian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven B-3000 Leuven Belgium
| |
Collapse
|
135
|
Schmidt CK, Medina-Sánchez M, Edmondson RJ, Schmidt OG. Engineering microrobots for targeted cancer therapies from a medical perspective. Nat Commun 2020; 11:5618. [PMID: 33154372 PMCID: PMC7645678 DOI: 10.1038/s41467-020-19322-7] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Systemic chemotherapy remains the backbone of many cancer treatments. Due to its untargeted nature and the severe side effects it can cause, numerous nanomedicine approaches have been developed to overcome these issues. However, targeted delivery of therapeutics remains challenging. Engineering microrobots is increasingly receiving attention in this regard. Their functionalities, particularly their motility, allow microrobots to penetrate tissues and reach cancers more efficiently. Here, we highlight how different microrobots, ranging from tailor-made motile bacteria and tiny bubble-propelled microengines to hybrid spermbots, can be engineered to integrate sophisticated features optimised for precision-targeting of a wide range of cancers. Towards this, we highlight the importance of integrating clinicians, the public and cancer patients early on in the development of these novel technologies.
Collapse
Affiliation(s)
- Christine K Schmidt
- Manchester Cancer Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester, M20 4GJ, UK.
| | - Mariana Medina-Sánchez
- Institute for Integrative Nanosciences, Leibniz IFW Dresden, Helmholtzstraße 20, 01069, Dresden, Germany.
| | - Richard J Edmondson
- Gynaecological Oncology, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- St. Mary's Hospital, Central Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Level 5, Research Floor, Oxford Road, Manchester, M13 9WL, UK
| | - Oliver G Schmidt
- Institute for Integrative Nanosciences, Leibniz IFW Dresden, Helmholtzstraße 20, 01069, Dresden, Germany.
| |
Collapse
|
136
|
Wong LW, Ong KS, Khoo JR, Goh CBS, Hor JW, Lee SM. Human intestinal parasitic infection: a narrative review on global prevalence and epidemiological insights on preventive, therapeutic and diagnostic strategies for future perspectives. Expert Rev Gastroenterol Hepatol 2020; 14:1093-1105. [PMID: 32755242 DOI: 10.1080/17474124.2020.1806711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Intestinal parasitic infection (IPI) is a global health concern among socioeconomically deprived communities in many developing countries. Many preventative strategies have been deployed to control IPI, however, there is a lack in standards on the techniques used to diagnose and monitor the prevalence of IPI. AREAS COVERED The present article will review the diseases associated with IPI and discuss the current IPI control strategies such as the water, sanitation, and hygiene (WASH) interventions, community-led total sanitation (CLTS) approach, and regular anthelminthic treatments. For the first time, this review will also evaluate all currently practised diagnostic techniques for the detection of intestinal parasites and provide insights on future IPI control strategies. EXPERT OPINION Advanced and improved diagnostic methods such as qPCR coupled with a high-resolution melting curve, aptamers, biosensors, and detection of extracellular vesicles can be used for detection of IPI. Vaccination against intestinal parasites can be made available to increase antibodies to interfere with the blood-feeding process by the parasites, which subsequently reduces the reproductive rates of the parasites. These methods collectively can serve as future management strategies for intestinal parasitic infections.
Collapse
Affiliation(s)
- Li Wen Wong
- School of Science, Monash University Malaysia , Bandar Sunway, Malaysia
| | - Kuan Shion Ong
- School of Science, Monash University Malaysia , Bandar Sunway, Malaysia.,Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia , Bandar Sunway, Malaysia
| | - Jun Rong Khoo
- School of Science, Monash University Malaysia , Bandar Sunway, Malaysia
| | - Calvin Bok Sun Goh
- School of Science, Monash University Malaysia , Bandar Sunway, Malaysia.,Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia , Bandar Sunway, Malaysia
| | - Jia Wei Hor
- Department of Medicine, Faculty of Medicine, University of Malaya , Kuala Lumpur, Malaysia
| | - Sui Mae Lee
- School of Science, Monash University Malaysia , Bandar Sunway, Malaysia.,Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia , Bandar Sunway, Malaysia
| |
Collapse
|
137
|
Larabi A, Dalmasso G, Delmas J, Barnich N, Nguyen HTT. Exosomes transfer miRNAs from cell-to-cell to inhibit autophagy during infection with Crohn's disease-associated adherent-invasive E. coli. Gut Microbes 2020; 11:1677-1694. [PMID: 32583714 PMCID: PMC7524154 DOI: 10.1080/19490976.2020.1771985] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Adherent-invasive E. coli (AIEC), which abnormally colonize the intestinal mucosa of Crohn's disease (CD) patients, are able to adhere to and invade intestinal epithelial cells (IECs), survive and replicate within macrophages and induce a pro-inflammatory response. AIEC infection of IECs induces secretion of exosomes that increase AIEC replication in exosome-receiving IECs and macrophages. Here, we investigated the mechanism underlying the increased AIEC replication in cells receiving exosomes from AIEC-infected cells. Exosomes released by uninfected human intestinal epithelial T84 cells (Exo-uninfected) or by T84 cells infected with the clinical AIEC LF82 strain (Exo-LF82), the nonpathogenic E. coli K12 strain (Exo-K12) or the commensal E. coli HS strain (Exo-HS) were purified and used to stimulate T84 cells. Stimulation of T84 cells with Exo-LF82 inhibited autophagy compared with Exo-uninfected, Exo-K12 and Exo-HS. qRT-PCR analysis revealed increased levels of miR-30c and miR-130a in Exo-LF82 compared to Exo-uninfected, Exo-K12 and Exo-HS. These miRNAs were transferred via exosomes to recipient cells, in which they targeted and inhibited ATG5 and ATG16L1 expression and thereby autophagy response, thus favoring AIEC intracellular replication. Inhibition of these miRNAs in exosome-donor cells infected with AIEC LF82 abolished the increase in miR-30c and miR-130a levels in the released Exo-LF82 and in Exo-LF82-receiving cells, thus suppressing the inhibitory effect of Exo-LF82 on ATG5 and ATG16L1 expression and on autophagy-mediated AIEC clearance in Exo-LF82-receiving cells. Our study shows that upon AIEC infection, IECs secrete exosomes that can transfer specific miRNAs to recipient IECs, inhibiting autophagy-mediated clearance of intracellular AIEC.
Collapse
Affiliation(s)
- Anaïs Larabi
- M2iSH, UMR 1071 Inserm, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Guillaume Dalmasso
- M2iSH, UMR 1071 Inserm, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Julien Delmas
- M2iSH, UMR 1071 Inserm, Université Clermont Auvergne, Clermont-Ferrand, France
- Service de Bactériologie, Centre Hospitalier Universitaire (CHU) Gabriel Montpied, Clermont-Ferrand, France
| | - Nicolas Barnich
- M2iSH, UMR 1071 Inserm, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Hang Thi Thu Nguyen
- M2iSH, UMR 1071 Inserm, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
138
|
Moore EM, Maestas DR, Comeau HY, Elisseeff JH. The Immune System and Its Contribution to Variability in Regenerative Medicine. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:39-47. [PMID: 32635878 DOI: 10.1089/ten.teb.2019.0335] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The immune system plays a critical role in directing tissue repair and regeneration outcomes. Tissue engineering technologies that are designed to promote new tissue growth will therefore be impacted by immune factors that are present in patients both locally at the site of intervention and systemically. The immune state of patients can be influenced by many factors, including infection, nutrition, and other disease comorbidities. As a result, the immune state is highly variable and may be a source of variability in tissue-engineered products in the clinic, which is not found in preclinical models. In this review, we will summarize key immune cells and evidence of their activity in tissue repair and potential in tissue engineering systems. We also discuss how clinical translation of tissue engineering strategies, in particular stem cells, helped elucidate the importance of the immune system. With increased understanding of the immune system's role in repair and tissue engineering systems, it will likely become a therapeutic target and component of future therapies.
Collapse
Affiliation(s)
- Erika M Moore
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida, USA
| | - David R Maestas
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hannah Y Comeau
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
139
|
Krayem I, Lipoldová M. Role of host genetics and cytokines in Leishmania infection. Cytokine 2020; 147:155244. [PMID: 33059974 DOI: 10.1016/j.cyto.2020.155244] [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: 05/22/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 12/29/2022]
Abstract
Cytokines and chemokines are important regulators of innate and specific responses in leishmaniasis, a disease that currently affects 12 million people. We overviewed the current information about influences of genetically engineered mouse models of cytokine and chemokine on leishmaniasis. We found that genetic background of the host, parasite species and sub-strain, as well as experimental design often modify effects of genetically engineered cytokine genes. Next we analyzed genes and QTLs (quantitative trait loci) that control response to Leishmania species in mouse in order to establish relationship between genetic control of cytokine expression and organ pathology. These studies revealed a network-like complexity of the combined effects of the multiple functionally diverse QTLs and their individual specificity. Genetic control of organ pathology and systemic immune response overlap only partially. Some QTLs control both organ pathology and systemic immune response, but the effects of genes and loci with the strongest impact on disease are cytokine-independent, whereas several loci modify cytokines levels in serum without influencing organ pathology. Understanding this genetic control might be important in development of vaccines designed to stimulate certain cytokine spectrum.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; Department of Natural Sciences, Faculty of Biomedical Engineering, Czech Technical University in Prague, Sítná 3105, 272 01 Kladno, Czech Republic.
| |
Collapse
|
140
|
Baillache DJ, Unciti-Broceta A. Recent developments in anticancer kinase inhibitors based on the pyrazolo[3,4- d]pyrimidine scaffold. RSC Med Chem 2020; 11:1112-1135. [PMID: 33479617 PMCID: PMC7652001 DOI: 10.1039/d0md00227e] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/01/2020] [Indexed: 12/24/2022] Open
Abstract
Pyrazolo[3,4-d]pyrimidines have become of significant interest for the medicinal chemistry community as a privileged scaffold for the development of kinase inhibitors to treat a range of diseases, including cancer. This fused nitrogen-containing heterocycle is an isostere of the adenine ring of ATP, allowing the molecules to mimic hinge region binding interactions in kinase active sites. Similarities in kinase ATP sites can be exploited to direct the activity and selectivity of pyrazolo[3,4-d]pyrimidines to multiple oncogenic targets through focussed chemical modification. As a result, pharma and academic efforts have succeeded in progressing several pyrazolo[3,4-d]pyrimidines to clinical trials, including the BTK inhibitor ibrutinib, which has been approved for the treatment of several B-cell cancers. In this review, we examine the pyrazolo[3,4-d]pyrimidines currently in clinical trials for oncology patients, as well as those published in the literature during the last 5 years for different anticancer indications.
Collapse
Affiliation(s)
- Daniel J Baillache
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre , Institute of Genetics and Molecular Medicine , University of Edinburgh , Crewe Road South , Edinburgh EH4 2XR , UK .
| |
Collapse
|
141
|
Jorgovanovic D, Song M, Wang L, Zhang Y. Roles of IFN-γ in tumor progression and regression: a review. Biomark Res 2020; 8:49. [PMID: 33005420 PMCID: PMC7526126 DOI: 10.1186/s40364-020-00228-x] [Citation(s) in RCA: 566] [Impact Index Per Article: 141.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Interferon-γ (IFN-γ) plays a key role in activation of cellular immunity and subsequently, stimulation of antitumor immune-response. Based on its cytostatic, pro-apoptotic and antiproliferative functions, IFN-γ is considered potentially useful for adjuvant immunotherapy for different types of cancer. Moreover, it IFN-γ may inhibit angiogenesis in tumor tissue, induce regulatory T-cell apoptosis, and/or stimulate the activity of M1 proinflammatory macrophages to overcome tumor progression. However, the current understanding of the roles of IFN-γ in the tumor microenvironment (TME) may be misleading in terms of its clinical application. MAIN BODY Some researchers believe it has anti-tumorigenic properties, while others suggest that it contributes to tumor growth and progression. In our recent work, we have shown that concentration of IFN-γ in the TME determines its function. Further, it was reported that tumors treated with low-dose IFN-γ acquired metastatic properties while those infused with high dose led to tumor regression. Pro-tumorigenic role may be described through IFN-γ signaling insensitivity, downregulation of major histocompatibility complexes, upregulation of indoleamine 2,3-dioxygenase, and checkpoint inhibitors such as programmed cell death ligand 1. CONCLUSION Significant research efforts are required to decipher IFN-γ-dependent pro- and anti-tumorigenic effects. This review discusses the current knowledge concerning the roles of IFN-γ in the TME as a part of the complex immune response to cancer and highlights the importance of identifying IFN-γ responsive patients to improve their sensitivity to immuno-therapies.
Collapse
Affiliation(s)
- Dragica Jorgovanovic
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052 Henan China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052 China
| | - Mengjia Song
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, 510060 China
| | - Liping Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052 Henan China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052 Henan China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052 China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052 Henan China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, 450052 China
| |
Collapse
|
142
|
Mxinwa V, Dludla PV, Nyambuya TM, Mokgalaboni K, Mazibuko-Mbeje SE, Nkambule BB. Natural killer cell levels in adults living with type 2 diabetes: a systematic review and meta-analysis of clinical studies. BMC Immunol 2020; 21:51. [PMID: 32907543 PMCID: PMC7487809 DOI: 10.1186/s12865-020-00378-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic immune activation and hyperglycaemia are a hallmark of type 2 diabetes mellitus (T2D) while natural killer (NK) cells are involved in the pathogenesis of T2D. Dysregulated NK cell responses are associated with an increased risk of cardiovascular disease in patients living with T2D. OBJECTIVE To provide a comprehensive and systematic evidence-based estimate on the levels of NK cells in patients living with T2D. RESULTS This systematic review and meta-analysis included 13 studies reporting on 491 adult patients with T2D and 1064 nondiabetic controls. The pooled effect estimates showed increased levels of NK cells in adult patients with T2D compared to controls (MD: 0.03 [- 3.20, 3.26], I2 = 97%, p < 0.00001). CONCLUSION Overall, the evidence presented in this systematic review shows that the changes in NK cells in patients living with T2D are still unclear and further studies are needed.
Collapse
Affiliation(s)
- Vuyolwethu Mxinwa
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000 South Africa
| | - Phiwayinkosi V. Dludla
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
- Biomedical Research and Innovation Platform, Medical Research Council, Tygerberg, South Africa
| | - Tawanda M. Nyambuya
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000 South Africa
- Department of Health Sciences, Faculty of Health and Applied Sciences, Namibia University of Science and Technology, Windhoek, 9000 Namibia
| | - Kabelo Mokgalaboni
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000 South Africa
| | - Sithandiwe E. Mazibuko-Mbeje
- Biomedical Research and Innovation Platform, Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, 4000 South Africa
| |
Collapse
|
143
|
Boros F, Vécsei L. Progress in the development of kynurenine and quinoline-3-carboxamide-derived drugs. Expert Opin Investig Drugs 2020; 29:1223-1247. [DOI: 10.1080/13543784.2020.1813716] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Fanni Boros
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Center, Faculty of Medicine, University of Szeged, Szeged, Hungary
- MTA-SZTE Neuroscience Research Group of the Hungarian Academy of Sciences and the University of Szeged, Szeged, Hungary
- Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
144
|
Takayasu T, Shah M, Dono A, Yan Y, Borkar R, Putluri N, Zhu JJ, Hama S, Yamasaki F, Tahara H, Sugiyama K, Kurisu K, Esquenazi Y, Ballester LY. Cerebrospinal fluid ctDNA and metabolites are informative biomarkers for the evaluation of CNS germ cell tumors. Sci Rep 2020; 10:14326. [PMID: 32868820 PMCID: PMC7459305 DOI: 10.1038/s41598-020-71161-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
Serum and cerebrospinal fluid (CSF) levels of α-fetoprotein and β-subunit of human chorionic gonadotropin are used as biomarkers for the management of central nervous system (CNS) germ cell tumors (GCTs). However, additional discriminating biomarkers are required. Especially, biomarkers to differentiate non-germinomatous germ cell tumors (NGGCTs) from germinomas are critical, as these have a distinct prognosis. We investigated CSF samples from 12 patients with CNS-GCT patients (8 germinomas and 4 NGGCTs). We analyzed circulating tumor DNA (ctDNA) in CSF to detect mutated genes. We also used liquid chromatography-mass spectrometry to characterize metabolites in CSF. We detected KIT and/or NRAS mutation, known as frequently mutated genes in GCTs, in 3/12 (25%) patients. We also found significant differences in the abundance of 15 metabolites between control and GCT, with unsupervised hierarchical clustering analysis. Metabolites related to the TCA cycle were increased in GCTs. Urea, ornithine, and short-chain acylcarnitines were decreased in GCTs. Moreover, we also detected several metabolites (e.g., betaine, guanidine acetic acid, and 2-aminoheptanoic acid) that displayed significant differences in abundance in patients with germinomas and NGGCTs. Our results suggest that ctDNA and metabolites in CSF can serve as novel biomarkers for CNS-GCTs and can be useful to differentiate germinomas from NGGCTs.
Collapse
Affiliation(s)
- Takeshi Takayasu
- Department of Pathology and Laboratory Medicine, Molecular Genetic Pathology and Neuropathology, The University of Texas Health Science Center, 6431 Fannin St., MSB 2.136, Houston, TX, 77030, USA.,Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ward, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Mauli Shah
- Department of Pathology and Laboratory Medicine, Molecular Genetic Pathology and Neuropathology, The University of Texas Health Science Center, 6431 Fannin St., MSB 2.136, Houston, TX, 77030, USA
| | - Antonio Dono
- Vivian L. Smith Department of Neurosurgery, UTHealth McGovern Medical School, the University of Texas Health Science Center, Houston, TX, USA
| | - Yuanqing Yan
- Vivian L. Smith Department of Neurosurgery, UTHealth McGovern Medical School, the University of Texas Health Science Center, Houston, TX, USA
| | - Roshan Borkar
- Metabolomics Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Metabolomics Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, UTHealth McGovern Medical School, the University of Texas Health Science Center, Houston, TX, USA.,Memorial Hermann Hospital-TMC, Houston, TX, USA
| | - Seiji Hama
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ward, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Fumiyuki Yamasaki
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ward, Hiroshima City, Hiroshima, 734-8551, Japan.
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology and Neuro-Oncology Program, Hiroshima University Hospital, Hiroshima City, Hiroshima, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ward, Hiroshima City, Hiroshima, 734-8551, Japan
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, UTHealth McGovern Medical School, the University of Texas Health Science Center, Houston, TX, USA.,Memorial Hermann Hospital-TMC, Houston, TX, USA.,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, USA
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, Molecular Genetic Pathology and Neuropathology, The University of Texas Health Science Center, 6431 Fannin St., MSB 2.136, Houston, TX, 77030, USA. .,Vivian L. Smith Department of Neurosurgery, UTHealth McGovern Medical School, the University of Texas Health Science Center, Houston, TX, USA. .,Memorial Hermann Hospital-TMC, Houston, TX, USA.
| |
Collapse
|
145
|
Zanwar S, Nandakumar B, Kumar S. Immune-based therapies in the management of multiple myeloma. Blood Cancer J 2020; 10:84. [PMID: 32829378 PMCID: PMC7443188 DOI: 10.1038/s41408-020-00350-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a clonal plasma cell malignancy affecting a predominantly elderly population. The continued development of newer therapies with novel mechanisms of action has reshaped the treatment paradigm of this disorder in the last two decades, leading to a significantly improved prognosis. This has in turn resulted in an increasing number of patients in need of therapy for relapsed/refractory disease. Immune-based therapies, including monoclonal antibodies, immune checkpoint inhibitors, and most promisingly, adoptive cellular therapies represent important therapeutic strategies in these patients due to their non-cross resistant mechanisms of actions with the usual frontline therapies comprising of immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs). The anti-CD38 antibodies daratumumab and more recently isatuximab, with their excellent efficacy and safety profile along with its synergy in combination with IMiDs and PIs, are being increasingly incorporated in the frontline setting. Chimeric antigen receptor-T cell (CART) therapies and bi-specific T-cell engager (BiTE) represent exciting new options that have demonstrated efficacy in heavily pretreated and refractory MM. In this review, we discuss the rationale for use of immune-based therapies in MM and summarize the currently available literature for common antibodies and CAR-T therapies that are utilized in MM.
Collapse
Affiliation(s)
- Saurabh Zanwar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
146
|
Das A, Dasgupta S, Pathak T. Design of configuration-restricted triazolylated β-d-ribofuranosides: a unique family of crescent-shaped RNase A inhibitors. Org Biomol Chem 2020; 18:6340-6356. [PMID: 32766618 DOI: 10.1039/d0ob01286f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Seven new carbohydrate-bistriazole hybrid molecules were designed taking into consideration the crescent shaped active site of ribonuclease A (RNase A). In this case, the β-d-ribofuranose structure was used as the basic building unit; both the C1 and C4 arms protruding out towards the β-face of the tetrahydrofuran moiety of the ribose sugar provided an overall "U" shape to the basic building block. Several combinations of bistriazole moieties were constructed on the two arms of this basic building block. These mono- and/or bis-substituted 1,2,3-triazole units were linked to acidic functional groups because of the overall basic nature of the hydrolytic site of RNase A. All these compounds were efficient competitive inhibitors of RNase A with inhibition constants (Ki) in the micromolar range. In contrast to the carboxylic acid-modified hybrid molecules, molecules carrying sulfonic acids were found to be more potent because of the stronger interactions with the positively charged active site. The most efficient inhibitor of the series was the disulfonic acid-functionalized carbohydrate-bis-triazole hybrid molecule. Docking studies disclosed that the molecule, because of its well defined "U" shape with flexible arms, fits effectively in the active site; moreover, in all cases, besides the acid groups, the triazole and sugar rings also actively participated in creating the hydrogen bonding network in the cavity of the enzyme active site.
Collapse
Affiliation(s)
- Ashrukana Das
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721 302, West Bengal, India.
| | - Swagata Dasgupta
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721 302, West Bengal, India.
| | - Tanmaya Pathak
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721 302, West Bengal, India.
| |
Collapse
|
147
|
Reeves PM, Raju Paul S, Baeten L, Korek SE, Yi Y, Hess J, Sobell D, Scholzen A, Garritsen A, De Groot AS, Moise L, Brauns T, Bowen R, Sluder AE, Poznansky MC. Novel multiparameter correlates of Coxiella burnetii infection and vaccination identified by longitudinal deep immune profiling. Sci Rep 2020; 10:13311. [PMID: 32770104 PMCID: PMC7414860 DOI: 10.1038/s41598-020-69327-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Q-fever is a flu-like illness caused by Coxiella burnetii (Cb), a highly infectious intracellular bacterium. There is an unmet need for a safe and effective vaccine for Q-fever. Correlates of immune protection to Cb infection are limited. We proposed that analysis by longitudinal high dimensional immune (HDI) profiling using mass cytometry combined with other measures of vaccination and protection could be used to identify novel correlates of effective vaccination and control of Cb infection. Using a vaccine-challenge model in HLA-DR transgenic mice, we demonstrated significant alterations in circulating T-cell and innate immune populations that distinguished vaccinated from naïve mice within 10 days, and persisted until at least 35 days post-vaccination. Following challenge, vaccinated mice exhibited reduced bacterial burden and splenomegaly, along with distinct effector T-cell and monocyte profiles. Correlation of HDI data to serological and pathological measurements was performed. Our data indicate a Th1-biased response to Cb, consistent with previous reports, and identify Ly6C, CD73, and T-bet expression in T-cell, NK-cell, and monocytic populations as distinguishing features between vaccinated and naïve mice. This study refines the understanding of the integrated immune response to Cb vaccine and challenge, which can inform the assessment of candidate vaccines for Cb.
Collapse
Affiliation(s)
- P M Reeves
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA.
| | - S Raju Paul
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - L Baeten
- Colorado State University, Fort Collins, CO, USA
| | - S E Korek
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - Y Yi
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - J Hess
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - D Sobell
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - A Scholzen
- InnatOss Laboratories B.V, Oss, The Netherlands
| | - A Garritsen
- InnatOss Laboratories B.V, Oss, The Netherlands
| | - A S De Groot
- EpiVax, Inc, Providence, RI, USA.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - L Moise
- EpiVax, Inc, Providence, RI, USA.,Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | - T Brauns
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - R Bowen
- Colorado State University, Fort Collins, CO, USA
| | - A E Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA
| | - M C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
148
|
Van Den Eeckhout B, Van Hoecke L, Burg E, Van Lint S, Peelman F, Kley N, Uzé G, Saelens X, Tavernier J, Gerlo S. Specific targeting of IL-1β activity to CD8 + T cells allows for safe use as a vaccine adjuvant. NPJ Vaccines 2020; 5:64. [PMID: 32714571 PMCID: PMC7378068 DOI: 10.1038/s41541-020-00211-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 06/19/2020] [Indexed: 11/08/2022] Open
Abstract
Annual administration and reformulation of influenza vaccines is required for protection against seasonal infections. However, the induction of strong and long-lasting T cells is critical to reach broad and potentially lifelong antiviral immunity. The NLRP3 inflammasome and its product interleukin-1β (IL-1β) are pivotal mediators of cellular immune responses to influenza, yet, overactivation of these systems leads to side effects, which hamper clinical applications. Here, we present a bypass around these toxicities by targeting the activity of IL-1β to CD8+ T cells. Using this approach, we demonstrate safe inclusion of IL-1β as an adjuvant in vaccination strategies, leading to full protection of mice against a high influenza virus challenge dose by raising potent T cell responses. In conclusion, this paper proposes a class of IL-1β-based vaccine adjuvants and also provides further insight in the mechanics of cellular immune responses driven by IL-1β.
Collapse
Affiliation(s)
- Bram Van Den Eeckhout
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lien Van Hoecke
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Elianne Burg
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Sandra Van Lint
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Frank Peelman
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Niko Kley
- Orionis Biosciences Inc, Waltham, MA 02451 USA
| | - Gilles Uzé
- CNRS 5235, University of Montpellier, 34090 Montpellier, France
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Orionis Biosciences Inc, Waltham, MA 02451 USA
| | - Sarah Gerlo
- VIB-UGent Center for Medical Biotechnology, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
149
|
Finke H, Winkelbeiner N, Lossow K, Hertel B, Wandt VK, Schwarz M, Pohl G, Kopp JF, Ebert F, Kipp AP, Schwerdtle T. Effects of a Cumulative, Suboptimal Supply of Multiple Trace Elements in Mice: Trace Element Status, Genomic Stability, Inflammation, and Epigenetics. Mol Nutr Food Res 2020; 64:e2000325. [PMID: 32609929 DOI: 10.1002/mnfr.202000325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Indexed: 12/15/2022]
Abstract
SCOPE Trace element (TE) deficiencies often occur accumulated, as nutritional intake is inadequate for several TEs, concurrently. Therefore, the impact of a suboptimal supply of iron, zinc, copper, iodine, and selenium on the TE status, health parameters, epigenetics, and genomic stability in mice are studied. METHODS AND RESULTS Male mice receive reduced or adequate amounts of TEs for 9 weeks. The TE status is analyzed mass-spectrometrically in serum and different tissues. Furthermore, gene and protein expression of TE biomarkers are assessed with focus on liver. Iron concentrations are most sensitive toward a reduced supply indicated by increased serum transferrin levels and altered hepatic expression of iron-related genes. Reduced TE supply results in smaller weight gain but higher spleen and heart weights. Additionally, inflammatory mediators in serum and liver are increased together with hepatic genomic instability. However, global DNA (hydroxy)methylation is unaffected by the TE modulation. CONCLUSION Despite homeostatic regulation of most TEs in response to a low intake, this condition still has substantial effects on health parameters. It appears that the liver and immune system react particularly sensitive toward changes in TE intake. The reduced Fe status might be the primary driver for the observed effects.
Collapse
Affiliation(s)
- Hannah Finke
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany
| | - Nicola Winkelbeiner
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Kristina Lossow
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich-Schiller University Jena, Dornburger Straße 24, Jena, 07743, Germany.,German Institute of Human Nutrition, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Barbara Hertel
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany
| | - Viktoria K Wandt
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Maria Schwarz
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich-Schiller University Jena, Dornburger Straße 24, Jena, 07743, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany
| | - Johannes F Kopp
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Anna P Kipp
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich-Schiller University Jena, Dornburger Straße 24, Jena, 07743, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Germany.,TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany.,German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, Berlin, 10589, Germany
| |
Collapse
|
150
|
Menanteau-Ledouble S, Nöbauer K, Razzazi-Fazeli E, El-Matbouli M. Effects of Yersinia ruckeri invasion on the proteome of the Chinook salmon cell line CHSE-214. Sci Rep 2020; 10:11840. [PMID: 32678312 PMCID: PMC7366648 DOI: 10.1038/s41598-020-68903-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 07/03/2020] [Indexed: 12/18/2022] Open
Abstract
Yersinia ruckeri is an important bacterial pathogen of fish, in particular salmonids, it has been associated with systemic infections worldwide and, like many enteric bacteria, it is a facultative intracellular pathogen. However, the effect of Y. ruckeri's interactions with the host at the cellular level have received little investigation. In the present study, a culture of Chinook Salmon Embryo (CHSE) cell line was exposed to Y. ruckeri. Afterwards, the proteins were investigated and identified by mass spectrometry and compared to the content of unexposed cultures. The results of this comparison showed that 4.7% of the identified proteins were found at significantly altered concentrations following infection. Interestingly, infection with Y. ruckeri was associated with significant changes in the concentration of surface adhesion proteins, including a significantly decreased presence of β-integrins. These surface adhesion molecules are known to be the target for several adhesion molecules of Yersiniaceae. The concentration of several anti-apoptotic regulators (HSP90 and two DNAj molecules) appeared similarly downregulated. Taken together, these findings suggest that Y. ruckeri affects the proteome of infected cells in a notable manner and our results shed some light on the interaction between this important bacterial pathogen and its host.
Collapse
Affiliation(s)
- Simon Menanteau-Ledouble
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria.
| | - Katharina Nöbauer
- VetCore Facility for Research, University of Veterinary Medicine, Vienna, Austria
| | | | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| |
Collapse
|