99951
|
Lawrence SM, Ruoss JL, Wynn JL. IL-17 in neonatal health and disease. Am J Reprod Immunol 2017; 79:e12800. [PMID: 29243317 DOI: 10.1111/aji.12800] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/21/2017] [Indexed: 12/11/2022] Open
Abstract
Over the last few years, scientific interest in the cytokine IL-17A has intensified as its role in human health and disease has been elucidated. Discovered almost a quarter century ago, IL-17A is known to have poor biologic activity when acting alone, but attains robust actions when working synergistically with potent mediators of proinflammatory immune responses, such as IL-6 and IL-8. IL-17A is produced by specialized innate immune cells that protect host barriers from the outside world. Like sentries, these innate immune cells can "sound the alarm" through increased production of IL-17A, causing activation and recruitment of primed neutrophils and monocytes when pathogens escape initial host defenses. In this way, IL-17A promulgates mechanisms responsible for pathogen death and clearance. However, when IL-17A pathways are triggered during fetal development, due to chorioamnionitis or in utero inflammatory conditions, IL-17A can instigate and/or exacerbate fetal inflammatory responses that increase neonatal morbidities and mortality associated with common neonatal conditions such as sepsis, bronchopulmonary dysplasia (BPD), patent ductus arteriosus (PDA), and necrotizing enterocolitis (NEC). This review details the ontogeny of IL-17A in the fetus and newborn, discusses how derangements in its production can lead to pathology, and describes known and evolving therapies that may attenuate IL-17A-mediated human conditions.
Collapse
Affiliation(s)
- Shelley M Lawrence
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of California, San Diego, La Jolla, CA, USA.,Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Lauren Ruoss
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - James L Wynn
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
99952
|
Gut-Bioreactor and Human Health in Future. Indian J Microbiol 2017; 58:3-7. [PMID: 29434391 DOI: 10.1007/s12088-017-0697-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/09/2017] [Indexed: 12/22/2022] Open
Abstract
Gut-microbiome provides the complementary metabolic potential to the human system. To understand the active participation and the performance of the microbial community in human health, the concept of gut as a plug-flow reactor with the fed-batch mode of operation can provide better insight. The concept suggests the virtual compartmentalized gut with sequential stratification of the microbial community in response to a typical host genotype. It also provides the analysis plan for gut microbiome; and its relevance in developing health management options under the identified clinical conditions.
Collapse
|
99953
|
Stamenković B, Bojanović M, Stojanović S, Živković V, Nedović J, Jovanović J, Đorđević D. BONE MINERAL DENSITY IN FEMALE PATIENTS WITH SYSTEMIC SCLEROSIS AND DIFFERENT SEROLOGICAL STATUS OF DISEASE. ACTA MEDICA MEDIANAE 2017. [DOI: 10.5633/amm.2017.0417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
99954
|
Lee SH, Charmoy M, Romano A, Paun A, Chaves MM, Cope FO, Ralph DA, Sacks DL. Mannose receptor high, M2 dermal macrophages mediate nonhealing Leishmania major infection in a Th1 immune environment. J Exp Med 2017; 215:357-375. [PMID: 29247046 PMCID: PMC5748861 DOI: 10.1084/jem.20171389] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 11/10/2017] [Indexed: 12/24/2022] Open
Abstract
The origin and functional specialization of dermal macrophages in cutaneous infections have been little studied. In this paper, we show that a strain of Leishmania major (L. major Seidman [LmSd]) that produces nonhealing cutaneous lesions in conventionally resistant C57BL/6 mice was more efficiently taken up by M2-polarized bone marrow (BM)-derived macrophages (BMDMs) in vitro and by mannose receptor (MR)hi dermal macrophages in vivo compared with a healing strain (L. major Friedlin V1). Both in steady and in T helper type 1 (Th1) cell-driven inflammatory states, the MRhi dermal macrophages showed M2 characteristics. The dermal macrophages were radio resistant and not replaced by monocytes or adult BM-derived cells during infection, but were locally maintained by IL-4 and IL-10. Notably, the favored infection of M2 BMDMs by LmSd in vitro was MR dependent, and genetic deletion of MR or selective depletion of MRhi dermal macrophages by anti-CSF-1 receptor antibody reversed the nonhealing phenotype. We conclude that embryonic-derived, MRhi dermal macrophages are permissive for parasite growth even in a strong Th1-immune environment, and the preferential infection of these cells plays a crucial role in the severity of cutaneous disease.
Collapse
Affiliation(s)
- Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Melanie Charmoy
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Audrey Romano
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mariana M Chaves
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | - David L Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
99955
|
Mouri Y, Ueda Y, Yamano T, Matsumoto M, Tsuneyama K, Kinashi T, Matsumoto M. Mode of Tolerance Induction and Requirement for Aire Are Governed by the Cell Types That Express Self-Antigen and Those That Present Antigen. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:3959-3971. [PMID: 29101311 DOI: 10.4049/jimmunol.1700892] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
Aire controls the fate of autoreactive thymocytes (i.e., clonal deletion or development into regulatory T cells [Tregs]) through transcriptional control of the expression of tissue-restricted self-antigens (TRAs) from medullary thymic epithelial cells (mTECs) and bone marrow (BM)-derived cells. Although TRAs expressed by mTECs and BM-derived cells are suggested to complement each other to generate a full spectrum of TRAs, little is known about the relative contribution of TRAs from each component for establishment of self-tolerance. Furthermore, the precise role of Aire in specific types of Aire-expressing APCs remains elusive. We have approached these issues by generating two different types of transgenic mouse (Tg) model, which express a prefixed model self-antigen driven by the insulin promoter or the Aire promoter. In the insulin-promoter Tg model, mTECs alone were insufficient for clonal deletion, and BM-derived APCs were required for this action by utilizing Ag transferred from mTECs. In contrast, mTECs alone were able to induce Tregs, although at a much lower efficiency in the absence of BM-derived APCs. Importantly, lack of Aire in mTECs, but not in BM-derived APCs, impaired both clonal deletion and production of Tregs. In the Aire-promoter Tg model, both mTECs and BM-derived APCs could independently induce clonal deletion without Aire, and production of Tregs was impaired by the lack of Aire in mTECs, but not in BM-derived APCs. These results suggest that the fate of autoreactive thymocytes together with the requirement for Aire depend on the cell types that express self-antigens and the types of APCs involved in tolerance induction.
Collapse
Affiliation(s)
- Yasuhiro Mouri
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
| | - Yoshihiro Ueda
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Osaka 570-8506, Japan
| | - Tomoyoshi Yamano
- Institute for Immunology, Ludwig-Maximilians-University Munich, Munich 80336, Germany
| | - Minoru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan; and
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan; and
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Osaka 570-8506, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima 770-8503, Japan;
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Tokyo 100-0004, Japan
| |
Collapse
|
99956
|
Janeva-Jovanovska E, Dokic D, Jovkovska-Kaeva B, Breskovska G, Goseva Z, Minov J, Trajkov D, Dimitrova-Genadieva M, Zafirovska-Ivanovska B. Relationship between Vitamin D, Inflammation and Lung Function In Patients with Severe Uncontrolled Asthma. Open Access Maced J Med Sci 2017; 5:899-903. [PMID: 29362615 PMCID: PMC5771291 DOI: 10.3889/oamjms.2017.190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Recently epidemiological studies showed that low vitamin D is linked to airway hyperresponsiveness, decreased lung function, poor asthma control, and steroid-resistant asthma. AIM We investigated the relationship between Vitamin D, inflammation with circulating IL-33 and lung function in 30 patients with severe uncontrolled asthma. MATERIALS AND METHODS The study included 30 patients with severe uncontrolled asthma. In each of them were measured serum levels of IL-33 and Vitamin D by the ELISA method. The pulmonary function is measured by basic spirometry parameters, FEV1. The results were statistically elaborated according to the Pearson's Correlation Tests. RESULTS The results showed statistically insignificant correlation between Vitamin D and IL-33, and Vitamin D with FEV1 (Vit.D/IL-33; r = 0.11323, p = 0.551); (Vit.D/FEV1; r = -0.1005; p = 0.597) Correlation between IL-33 and FEV1 is negative but statistically significant (IL-33/FEV1; r = -0.5248; p = 0.003). CONCLUSION Because there are little studies about the link between vitamin D and asthma, further research to clarify the mechanism how vitamin D control the activity of CD4+ T cells and the related Th2-type cytokines in the parthenogenesis of asthma.
Collapse
Affiliation(s)
- Elena Janeva-Jovanovska
- PHI University Clinic of Pulmonology and Allergy, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Dejan Dokic
- PHI University Clinic of Pulmonology and Allergy, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Biserka Jovkovska-Kaeva
- PHI University Clinic of Pulmonology and Allergy, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Gorica Breskovska
- PHI University Clinic of Pulmonology and Allergy, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Zlatica Goseva
- PHI University Clinic of Pulmonology and Allergy, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Jordan Minov
- Institute for Occupational Health of RM - Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Dejan Trajkov
- Institute of Immunobiology and Human Genetics, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Magdalena Dimitrova-Genadieva
- PHI University Clinic of Gastroenterohepatology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Beti Zafirovska-Ivanovska
- Institute of Epidemiology and Biostatistics, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
99957
|
Chen M, Jiang L, Li Y, Bai G, Zhao J, Zhang M, Zhang J. Hydrogen protects against liver injury during CO 2 pneumoperitoneum in rats. Oncotarget 2017; 9:2631-2645. [PMID: 29416797 PMCID: PMC5788665 DOI: 10.18632/oncotarget.23498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the current study was to identify the protective effect of hydrogen gas against liver injury during CO2 pneumoperitoneum. Rats were randomly divided into three groups: control group (C group), pneumoperitoneum group (P15 group) and hydrogen group (H2 group). Rats in the C group were subjected to anesthesia for 90 min. Rats in the P15 group received an abdominal insufflation of CO2 for 90 min at an intra-abdominal pressure of 15 mmHg. Rats in the H2 group received a hypodermic injection of hydrogen gas (0.2 mL/kg) and after 10 min they received an abdominal insufflation of CO2 for 90 min at an intra-abdominal pressure of 15 mmHg. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured to evaluate liver function. Malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione (GSH) content were measured to evaluate oxidative stress. Nuclear factor E2-related factor 2 (Nrf2) and Nrf2 downstream target genes, apoptosis-related genes and inflammatory cytokine mRNA and protein expression were detected. Liver injury was detected under the microscope. Our results revealed that liver function, antioxidants content, inflammation and liver injury were improved after hydrogen preconditioning in H2 group compared with P15 group. Overall, our results revealed that subcutaneous hydrogen injection could exert a protective effect against liver injury during CO2 pneumoperitoneum through reducing oxidative stress, cell apoptosis and inflammatory cytokines release.
Collapse
Affiliation(s)
- Mingzi Chen
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Lihong Jiang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yue Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ge Bai
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jinghua Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ming Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiantao Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
99958
|
Leptin and ghrelin: Sewing metabolism onto neurodegeneration. Neuropharmacology 2017; 136:307-316. [PMID: 29248481 DOI: 10.1016/j.neuropharm.2017.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/23/2022]
Abstract
Life expectancy has considerably increased over the last decades. The negative consequence of this augmented longevity has been a dramatic increase of age-related chronic neurodegenerative diseases, such as Alzheimer's, Parkinson's and multiple sclerosis. Epidemiology is telling us there exists a strong correlation between the neuronal loss characterizing these disorders and metabolic dysfunction. This review aims at presenting the evidence supporting the existence of a molecular system linking metabolism with neurodegeneration, with a specific focus on the role of two hormones with a key role in the regulatory cross talk between metabolic imbalance and the damage of nervous system: leptin and ghrelin. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
|
99959
|
Min Y, Lee S, Kim MJ, Chun E, Lee KY. Ubiquitin-Specific Protease 14 Negatively Regulates Toll-Like Receptor 4-Mediated Signaling and Autophagy Induction by Inhibiting Ubiquitination of TAK1-Binding Protein 2 and Beclin 1. Front Immunol 2017; 8:1827. [PMID: 29326710 PMCID: PMC5736539 DOI: 10.3389/fimmu.2017.01827] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/04/2017] [Indexed: 11/24/2022] Open
Abstract
Ubiquitin-specific protease 14 (USP14), one of three proteasome-associated deubiquitinating enzymes, has multifunctional roles in cellular context. Here, we report a novel molecular mechanism and function of USP14 in regulating autophagy induction and nuclear factor-kappa B (NF-κB) activation induced by toll-like receptor (TLR) 4 (TLR4). USP14 interacted with tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) and interrupted the association of Beclin 1 with TRAF6, leading to inhibition of TRAF6-mediated ubiquitination of Beclin 1. Reduced expression of USP14 in USP14-knockdown (USP14KD) THP-1 cells enhanced autophagy induction upon TLR4 stimulation as shown by the increased conversion of cytosolic LC3-I to membrane-bound LC3-II. Moreover, USP14KD human breast carcinoma MDA-MB-231 cells and USP14KD human hepatic adenocarcinoma SK-HEP-1 cells showed increased cell migration and invasion, indicating that USP14 is negatively implicated in the cancer progression by the inhibition of autophagy induction. Furthermore, we found that USP14 interacted with TAK1-binding protein (TAB) 2 protein and induced deubiquitination of TAB 2, a key factor in the activation of NF-κB. Functionally, overexpression of USP14 suppressed TLR4-induced activation of NF-κB. In contrast, USP14KD THP-1 cells showed enhanced activation of NF-κB, NF-κB-dependent gene expression, and production of pro-inflammatory cytokines such as IL-6, IL-1β, and tumor necrosis factor-α. Taken together, our data demonstrate that USP14 can negatively regulate autophagy induction by inhibiting Beclin 1 ubiquitination, interrupting association between TRAF6 and Beclin 1, and affecting TLR4-induced activation of NF-κB through deubiquitination of TAB 2 protein.
Collapse
Affiliation(s)
- Yoon Min
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Sena Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Mi-Jeong Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Eunyoung Chun
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, United States.,The Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Ki-Young Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Samsung Medical Center, Seoul, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
99960
|
Dai L, Sahin O, Tang Y, Zhang Q. A Mutator Phenotype Promoting the Emergence of Spontaneous Oxidative Stress-Resistant Mutants in Campylobacter jejuni. Appl Environ Microbiol 2017; 83:e01685-17. [PMID: 29030436 PMCID: PMC5717198 DOI: 10.1128/aem.01685-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/03/2017] [Indexed: 12/27/2022] Open
Abstract
Campylobacter jejuni is a leading cause of foodborne illnesses worldwide. As a microaerophilic organism, C. jejuni must be able to defend against oxidative stress encountered both in the host and in the environment. How Campylobacter utilizes a mutation-based mechanism for adaptation to oxidative stress is still unknown. Here we present a previously undescribed phenotypic and genetic mechanism that promotes the emergence of oxidative stress-resistant mutants. Specifically, we showed that a naturally occurring mutator phenotype, resulting from a loss of function mutation in the DNA repair enzyme MutY, increased oxidative stress resistance (OXR) in C. jejuni We further demonstrated that MutY malfunction did not directly contribute to the OXR phenotype but increased the spontaneous mutation rate in the peroxide regulator gene perR, which functions as a repressor for multiple genes involved in oxidative stress resistance. Mutations in PerR resulted in loss of its DNA binding function and derepression of PerR-controlled oxidative stress defense genes, thereby conferring an OXR phenotype and facilitating Campylobacter survival under oxidative stress. These findings reveal a new mechanism that promotes the emergence of spontaneous OXR mutants in bacterial organisms.IMPORTANCE Although a mutator phenotype has been shown to promote antibiotic resistance in many bacterial species, little is known about its contribution to the emergence of OXR mutants. This work describes the link between a mutator phenotype and the enhanced emergence of OXR mutants as well as its underlying mechanism involving DNA repair and mutations in PerR. Since DNA repair systems and PerR are well conserved in many bacterial species, especially in Gram positives, the same mechanism may operate in multiple bacterial species. Additionally, we developed a novel method that allows for rapid quantification of spontaneous OXR mutants in a bacterial population. This method represents a technical innovation and may also be applied to other bacterial species. These findings significantly advance our understanding of bacterial mechanisms for survival under oxidative stress.
Collapse
Affiliation(s)
- Lei Dai
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Orhan Sahin
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Yizhi Tang
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Qijing Zhang
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
99961
|
Abstract
The characterization of protein interactions has become essential in many fields of life science, especially drug discovery. Microscale thermophoresis (MST) is a powerful new method for the quantitative analysis of protein-protein interactions (PPIs) with low sample consumption. In addition, one of the major advantages of this technique is that no tedious purification step is necessary to access the protein of interest. Here, we describe a protocol using MST to determine the binding affinity of the PD-1/PD-L1 couple, which is involved in tumour escape processes, without purification of the target protein from cell lysates. The method requires the overexpression of fluorescent proteins in CHO-K1 cells and describes the optimal conditions for determining the dissociation constant. The protocol has a variety of potential applications in studying the interactions of these proteins with small molecules and demonstrates that MST is a valuable method for studying the PD-1/PD-L1 pathway.
Collapse
|
99962
|
Mariappan L, Jiang XY, Jackson J, Drew Y. Emerging treatment options for ovarian cancer: focus on rucaparib. Int J Womens Health 2017; 9:913-924. [PMID: 29290694 PMCID: PMC5735986 DOI: 10.2147/ijwh.s151194] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are an exciting class of anticancer drugs, which have revolutionized the management of BRCA mutant/homologous recombination-deficient recurrent high-grade serous ovarian cancer (HGSOC). With three PARPi now approved by the US Food and Drug Administration, olaparib (Lynparza™), niraparib (Zejula™), and rucaparib (Rubraca™) in 2014 (and 2017 for the tablet formulation), 2016, and 2017, respectively, these drugs have now entered routine clinical practice. The marked single-agent efficacy of PARPi either as maintenance following response to platinum-based chemotherapy or as up-front treatment in these indications is based on the well-known concept of synthetic lethality. PARPi themselves work by blocking the repair of single-strand DNA breaks by the base excision/single-strand break repair pathway and can also be directly cytotoxic by the mechanism of PARP trapping. The greatest benefit in terms of progression-free survival, in all three PARPi maintenance registration studies, was seen in women with platinum-sensitive BRCA mutation-associated HGSOC. However, it is clear that non-BRCA HGSOC can benefit from PARPi and the ongoing challenge of biomarker driven studies is how best to define these patients. PARPi are well tolerated, but more information is needed to assess the longer-term/later onset toxicities as these agents are investigated in the first-line setting. The future direction and challenges for PARPi will be to continue to expand beyond BRCA and ovarian cancer by identifying molecular or functional signatures of response; to see if the durable responses in ovarian cancer can be improved and efficacy can be achieved in other cancer sub-types by combining with novel targeted agents. This review summarizes the development of PARPi as a class in ovarian cancer with particular focus on the PARPi rucaparib.
Collapse
Affiliation(s)
| | | | - Josie Jackson
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| | - Yvette Drew
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
99963
|
Sumner JA, Chen Q, Roberts AL, Winning A, Rimm EB, Gilsanz P, Glymour MM, Tworoger SS, Koenen KC, Kubzansky LD. Cross-Sectional and Longitudinal Associations of Chronic Posttraumatic Stress Disorder With Inflammatory and Endothelial Function Markers in Women. Biol Psychiatry 2017; 82:875-884. [PMID: 28778657 PMCID: PMC5683901 DOI: 10.1016/j.biopsych.2017.06.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/24/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD) may contribute to heightened cardiovascular disease risk by promoting a proinflammatory state and impaired endothelial function. Previous research has demonstrated associations of PTSD with inflammatory and endothelial function biomarkers, but most work has been cross-sectional and does not separate the effects of trauma exposure from those of PTSD. METHODS We investigated associations of trauma exposure and chronic PTSD with biomarkers of inflammation (C-reactive protein and tumor necrosis factor alpha receptor II) and endothelial function (intercellular adhesion molecule-1 and vascular cell adhesion molecule-1) in 524 middle-aged women in the Nurses' Health Study II. Using linear mixed models, we examined associations of trauma/PTSD status with biomarkers measured twice, 10 to 16 years apart, in cardiovascular disease-free women, considering either average levels over time (cross-sectional) or change in levels over time (longitudinal). Biomarker levels were log-transformed. Trauma/PTSD status (based on structured diagnostic interviews) was defined as no trauma at either blood draw (n = 175), trauma at blood draw 1 but no PTSD at either draw (n = 175), and PTSD that persisted beyond blood draw 1 (chronic PTSD; n = 174). The reference group was women without trauma. RESULTS In models adjusted for known potential confounders, women with chronic PTSD had higher average C-reactive protein (B = 0.27, p < .05), tumor necrosis factor alpha receptor II (B = 0.07, p < .01), and intercellular adhesion molecule-1 (B = 0.04, p < .05) levels. Women with trauma but without PTSD had higher average tumor necrosis factor alpha receptor II levels (B = 0.05, p < .05). In addition, women with chronic PTSD had a greater increase in vascular cell adhesion molecule-1 over time (B = 0.003, p < .05). CONCLUSIONS Increased inflammation and impaired endothelial function may be pathways by which chronic PTSD increases cardiovascular disease risk.
Collapse
Affiliation(s)
- Jennifer A. Sumner
- Center for Behavioral Cardiovascular Health, Columbia University Medical Center, New York, NY,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Correspondence to: Jennifer Sumner, Center for Behavioral Cardiovascular Health, Columbia University Medical Center, 622 W. 168th St, PH 9-315, New York, NY 10032. Tel: 212-342-3133; Fax: 212-342-3431;
| | - Qixuan Chen
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY
| | - Andrea L. Roberts
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Ashley Winning
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Eric B. Rimm
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Paola Gilsanz
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA
| | - M. Maria Glymour
- Department of Epidemiology and Biostatistics, University of California San Francisco School of Medicine, San Francisco, CA
| | - Shelley S. Tworoger
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Karestan C. Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Psychiatric and Neurodevelopmental Genetics Unit and Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Laura D. Kubzansky
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
99964
|
Feng Y, Liao S, Wei C, Jia D, Wood K, Liu Q, Wang X, Shi FD, Jin WN. Infiltration and persistence of lymphocytes during late-stage cerebral ischemia in middle cerebral artery occlusion and photothrombotic stroke models. J Neuroinflammation 2017; 14:248. [PMID: 29246244 PMCID: PMC5732427 DOI: 10.1186/s12974-017-1017-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/29/2017] [Indexed: 01/05/2023] Open
Abstract
Background Evidence suggests that brain infiltration of lymphocytes contributes to acute neural injury after cerebral ischemia. However, the spatio-temporal dynamics of brain-infiltrating lymphocytes during the late stage after cerebral ischemia remains unclear. Methods C57BL/6 (B6) mice were subjected to sham, photothrombosis, or 60-min transient middle cerebral artery occlusion (MCAO) procedures. Infarct volume, neurodeficits, production of reactive oxygen species (ROS) and inflammatory factors, brain-infiltrating lymphocytes, and their activation as well as pro-inflammatory cytokine IFN-γ production were assessed. Brain-infiltrating lymphocytes were also measured in tissue sections from post-mortem patients after ischemic stroke by immunostaining. Results In mice subjected to transient MCAO or photothrombotic stroke, we found that lymphocyte infiltration persists in the ischemic brain until at least day 14 after surgery, during which brain infarct volume significantly diminished. These brain-infiltrating lymphocytes express activation marker CD69 and produce proinflammatory cytokines such as IFN-γ, accompanied with a sustained increase of reactive oxygen species (ROS) and inflammatory cytokines release in the brain. In addition, brain-infiltrating lymphocytes were observed in post-mortem brain sections from patients during the late stage of ischemic stroke. Conclusion Our results demonstrate that brain-infiltration of lymphocytes persists after the acute stage of cerebral ischemia, facilitating future advanced studies to reveal the precise role of lymphocytes during late stage of stroke. Electronic supplementary material The online version of this article (10.1186/s12974-017-1017-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Feng
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Center for Neuroinflammation, Beijing TianTan Hospital, Beijing, 100070, China
| | - Shiwei Liao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Changjuan Wei
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dongmei Jia
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Kristofer Wood
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, 02129, MA, USA
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Center for Neuroinflammation, Beijing TianTan Hospital, Beijing, 100070, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA
| | - Wei-Na Jin
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China. .,Center for Neuroinflammation, Beijing TianTan Hospital, Beijing, 100070, China. .,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA.
| |
Collapse
|
99965
|
Khanna P, Chua PJ, Wong BSE, Yin C, Thike AA, Wan WK, Tan PH, Baeg GH. GRAM domain-containing protein 1B (GRAMD1B), a novel component of the JAK/STAT signaling pathway, functions in gastric carcinogenesis. Oncotarget 2017; 8:115370-115383. [PMID: 29383166 PMCID: PMC5777778 DOI: 10.18632/oncotarget.23265] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/03/2017] [Indexed: 12/31/2022] Open
Abstract
Dysregulated JAK/STAT signaling has been implicated in the molecular pathogenesis of gastric cancer. However, downstream effectors of STAT signaling that facilitate gastric carcinogenesis remain to be explored. We previously identified the Drosophila ortholog of human GRAMD1B in our genome-wide RNAi screen to identify novel components of the JAK/STAT signaling pathway in Drosophila. Here, we examined the involvement of GRAMD1B in JAK/STAT-associated gastric carcinogenesis. We found that GRAMD1B expression is positively regulated by JAK/STAT signaling and GRAMD1B inhibition decreases STAT3 levels, suggesting the existence of a positive feedback loop. Consistently, GRAMD1B and JAK/STAT signaling acted synergistically to promote gastric cancer cell survival by upregulating the expression of the anti-apoptotic molecule Bcl-xL. Interestingly, our immunohistochemical analysis for GRAMD1B revealed a gradual loss of cytoplasmic staining but an increase in the nuclear accumulation of GRAMD1B, as gastric tissue becomes malignant. GRAMD1B expression levels were also found to be significantly associated with clinicopathological features of the gastric cancer patients, particularly the tumor grades and lymph node status. Moreover, GRAMD1B and pSTAT3 (Tyr705) showed a positive correlation in gastric tissues, thereby confirming the existence of a close link between these two signaling molecules in vivo. This new knowledge about JAK/STAT-GRAMD1B regulation deepens our understanding of JAK/STAT signaling in gastric carcinogenesis and provides a foundation for the development of novel biomarkers in gastric cancer.
Collapse
Affiliation(s)
- Puja Khanna
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
| | - Pei Jou Chua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
| | - Belinda Shu Ee Wong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
| | - Changhong Yin
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Wei Keat Wan
- Division of Pathology, Singapore General Hospital, Singapore 169856, Singapore.,Academic Clinical Program for Pathology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore 169856, Singapore
| | - Gyeong Hun Baeg
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
| |
Collapse
|
99966
|
Esteso G, Guerra S, Valés-Gómez M, Reyburn HT. Innate immune recognition of double-stranded RNA triggers increased expression of NKG2D ligands after virus infection. J Biol Chem 2017; 292:20472-20480. [PMID: 28986447 PMCID: PMC5733586 DOI: 10.1074/jbc.m117.818393] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 09/28/2017] [Indexed: 12/16/2022] Open
Abstract
Self/non-self-discrimination by the innate immune system relies on germline-encoded, non-rearranging receptors expressed by innate immune cells recognizing conserved pathogen-associated molecular patterns. The natural killer group 2D (NKG2D) receptor is a potent immune-activating receptor that binds human genome-encoded ligands, whose expression is negligible in normal tissues, but increased in stress and disease conditions for reasons that are incompletely understood. Here it is not clear how the immune system reconciles receptor binding of self-proteins with self/non-self-discrimination to avoid autoreactivity. We now report that increased expression of NKG2D ligands after virus infection depends on interferon response factors activated by the detection of viral double-stranded RNA by pattern-recognition receptors (RIG-I/MDA-5) and that NKG2D ligand up-regulation can be blocked by the expression of viral dsRNA-binding proteins. Thus, innate immunity-mediated recognition of viral nucleic acids triggers the infected cell to release interferon for NK cell recruitment and to express NKG2D ligands to become more visible to the immune system. Finally, the observation that NKG2D-ligand induction is a consequence of signaling by pattern-recognition receptors that have been selected over evolutionary time to be highly pathogen-specific explains how the risks of autoreactivity in this system are minimized.
Collapse
MESH Headings
- Amino Acid Substitution
- Animals
- Cell Line
- Cells, Cultured
- Cricetinae
- DEAD Box Protein 58/chemistry
- DEAD Box Protein 58/genetics
- DEAD Box Protein 58/metabolism
- Gene Expression Regulation
- Gene Expression Regulation, Viral
- Genes, Reporter
- Humans
- Immunity, Innate
- Interferon-Induced Helicase, IFIH1/chemistry
- Interferon-Induced Helicase, IFIH1/genetics
- Interferon-Induced Helicase, IFIH1/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/virology
- Lentivirus/immunology
- Lentivirus/physiology
- Ligands
- Mutation
- NK Cell Lectin-Like Receptor Subfamily K/agonists
- NK Cell Lectin-Like Receptor Subfamily K/genetics
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- RNA/metabolism
- RNA, Viral/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptors, Immunologic
- Recombinant Proteins/metabolism
- Viral Proteins/genetics
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Gloria Esteso
- From the Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas (CNB-CSIC), 28049 Madrid and
| | - Susana Guerra
- the Department of Preventive Medicine and Public Health, Universidad Autónoma, 28029 Madrid, Spain
| | - Mar Valés-Gómez
- From the Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas (CNB-CSIC), 28049 Madrid and
| | - Hugh T Reyburn
- From the Department of Immunology and Oncology, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas (CNB-CSIC), 28049 Madrid and
| |
Collapse
|
99967
|
Huang L, Betjes MGH, Klepper M, Langerak AW, Baan CC, Litjens NHR. End-Stage Renal Disease Causes Skewing in the TCR Vβ-Repertoire Primarily within CD8 + T Cell Subsets. Front Immunol 2017; 8:1826. [PMID: 29326709 PMCID: PMC5736542 DOI: 10.3389/fimmu.2017.01826] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/04/2017] [Indexed: 12/12/2022] Open
Abstract
A broad T cell receptor (TCR-) repertoire is required for an effective immune response. TCR-repertoire diversity declines with age. End-stage renal disease (ESRD) patients have a prematurely aged T cell system which is associated with defective T cell-mediated immunity. Recently, we showed that ESRD may significantly skew the TCR Vβ-repertoire. Here, we assessed the impact of ESRD on the TCR Vβ-repertoire within different T cell subsets using a multiparameter flow-cytometry-based assay, controlling for effects of aging and CMV latency. Percentages of 24 different TCR Vβ-families were tested in circulating naive and memory T cell subsets of 10 ESRD patients and 10 age- and CMV-serostatus-matched healthy individuals (HI). The Gini-index, a parameter used in economics to describe the distribution of income, was calculated to determine the extent of skewing at the subset level taking into account frequencies of all 24 TCR Vβ-families. In addition, using HI as reference population, the differential impact of ESRD was assessed on clonal expansion at the level of an individual TCR Vβ-family. CD8+, but not CD4+, T cell differentiation was associated with higher Gini-TCR indices. Gini-TCR indices were already significantly higher for different CD8+ memory T cell subsets of younger ESRD patients compared to their age-matched HI. ESRD induced expansions of not one TCR Vβ-family in particular and expansions were predominantly observed within the CD8+ T cell compartment. All ESRD patients had expanded TCR Vβ-families within total CD8+ T cells and the median (IQ range) number of expanded TCR Vβ-families/patient amounted to 2 (1-4). Interestingly, ESRD also induced clonal expansions of TCR Vβ-families within naive CD8+ T cells as 8 out of 10 patients had expanded TCR Vβ-families. The median (IQ range) number of expanded families/patient amounted to 1 (1-1) within naive CD8+ T cells. In conclusion, loss of renal function skews the TCR Vβ-repertoire already in younger patients by inducing expansions of different TCR Vβ-families within the various T cell subsets, primarily affecting the CD8+ T cell compartment. This skewed TCR Vβ-repertoire may be associated with a less broad and diverse T cell-mediated immunity.
Collapse
Affiliation(s)
- Ling Huang
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Nephrology and Transplantation, Rotterdam, Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Nephrology and Transplantation, Rotterdam, Netherlands
| | - Mariska Klepper
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Nephrology and Transplantation, Rotterdam, Netherlands
| | - Anton W Langerak
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Nephrology and Transplantation, Rotterdam, Netherlands
| | - Nicolle H R Litjens
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Nephrology and Transplantation, Rotterdam, Netherlands
| |
Collapse
|
99968
|
Sutton TC, Chakraborty S, Mallajosyula VVA, Lamirande EW, Ganti K, Bock KW, Moore IN, Varadarajan R, Subbarao K. Protective efficacy of influenza group 2 hemagglutinin stem-fragment immunogen vaccines. NPJ Vaccines 2017; 2:35. [PMID: 29263889 PMCID: PMC5732283 DOI: 10.1038/s41541-017-0036-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/30/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022] Open
Abstract
The stem of the influenza A virus hemagglutinin (HA) is highly conserved and represents an attractive target for a universal influenza vaccine. The 18 HA subtypes of influenza A are phylogenetically divided into two groups, and while protection with group 1 HA stem vaccines has been demonstrated in animal models, studies on group 2 stem vaccines are limited. Thus, we engineered group 2 HA stem-immunogen (SI) vaccines targeting the epitope for the broadly neutralizing monoclonal antibody CR9114 and evaluated vaccine efficacy in mice and ferrets. Immunization induced antibodies that bound to recombinant HA protein and viral particles, and competed with CR9114 for binding to the HA stem. Mice vaccinated with H3 and H7-SI were protected from lethal homologous challenge with X-79 (H3N2) or A/Anhui/1/2013 (H7N9), and displayed moderate heterologous protection. In ferrets, H7-SI vaccination did not significantly reduce weight loss or nasal wash titers after robust 107 TCID50 H7N9 virus challenge. Epitope mapping revealed ferrets developed lower titers of antibodies that bound a narrow range of HA stem epitopes compared to mice, and this likely explains the lower efficacy in ferrets. Collectively, these findings indicate that while group 2 SI vaccines show promise, their immunogenicity and efficacy are reduced in larger outbred species, and will have to be enhanced for successful translation to a universal vaccine.
Collapse
Affiliation(s)
- Troy C Sutton
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD USA
| | - Saborni Chakraborty
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka India
| | | | | | - Ketaki Ganti
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD USA
| | - Kevin W Bock
- Comparative Medicine Branch, Infectious Disease Pathogenesis Section, NIAID, NIH, Bethesda, MD USA
| | - Ian N Moore
- Comparative Medicine Branch, Infectious Disease Pathogenesis Section, NIAID, NIH, Bethesda, MD USA
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, Karnataka India
| | - Kanta Subbarao
- Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD USA.,Present Address: WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute, 792 Elizabeth Street, Melbourne, VIC Australia
| |
Collapse
|
99969
|
Grössinger EM, Kang M, Bouchareychas L, Sarin R, Haudenschild DR, Borodinsky LN, Adamopoulos IE. Ca 2+-Dependent Regulation of NFATc1 via KCa3.1 in Inflammatory Osteoclastogenesis. THE JOURNAL OF IMMUNOLOGY 2017; 200:749-757. [PMID: 29246953 DOI: 10.4049/jimmunol.1701170] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/02/2017] [Indexed: 12/21/2022]
Abstract
In inflammatory arthritis, the dysregulation of osteoclast activity by proinflammatory cytokines, including TNF, interferes with bone remodeling during inflammation through Ca2+-dependent mechanisms causing pathological bone loss. Ca2+-dependent CREB/c-fos activation via Ca2+-calmodulin kinase IV (CaMKIV) induces transcriptional regulation of osteoclast-specific genes via NFATc1, which facilitate bone resorption. In leukocytes, Ca2+ regulation of NFAT-dependent gene expression oftentimes involves the activity of the Ca2+-activated K+ channel KCa3.1. In this study, we evaluate KCa3.1 as a modulator of Ca2+-induced NFAT-dependent osteoclast differentiation in inflammatory bone loss. Microarray analysis of receptor activator of NF-κB ligand (RANKL)-activated murine bone marrow macrophage (BMM) cultures revealed unique upregulation of KCa3.1 during osteoclastogenesis. The expression of KCa3.1 in vivo was confirmed by immunofluorescence staining on multinucleated cells at the bone surface of inflamed mouse joints. Experiments on in vitro BMM cultures revealed that KCa3.1-/- and TRAM-34 treatment significantly reduced the expression of osteoclast-specific genes (p < 0.05) alongside decreased osteoclast formation (p < 0.0001) in inflammatory (RANKL+TNF) and noninflammatory (RANKL) conditions. In particular, live cell Ca2+ imaging and Western blot analysis showed that TRAM-34 pretreatment decreased transient RANKL-induced Ca2+ amplitudes in BMMs by ∼50% (p < 0.0001) and prevented phosphorylation of CaMKIV. KCa3.1-/- reduced RANKL+/-TNF-stimulated phosphorylation of CREB and expression of c-fos in BMMs (p < 0.01), culminating in decreased NFATc1 protein expression and transcriptional activity (p < 0.01). These data indicate that KCa3.1 regulates Ca2+-dependent NFATc1 expression via CaMKIV/CREB during inflammatory osteoclastogenesis in the presence of TNF, corroborating its role as a target candidate for the treatment of bone erosion in inflammatory arthritis.
Collapse
Affiliation(s)
- Eva M Grössinger
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | - Mincheol Kang
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | - Laura Bouchareychas
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | - Ritu Sarin
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616
| | | | - Laura N Borodinsky
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616; and.,Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children - Northern California, Sacramento, CA 95817
| | - Iannis E Adamopoulos
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California Davis, Davis, CA 95616; .,Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children - Northern California, Sacramento, CA 95817
| |
Collapse
|
99970
|
Liu W, Zhu M, Yu Z, Yin D, Lu F, Pu Y, Zhao C, He C, Cao L. Therapeutic effects of diosgenin in experimental autoimmune encephalomyelitis. J Neuroimmunol 2017; 313:152-160. [PMID: 29132838 DOI: 10.1016/j.jneuroim.2017.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 01/10/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system. Currently, there is no drug available to cure this kind of disease. Diosgenin is a plant-derived steroid saponin. A previous study in our lab revealed that diosgenin can promote oligodendrocyte progenitor cell differentiation and accelerate remyelination. In the present study, we found that diosgenin dose-dependently alleviated the progression of experimental autoimmune encephalomyelitis with reduced central nervous system inflammation and demyelination. We also found that diosgenin treatment can significantly inhibit the activation of microglia and macrophages, suppress CD4+ T cell proliferation and hinder Th1/Th17 cell differentiation. Therefore, we suggested that diosgenin may be a potential therapeutic drug for inflammatory demyelinating diseases, such as MS.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/therapeutic use
- Antigens, CD/metabolism
- Diosgenin/therapeutic use
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalitis/drug therapy
- Encephalitis/etiology
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/pathology
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Statistics, Nonparametric
- Treatment Outcome
Collapse
Affiliation(s)
- Weili Liu
- Institute of Health Sciences, Anhui University, Hefei 230601, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Mei Zhu
- Institute of Health Sciences, Anhui University, Hefei 230601, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Zhongwang Yu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Dou Yin
- Institute of Health Sciences, Anhui University, Hefei 230601, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Fengfeng Lu
- Institute of Health Sciences, Anhui University, Hefei 230601, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Yingyan Pu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, England, UK
| | - Cheng He
- Institute of Health Sciences, Anhui University, Hefei 230601, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China.
| | - Li Cao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
99971
|
Xiao Y, Sun L, Fu Y, Huang Y, Zhou R, Hu X, Zhou P, Quan J, Li N, Fan XG. High mobility group box 1 promotes sorafenib resistance in HepG2 cells and in vivo. BMC Cancer 2017; 17:857. [PMID: 29246127 PMCID: PMC5731191 DOI: 10.1186/s12885-017-3868-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/29/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Primary liver cancer is a lethal malignancy with a high mortality worldwide. Currently, sorafenib is the most effective molecular-targeted drug against hepatocellular carcinoma (HCC). However, the sorafenib resistance rate is high. The molecular mechanism of this resistance has not been fully elucidated. High mobility group box 1 (HMGB1) is a multifaceted protein that plays a key role in the proliferation, apoptosis, metastasis and angiogenesis of HCC cells. In addition, HMGB1 has been suggested to contribute to chemotherapy resistance in tumours, including lung cancer, osteosarcoma, neuroblastoma, leukaemia, and colorectal cancer. This study investigated the association between HMGB1 and sorafenib resistance in HCC. METHODS HepG2 cells with HMGB1 knockdown or overexpression were generated. The efficacy of sorafenib in these cells was tested using flow cytometry and a cell counting assay. The subcellular localization of HMGB1 in HepG2 cells following sorafenib treatment was measured by western blotting and confocal microscopy. A murine subcutaneous HCC model was generated to examine the association between HMGB1 and the sensitivity of sorafenib treatment. RESULTS The HMGB1 knockdown cells exhibited a significantly higher apoptotic level and lower cell viability than the normal HMGB1 expressing cells following the sorafenib treatment. In addition, the cell viability observed in the HMGB1 overexpressing cells was higher than that observed in the control cells following the sorafenib intervention. Sorafenib had a better tumour inhibition effect in the HMGB1 knockdown group in vivo. The amount of mitochondrial HMGB1 decreased, while the amount of cytosolic HMGB1 increased following the exposure to sorafenib. Altogether, HMGB1 translocated from the mitochondria to the cytoplasm outside the mitochondria following the exposure of HepG2 cells to sorafenib. CONCLUSIONS A novel potential role of HMGB1 in the regulation of sorafenib therapy resistance in HCC was observed. The knockdown of HMGB1 restores sensitivity to sorafenib and enhances HepG2 cell death, while HMGB1 overexpression blunts these effects. The translocation of HMGB1 from the mitochondria to the cytosol following sorafenib treatment provides new insight into sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Yinzong Xiao
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Lunquan Sun
- Center for Molecular Medicine, Xiangya Hospital, Key Laboratory of Molecular Radiation Oncology of Hunan Province, Central South University, Changsha, 410008, China
| | - Yongming Fu
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yan Huang
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Rongrong Zhou
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xingwang Hu
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Pengcheng Zhou
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jun Quan
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ning Li
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Xue-Gong Fan
- Hunan Key Laboratory of Viral Hepatitis, Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
99972
|
Nagendraprabhu P, Khatiwada S, Chaulagain S, Delhon G, Rock DL. A parapoxviral virion protein targets the retinoblastoma protein to inhibit NF-κB signaling. PLoS Pathog 2017; 13:e1006779. [PMID: 29244863 PMCID: PMC5747488 DOI: 10.1371/journal.ppat.1006779] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/29/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022] Open
Abstract
Poxviruses have evolved multiple strategies to subvert signaling by Nuclear Factor κB (NF-κB), a crucial regulator of host innate immune responses. Here, we describe an orf virus (ORFV) virion-associated protein, ORFV119, which inhibits NF-κB signaling very early in infection (≤ 30 min post infection). ORFV119 NF-κB inhibitory activity was found unimpaired upon translation inhibition, suggesting that virion ORFV119 alone is responsible for early interference in signaling. A C-terminal LxCxE motif in ORFV119 enabled the protein to interact with the retinoblastoma protein (pRb) a multifunctional protein best known for its tumor suppressor activity. Notably, experiments using a recombinant virus containing an ORFV119 mutation which abrogates its interaction with pRb together with experiments performed in cells lacking or with reduced pRb levels indicate that ORFV119 mediated inhibition of NF-κB signaling is largely pRb dependent. ORFV119 was shown to inhibit IKK complex activation early in infection. Consistent with IKK inhibition, ORFV119 also interacted with TNF receptor associated factor 2 (TRAF2), an adaptor protein recruited to signaling complexes upstream of IKK in infected cells. ORFV119-TRAF2 interaction was enhanced in the presence of pRb, suggesting that ORFV119-pRb complex is required for efficient interaction with TRAF2. Additionally, transient expression of ORFV119 in uninfected cells was sufficient to inhibit TNFα-induced IKK activation and NF-κB signaling, indicating that no other viral proteins are required for the effect. Infection of sheep with ORFV lacking the ORFV119 gene led to attenuated disease phenotype, indicating that ORFV119 contributes to virulence in the natural host. ORFV119 represents the first poxviral protein to interfere with NF-κB signaling through interaction with pRb.
Collapse
Affiliation(s)
- Ponnuraj Nagendraprabhu
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
| | - Sushil Khatiwada
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
| | - Sabal Chaulagain
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
| | - Gustavo Delhon
- School of Veterinary and Biomedical Sciences, Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- * E-mail: (GD); (DLR)
| | - Daniel L. Rock
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
- * E-mail: (GD); (DLR)
| |
Collapse
|
99973
|
Chen Y, Yu S, Wu Q, Tang Y, Jiang C, Zhuang Z, Zhao N, Huang B, Xie L. [Effect of bone marrow mesenchymal stem cells conditioned medium on microglia and its secretion of arginase 1 in rats]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:1500-1505. [PMID: 29806395 PMCID: PMC8498264 DOI: 10.7507/1002-1892.201710038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/30/2017] [Indexed: 11/03/2022]
Abstract
Objective To observe the effect of bone marrow mesenchymal stem cells (BMSCs) conditioned medium on microglia (MGs) and its secretion of arginase 1 (Arg1). Methods The BMSCs separated through differential adhesion method from the femur and tibia marrow of 4-week-old Sprague Dawley (SD) rats were cultured and identified by Vimentin immunofluorescence staining; whereas MGs separated through trypsin digestion method from the brain of 3-day-old SD rats were cultured and identified by Iba1 immunofluorescence staining. The primary MGs were cultured with DMEM/F12 medium containing BMSCs conditioned medium (experimental group) and with single DMEM/F12 medium (control group), respectively. After 48 hours of culture, the morphology of MGs was observed by inverted phase contrast microscope, the activated state of MGs was detected by using Iba1 immunofluorescence staining, and Arg1 expression of MGs was assessed by Iba1-Arg1 double-labelling immunofluorescence staining and Western blot method. Results Inverted phase contrast microscope observation showed that BMSCs entered logarithmic growth phase at 14 days after culture, and more than 98% cells were positive to Vimentin immunofluorescence staining; whereas MGs entered logarithmic growth phase at 21 days after culture, and around 80% cells were positive to Iba1 immunofluorescence staining. Inverted phase contrast microscope observation displayed that in the experimental group, MGs were activated with increased size of soma, shortened process, and amoeba change. Immunofluorescence staining displayed that the Iba1 positive cells number in the experimental group was significantly higher than that in the control group ( t=0.007, P=0.000); double-labelling immunofluorescence staining revealed that the Iba1-Arg1 positive cells number in the experimental group was significantly higher than that in the control group ( t=0.007, P=0.000); and Western blot results elucidated that the relative expression of Arg1 protein in the experimental group was significantly higher than that in the control group ( t=0.001, P=0.000). Conclusion BMSCs conditioned medium can activate MGs and induce MGs to express Arg1.
Collapse
Affiliation(s)
- Yunhui Chen
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Shuguang Yu
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Qiaofeng Wu
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Yong Tang
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Cen Jiang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Zhiqi Zhuang
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Na Zhao
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Biao Huang
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Lushuang Xie
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China;College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075,
| |
Collapse
|
99974
|
Lopez Aguilar A, Gao Y, Hou X, Lauvau G, Yates JR, Wu P. Profiling of Protein O-GlcNAcylation in Murine CD8 + Effector- and Memory-like T Cells. ACS Chem Biol 2017; 12:3031-3038. [PMID: 29125738 DOI: 10.1021/acschembio.7b00869] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
During an acute infection, antigenic stimulation leads to activation, expansion, and differentiation of naïve CD8+ T cells, first into cytotoxic effector cells and eventually into long-lived memory cells. T cell antigen receptors (TCRs) detect antigens on antigen-presenting cells (APCs) in the form of antigenic peptides bound to major histocompatibility complex I (MHC-I)-encoded molecules and initiate TCR signal transduction network. This process is mediated by phosphorylation of many intracellular signaling proteins. Protein O-GlcNAc modification is another post-translational modification involved in this process, which often has either reciprocal or synergistic roles with phosphorylation. In this study, using a chemoenzymatic glycan labeling technique and proteomics analysis, we compared protein O-GlcNAcylation of murine effector and memory-like CD8+ T cells differentiated in vitro. By quantitative proteomics analysis, we identified 445 proteins that are significantly regulated in either effector- or memory-like T cell subsets. Furthermore, qualitative and quantitative analysis identified highly regulated protein clusters that suggest involvement of this post-translational modification in specific cellular processes. In effector-like T cells, protein O-GlcNAcylation is heavily involved in transcriptional and translational processes that drive fast effector T cells proliferation. During the formation of memory-like T cells, protein O-GlcNAcylation is involved in a more specific, perhaps more targeted regulation of transcription, mRNA processing, and translation. Significantly, O-GlcNAc plays a critical role as part of the "histone code" in both CD8+ T cells subgroups.
Collapse
Affiliation(s)
- Aime Lopez Aguilar
- Department
of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Yu Gao
- Department
of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Xiaomeng Hou
- Department
of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Gregoire Lauvau
- Department
of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - John R. Yates
- Department
of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Peng Wu
- Department
of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
99975
|
Current systemic therapies for metastatic renal cell carcinoma in older adults: A comprehensive review. J Geriatr Oncol 2017; 9:265-274. [PMID: 29249644 DOI: 10.1016/j.jgo.2017.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/24/2017] [Accepted: 11/24/2017] [Indexed: 12/16/2022]
Abstract
Physiological changes that occur during the aging process may impact drug metabolism and availability, consequently affecting treatment efficacy and tolerability. Despite being a disease of older adults, there is little data to guide treatment decisions for older patients with metastatic renal cell carcinoma (mRCC). The recent approval of many new agents for this disease poses a clinical challenge: how to best utilize these drugs in a population (older adults) who has been generally under-represented in clinical studies. Additionally, the presence of comorbid conditions, polypharmacy, frailty, and lack of social support place this group of patients in a very unique situation. In order to avoid under-treatment, international societies' guidelines recommend routine use of geriatric tools to assess patients' suitability for systemic treatments. Here we provide a thorough review of age-related metabolic differences, safety and efficacy data for each drug approved for mRCC, and cover specific considerations for the management of older adults with this disease.
Collapse
|
99976
|
Abstract
PURPOSE OF REVIEW Here, we explore the significant progress made in the treatment of multiple myeloma, focusing on immunotherapy and the promise it has offered to patients suffering from advanced disease. RECENT FINDINGS Multiple myeloma, a B-cell malignancy, is characterized by unregulated plasma cell growth in the bone marrow as well as strong immunosuppression in the tumor microenvironment. mAbs targeting tumor antigens overcome this, increasing T-cell activation, multiple myeloma cell death, and depth of response. Similarly, adoptive T-cell therapy aims to engineer or isolate tumor-specific T cells for a targeted approach. Finally, peptide and dendritic cell/tumor fusion vaccines reeducate the immune system, expanding the immune response and generating long-term memory to prevent relapse of disease. Many of these approaches have been combined with existing therapies to enhance antitumor immunity. SUMMARY Immunotherapeutic approaches have remarkably changed the treatment paradigm for multiple myeloma, and encouraging patient responses have warranted further investigation into mAbs, adoptive T-cell therapy, vaccines, and combination therapy.
Collapse
|
99977
|
Oberson A, Spagnuolo L, Puddinu V, Barchet W, Rittner K, Bourquin C. NAB2 is a novel immune stimulator of MDA-5 that promotes a strong type I interferon response. Oncotarget 2017; 9:5641-5651. [PMID: 29464024 PMCID: PMC5814164 DOI: 10.18632/oncotarget.23725] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/13/2017] [Indexed: 11/25/2022] Open
Abstract
Novel adjuvants are needed to increase the efficacy of vaccine formulations and immune therapies for cancer and chronic infections. In particular, adjuvants that promote a strong type I IFN response are required, since this cytokine is crucial for the development of efficient anti-tumoral and anti-viral immunity. Nucleic acid band 2 (NAB2) is a double-stranded RNA molecule isolated from yeast and identified as an agonist of the pattern-recognition receptors TLR3 and MDA-5. We compared the ability of NAB2 to activate innate immunity with that of poly(I:C), a well-characterized TLR3 and MDA-5 agonist known for the induction of type I IFN. NAB2 promoted stronger IFN-α production and induced a higher activation state of both murine and human innate immune cells compared to poly(I:C). This correlated with a stronger activation of the signalling pathway downstream of MDA-5, and IFN-α induction was dependent on MDA-5. Upon injection, NAB2 induced higher levels of serum IFN-α in mice than poly(I:C). These results suggest that NAB2 has the potential to become an efficient adjuvant for the induction of type-I IFN responses in therapeutic immunization against cancer or infections.
Collapse
Affiliation(s)
- Anne Oberson
- Chair of Pharmacology, Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland
| | - Lorenzo Spagnuolo
- Chair of Pharmacology, Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Viola Puddinu
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Winfried Barchet
- German Center for Infection Research, Cologne-Bonn, Germany.,Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Germany
| | - Karola Rittner
- Transgene S.A., Parc d'Innovation, CS80166, 67405 Illkirch-Graffenstaden Cedex, France
| | - Carole Bourquin
- Chair of Pharmacology, Department of Medicine, Faculty of Science, University of Fribourg, 1700 Fribourg, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, 1211 Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
99978
|
Sic H, Speletas M, Cornacchione V, Seidl M, Beibel M, Linghu B, Yang F, Sevdali E, Germenis AE, Oakeley EJ, Vangrevelinghe E, Sailer AW, Traggiai E, Gram H, Eibel H. An Activating Janus Kinase-3 Mutation Is Associated with Cytotoxic T Lymphocyte Antigen-4-Dependent Immune Dysregulation Syndrome. Front Immunol 2017; 8:1824. [PMID: 29375547 PMCID: PMC5770691 DOI: 10.3389/fimmu.2017.01824] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/04/2017] [Indexed: 11/13/2022] Open
Abstract
Heterozygous mutations in the cytotoxic T lymphocyte antigen-4 (CTLA-4) are associated with lymphadenopathy, autoimmunity, immune dysregulation, and hypogammaglobulinemia in about 70% of the carriers. So far, the incomplete penetrance of CTLA-4 haploinsufficiency has been attributed to unknown genetic modifiers, epigenetic changes, or environmental effects. We sought to identify potential genetic modifiers in a family with differential clinical penetrance of CTLA-4 haploinsufficiency. Here, we report on a rare heterozygous gain-of-function mutation in Janus kinase-3 (JAK3) (p.R840C), which is associated with the clinical manifestation of CTLA-4 haploinsufficiency in a patient carrying a novel loss-of-function mutation in CTLA-4 (p.Y139C). While the asymptomatic parents carry either the CTLA-4 mutation or the JAK3 variant, their son has inherited both heterozygous mutations and suffers from hypogammaglobulinemia combined with autoimmunity and lymphoid hyperplasia. Although the patient's lymph node and spleen contained many hyperplastic germinal centers with follicular helper T (TFH) cells and immunoglobulin (Ig) G-positive B cells, plasma cell, and memory B cell development was impaired. CXCR5+PD-1+TIGIT+ TFH cells contributed to a large part of circulating T cells, but they produced only very low amounts of interleukin (IL)-4, IL-10, and IL-21 required for the development of memory B cells and plasma cells. We, therefore, suggest that the combination of the loss-of-function mutation in CTLA-4 with the gain-of-function mutation in JAK3 directs the differentiation of CD4 T cells into dysfunctional TFH cells supporting the development of lymphadenopathy, hypogammaglobulinemia, and immunodeficiency. Thus, the combination of rare genetic heterozygous variants that remain clinically unnoticed individually may lead to T cell hyperactivity, impaired memory B cell, and plasma cell development resulting finally in combined immunodeficiency.
Collapse
Affiliation(s)
- Heiko Sic
- Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Matthaios Speletas
- Department of Immunology and Histocompatibility, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | | | - Maximillian Seidl
- Institute for Surgical Pathology, University Medical Center Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - Martin Beibel
- Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Bolan Linghu
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States
| | - Fan Yang
- Novartis Institutes for Biomedical Research, Cambridge, MA, United States
| | - Eirini Sevdali
- Department of Immunology and Histocompatibility, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Anastasios E Germenis
- Department of Immunology and Histocompatibility, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | | | | | | | | | - Hermann Gram
- Novartis Institute for Biomedical Research, Basel, Switzerland
| | - Hermann Eibel
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
99979
|
Liu H, Xue F, Gong J, Wan Q, Fang S. Limited polymorphism of the functional MHC class II B gene in the black-spotted frog ( Pelophylax nigromaculatus) identified by locus-specific genotyping. Ecol Evol 2017; 7:9860-9868. [PMID: 29238521 PMCID: PMC5723586 DOI: 10.1002/ece3.3408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 07/31/2017] [Accepted: 08/19/2017] [Indexed: 12/23/2022] Open
Abstract
Amphibians can be more vulnerable to environmental changes and diseases than other species because of their complex life cycle and physiological requirements. Therefore, understanding the adaptation of amphibians to environmental changes is crucial for their conservation. Major histocompatibility complex (MHC) presents an excellent tool for the investigation of adaptive variations and the assessment of adaptive potential because it can be under strong diversifying selection. Here, we isolated the MHC class II B (MHCIIB) gene from cDNA sequences of the black-spotted frog (Pelophylax nigromaculatus), a widespread amphibian species in China, and designed locus-specific primers to characterize adaptive variability of this amphibian. Ten alleles were identified from 67 individual frogs of three populations and no more than two alleles were present in each individual animal. Furthermore, none of the sequences had indels or/and stop codons, which is in good agreement with locus-specific amplification of a functional gene. However, we found low polymorphism at both nucleotide and amino acid levels, even in the antigen-binding region. Purifying selection acting at this locus was supported by the findings that the dN/dS ratio across all alleles was below 1 and that negatively selected sites were detected by different methods. Allele frequency distributions were significantly different among geographic populations, indicating that physiographic factors may have strong effect on the genetic structure of the black-spotted frog. This study revealed limited polymorphism of three adjacent black-spotted frog populations at the functional MHCIIB locus, which may be attributed to region-specific differences. The locus-specific genotyping technique developed in this study would provide a foundation for future studies on adaptive divergence among different frog populations.
Collapse
Affiliation(s)
- Hong‐Yi Liu
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, and State Conservation Centre for Gene Resources of Endangered WildlifeCollege of Life SciencesZhejiang UniversityHangzhouChina
- Co‐Innovation Center for Sustainable Forestry in Southern ChinaCollege of Biology and the EnvironmentNanjing Forestry UniversityNanjingChina
| | - Fei Xue
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, and State Conservation Centre for Gene Resources of Endangered WildlifeCollege of Life SciencesZhejiang UniversityHangzhouChina
| | - Jie Gong
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, and State Conservation Centre for Gene Resources of Endangered WildlifeCollege of Life SciencesZhejiang UniversityHangzhouChina
| | - Qiu‐Hong Wan
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, and State Conservation Centre for Gene Resources of Endangered WildlifeCollege of Life SciencesZhejiang UniversityHangzhouChina
| | - Sheng‐Guo Fang
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, and State Conservation Centre for Gene Resources of Endangered WildlifeCollege of Life SciencesZhejiang UniversityHangzhouChina
| |
Collapse
|
99980
|
Li Z, Zhang S, Cao L, Li W, Ye YC, Shi ZX, Wang ZR, Sun LX, Wang JW, Jia LT, Wang W. Tanshinone IIA and Astragaloside IV promote the angiogenesis of mesenchymal stem cell-derived endothelial cell-like cells via upregulation of Cx37, Cx40 and Cx43. Exp Ther Med 2017; 15:1847-1854. [PMID: 29434774 PMCID: PMC5776521 DOI: 10.3892/etm.2017.5636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/29/2017] [Indexed: 12/17/2022] Open
Abstract
Tanshinone IIA (Tan IIA) and Astragaloside IV (AGS-IV) were used as therapeutic treatments for coronary heart diseases (CHDs) in ancient China. However, the underlying mechanisms mediating the effects of Tan IIA and AGS-IV in angiogenesis remain unknown. In the present study, mesenchymal stem cells (MSCs) were induced to differentiate into endothelial cell (EC)-like cells in vitro and the effects of Tan IIA and/or AGS-IV on the functions of these cells, including cell proliferation and tube formation, were assessed. Compared with the single-agent groups (Tan IIA or AGS-IV only), combined-agent (Tan IIA and AGS-IV) treatment significantly enhanced the proliferation and tube formation capacity of EC-like cells. In addition, the expression of connexin 37 (Cx37), Cx40 and Cx43 in the combined-agent group was significantly increased compared with the single-agent groups. Furthermore, enhanced gap junctional intercellular communication (GJIC) was identified in the combined-agent group, as evidenced by increased dye transfer in scrape-loading dye transfer assays. In conclusion, Tan IIA and AGS-IV may promote the angiogenesis of EC-like cells by upregulating the expression of Cx37, Cx40 and Cx43 and enhancing GJIC function. The results of the present study may provide experimental evidence for the clinical application of Tan IIA and AGS-IV as a treatment for CHDs.
Collapse
Affiliation(s)
- Zhe Li
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China.,Second Clinical Medical College, Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 710026, P.R. China
| | - Sha Zhang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Liang Cao
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wei Li
- Department of Histology and Embryology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yu-Chen Ye
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zi-Xuan Shi
- Department of Acupuncture, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710003, P.R. China
| | - Zong-Ren Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lian-Xu Sun
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jia-Wei Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wen Wang
- Department of Traditional Chinese Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
99981
|
Tuomela S, Rautio S, Ahlfors H, Öling V, Salo V, Ullah U, Chen Z, Hämälistö S, Tripathi SK, Äijö T, Rasool O, Soueidan H, Wessels L, Stockinger B, Lähdesmäki H, Lahesmaa R. Comparative analysis of human and mouse transcriptomes of Th17 cell priming. Oncotarget 2017; 7:13416-28. [PMID: 26967054 PMCID: PMC4924651 DOI: 10.18632/oncotarget.7963] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/24/2016] [Indexed: 01/17/2023] Open
Abstract
Uncontrolled Th17 cell activity is associated with cancer and autoimmune and inflammatory diseases. To validate the potential relevance of mouse models of targeting the Th17 pathway in human diseases we used RNA sequencing to compare the expression of coding and non-coding transcripts during the priming of Th17 cell differentiation in both human and mouse. In addition to already known targets, several transcripts not previously linked to Th17 cell polarization were found in both species. Moreover, a considerable number of human-specific long non-coding RNAs were identified that responded to cytokines stimulating Th17 cell differentiation. We integrated our transcriptomics data with known disease-associated polymorphisms and show that conserved regulation pinpoints genes that are relevant to Th17 cell-mediated human diseases and that can be modelled in mouse. Substantial differences observed in non-coding transcriptomes between the two species as well as increased overlap between Th17 cell-specific gene expression and disease-associated polymorphisms underline the need of parallel analysis of human and mouse models. Comprehensive analysis of genes regulated during Th17 cell priming and their classification to conserved and non-conserved between human and mouse facilitates translational research, pointing out which candidate targets identified in human are worth studying by using in vivo mouse models.
Collapse
Affiliation(s)
- Soile Tuomela
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sini Rautio
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Helena Ahlfors
- Division of Molecular Immunology, MRC National Institute for Medical Research, London, United Kingdom
| | - Viveka Öling
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Verna Salo
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Ubaid Ullah
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Zhi Chen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Saara Hämälistö
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Subhash K Tripathi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Tarmo Äijö
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Omid Rasool
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Hayssam Soueidan
- Computational Cancer Biology, Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lodewyk Wessels
- Computational Cancer Biology, Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Brigitta Stockinger
- Division of Molecular Immunology, MRC National Institute for Medical Research, London, United Kingdom
| | - Harri Lähdesmäki
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Computer Science, Aalto University, Espoo, Finland
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
99982
|
Tidwell WJ, Fowler JF. T-cell inhibitors for atopic dermatitis. J Am Acad Dermatol 2017; 78:S67-S70. [PMID: 29248519 DOI: 10.1016/j.jaad.2017.12.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/08/2017] [Accepted: 12/09/2017] [Indexed: 11/16/2022]
Abstract
The management of atopic dermatitis is changing with the development of novel biologic agents to target specific molecules in the inflammatory cascade. Following the ability of dupilumab has proved its ability to act on the interleukin 4 receptor in treating atopic dermatitis. Thymic stromal lymphopoietin monoclonal antibody (AMG157/MEDI9929) and OX40 blocking antibody (GBR 830) were developed by targeting the same pathway as dupilumab further upstream. The clinical data on the efficacy for these drugs are not yet known. There is some early evidence that AMG157/MEDI9929 attenuates most measures of allergen-induced asthmatic responses. However, there are no public data on its ability to treat atopic dermatitis. In a phase 2a study, GBR 830 showed at least a 50% reduction in the Eczema Area and Severity Index scores of 17 of 23 patients, but it was not sufficiently powered for identification of statistical differences between GBR 830 versus placebo. Although there is potential for these 2 drugs to greatly improve the management of severe atopic dermatitis, significant clinical trials have not yet been completed to prove efficacy, and there are not yet any available phase 3 clinical trials, which are needed to truly evaluate their efficacy in affecting T-cells.
Collapse
Affiliation(s)
- W James Tidwell
- Micrographic Surgery and Cutaneous Oncology, Scripps Clinic, La Jolla, California; Division of Dermatology, University of Louisville, Louisville, Kentucky.
| | - Joseph F Fowler
- Division of Dermatology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
99983
|
Carter DM, Darby CA, Johnson SK, Carlock MA, Kirchenbaum GA, Allen JD, Vogel TU, Delagrave S, DiNapoli J, Kleanthous H, Ross TM. Elicitation of Protective Antibodies against a Broad Panel of H1N1 Viruses in Ferrets Preimmune to Historical H1N1 Influenza Viruses. J Virol 2017; 91:e01283-17. [PMID: 28978709 PMCID: PMC5709581 DOI: 10.1128/jvi.01283-17] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/13/2017] [Indexed: 11/20/2022] Open
Abstract
Most preclinical animal studies test influenza vaccines in immunologically naive animal models, even though the results of vaccination may not accurately reflect the effectiveness of vaccine candidates in humans that have preexisting immunity to influenza. In this study, novel, broadly reactive influenza vaccine candidates were assessed in preimmune ferrets. These animals were infected with different H1N1 isolates before being vaccinated or infected with another influenza virus. Previously, our group has described the design and characterization of computationally optimized broadly reactive hemagglutinin (HA) antigens (COBRA) for H1N1 isolates. Vaccinating ferrets with virus-like particle (VLP) vaccines expressing COBRA HA proteins elicited antibodies with hemagglutination inhibition (HAI) activity against more H1N1 viruses in the panel than VLP vaccines expressing wild-type HA proteins. Specifically, ferrets infected with the 1986 virus and vaccinated with a single dose of the COBRA HA VLP vaccines elicited antibodies with HAI activity against 11 to 14 of the 15 H1N1 viruses isolated between 1934 and 2013. A subset of ferrets was infected with influenza viruses expressing the COBRA HA antigens. These COBRA preimmune ferrets had superior breadth of HAI activity after vaccination with COBRA HA VLP vaccines than COBRA preimmune ferrets vaccinated with VLP vaccines expressing wild-type HA proteins. Overall, priming naive ferrets with COBRA HA based viruses or using COBRA HA based vaccines to boost preexisting antibodies induced by wild-type H1N1 viruses, COBRA HA antigens elicited sera with the broadest HAI reactivity against multiple antigenic H1N1 viral variants. This is the first report demonstrating the effectiveness of a broadly reactive or universal influenza vaccine in a preimmune ferret model.IMPORTANCE Currently, many groups are testing influenza vaccine candidates to meet the challenge of developing a vaccine that elicits broadly reactive and long-lasting protective immune responses. The goal of these vaccines is to stimulate immune responses that react against most, if not all, circulating influenza strains, over a long period of time in all populations of people. Commonly, these experimental vaccines are tested in naive animal models that do not have anti-influenza immune responses; however, humans have preexisting immunity to influenza viral antigens, particularly antibodies to the HA and NA glycoproteins. Therefore, this study investigated how preexisting antibodies to historical influenza viruses influenced HAI-specific antibodies and protective efficacy using a broadly protective vaccine candidate.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Ferrets
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/classification
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Donald M Carter
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Christopher A Darby
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Scott K Johnson
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Michael A Carlock
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Greg A Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - James D Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Thorsten U Vogel
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Simon Delagrave
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Joshua DiNapoli
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Harold Kleanthous
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
99984
|
Albertsson AM, Zhang X, Vontell R, Bi D, Bronson RT, Supramaniam V, Baburamani AA, Hua S, Nazmi A, Cardell S, Zhu C, Cantor H, Mallard C, Hagberg H, Leavenworth JW, Wang X. γδ T Cells Contribute to Injury in the Developing Brain. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:757-767. [PMID: 29248460 PMCID: PMC5840494 DOI: 10.1016/j.ajpath.2017.11.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/11/2017] [Accepted: 11/07/2017] [Indexed: 01/04/2023]
Abstract
Brain injury in premature infants, especially periventricular leukomalacia, is an important cause of neurologic disabilities. Inflammation contributes to perinatal brain injury development, but the essential mediators that lead to early-life brain injury remain largely unknown. Neonates have reduced capacity for mounting conventional αβT-cell responses. However, γδT cells are already functionally competent during early development and are important in early-life immunity. We investigated the potential contribution of γδT cells to preterm brain injury using postmortem brains from human preterm infants with periventricular leukomalacia and two animal models of preterm brain injury—the hypoxic-ischemic mouse model and a fetal sheep asphyxia model. Large numbers of γδT cells were observed in the brains of mice, sheep, and postmortem preterm infants after injury, and depletion of γδT cells provided protection in the mouse model. The common γδT-cell–associated cytokines interferon-γ and IL-17A were not detectable in the brain. Although there were increased mRNA levels of Il17f and Il22 in the mouse brains after injury, neither IL-17F nor IL-22 cytokines contributed to preterm brain injury. These findings highlight unique features of injury in the developing brain, where, unlike injury in the mature brain, γδT cells function as initiators of injury independently of common γδT-cell–associated cytokines. This finding will help to identify therapeutic targets for preventing or treating preterm infants with brain injury.
Collapse
Affiliation(s)
- Anna-Maj Albertsson
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoli Zhang
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Provincial Key Laboratory of Child Brain Injury, Zhengzhou, China
| | - Regina Vontell
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Dan Bi
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Roderick T Bronson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts; Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Veena Supramaniam
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Ana A Baburamani
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom
| | - Sha Hua
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Cardiovascular Medicine, Luwan Branch of Shanghai Jiaotong University Medical School Ruijin Hospital, Shanghai, China
| | - Arshed Nazmi
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanna Cardell
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Henan Provincial Key Laboratory of Child Brain Injury, Zhengzhou, China; Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Harvey Cantor
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts; Department of Microbiology and Immunobiology, Division of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Carina Mallard
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Centre for the Developing Brain, Department of Perinatal Imaging and Health, King's College London, St. Thomas' Hospital, London, United Kingdom; Department of Clinical Sciences, East Hospital, Gothenburg, Sweden
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama; Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Xiaoyang Wang
- Perinatal Center, Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
99985
|
Silver J, Zuo T, Chaudhary N, Kumari R, Tong P, Giguere S, Granato A, Donthula R, Devereaux C, Wesemann DR. Stochasticity enables BCR-independent germinal center initiation and antibody affinity maturation. J Exp Med 2017; 215:77-90. [PMID: 29247044 PMCID: PMC5748855 DOI: 10.1084/jem.20171022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/12/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Two immunoglobulin (Ig) diversification mechanisms collaborate to provide protective humoral immunity. Combinatorial assembly of IgH and IgL V region exons from gene segments generates preimmune Ig repertoires, expressed as B cell receptors (BCRs). Secondary diversification occurs when Ig V regions undergo somatic hypermutation (SHM) and affinity-based selection toward antigen in activated germinal center (GC) B cells. Secondary diversification is thought to only ripen the antigen-binding affinity of Igs that already exist (i.e., cognate Igs) because of chance generation during preimmune Ig diversification. However, whether stochastic activation of noncognate B cells can generate new affinity to antigen in GCs is unclear. Using a mouse model whose knock-in BCR does not functionally engage with immunizing antigen, we found that chronic immunization induced antigen-specific serological responses with diverse SHM-mediated antibody affinity maturation pathways and divergent epitope targeting. Thus, intrinsic GC B cell flexibility allows for somatic, noncognate B cell evolution, permitting de novo antigen recognition and subsequent antibody affinity maturation without initial preimmune BCR engagement.
Collapse
Affiliation(s)
- Jared Silver
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Teng Zuo
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Neha Chaudhary
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Rupa Kumari
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Pei Tong
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Sophie Giguere
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Alessandra Granato
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Rakesh Donthula
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Colby Devereaux
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Duane R Wesemann
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
99986
|
Sarkar S, Sabhachandani P, Ravi D, Potdar S, Purvey S, Beheshti A, Evens AM, Konry T. Dynamic Analysis of Human Natural Killer Cell Response at Single-Cell Resolution in B-Cell Non-Hodgkin Lymphoma. Front Immunol 2017; 8:1736. [PMID: 29312292 PMCID: PMC5735063 DOI: 10.3389/fimmu.2017.01736] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/23/2017] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are phenotypically and functionally diverse lymphocytes that recognize and kill cancer cells. The susceptibility of target cancer cells to NK cell-mediated cytotoxicity depends on the strength and balance of regulatory (activating/inhibitory) ligands expressed on target cell surface. We performed gene expression arrays to determine patterns of NK cell ligands associated with B-cell non-Hodgkin lymphoma (b-NHL). Microarray analyses revealed significant upregulation of a multitude of NK-activating and costimulatory ligands across varied b-NHL cell lines and primary lymphoma cells, including ULBP1, CD72, CD48, and SLAMF6. To correlate genetic signatures with functional anti-lymphoma activity, we developed a dynamic and quantitative cytotoxicity assay in an integrated microfluidic droplet generation and docking array. Individual NK cells and target lymphoma cells were co-encapsulated in picoliter-volume droplets to facilitate monitoring of transient cellular interactions and NK cell effector outcomes at single-cell level. We identified significant variability in NK-lymphoma cell contact duration, frequency, and subsequent cytolysis. Death of lymphoma cells undergoing single contact with NK cells occurred faster than cells that made multiple short contacts. NK cells also killed target cells in droplets via contact-independent mechanisms that partially relied on calcium-dependent processes and perforin secretion, but not on cytokines (interferon-γ or tumor necrosis factor-α). We extended this technique to characterize functional heterogeneity in cytolysis of primary cells from b-NHL patients. Tumor cells from two diffuse large B-cell lymphoma patients showed similar contact durations with NK cells; primary Burkitt lymphoma cells made longer contacts and were lysed at later times. We also tested the cytotoxic efficacy of NK-92, a continuously growing NK cell line being investigated as an antitumor therapy, using our droplet-based bioassay. NK-92 cells were found to be more efficient in killing b-NHL cells compared with primary NK cells, requiring shorter contacts for faster killing activity. Taken together, our combined genetic and microfluidic analysis demonstrate b-NHL cell sensitivity to NK cell-based cytotoxicity, which was associated with significant heterogeneity in the dynamic interaction at single-cell level.
Collapse
Affiliation(s)
- Saheli Sarkar
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Pooja Sabhachandani
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Dashnamoorthy Ravi
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Sayalee Potdar
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Sneha Purvey
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Afshin Beheshti
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Andrew M Evens
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| |
Collapse
|
99987
|
Viperin Targets Flavivirus Virulence by Inducing Assembly of Noninfectious Capsid Particles. J Virol 2017; 92:JVI.01751-17. [PMID: 29046456 PMCID: PMC5730767 DOI: 10.1128/jvi.01751-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022] Open
Abstract
Efficient antiviral immunity requires interference with virus replication at multiple layers targeting diverse steps in the viral life cycle. We describe here a novel flavivirus inhibition mechanism that results in interferon-mediated obstruction of tick-borne encephalitis virus particle assembly and involves release of malfunctioning membrane-associated capsid (C) particles. This mechanism is controlled by the activity of the interferon-induced protein viperin, a broad-spectrum antiviral interferon-stimulated gene. Through analysis of the viperin-interactome, we identified the Golgi brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1) as the cellular protein targeted by viperin. Viperin-induced antiviral activity, as well as C-particle release, was stimulated by GBF1 inhibition and knockdown and reduced by elevated levels of GBF1. Our results suggest that viperin targets flavivirus virulence by inducing the secretion of unproductive noninfectious virus particles via a GBF1-dependent mechanism. This as-yet-undescribed antiviral mechanism allows potential therapeutic intervention. IMPORTANCE The interferon response can target viral infection on almost every level; however, very little is known about the interference of flavivirus assembly. We show here that interferon, through the action of viperin, can disturb the assembly of tick-borne encephalitis virus. The viperin protein is highly induced after viral infection and exhibit broad-spectrum antiviral activity. However, the mechanism of action is still elusive and appears to vary between the different viruses, indicating that cellular targets utilized by several viruses might be involved. In this study, we show that viperin induces capsid particle release by interacting and inhibiting the function of the cellular protein Golgi brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1). GBF1 is a key protein in the cellular secretory pathway and is essential in the life cycle of many viruses, also targeted by viperin, implicating GBF1 as a novel putative drug target.
Collapse
|
99988
|
The mitochondrial and death receptor pathways involved in the thymocytes apoptosis induced by aflatoxin B1. Oncotarget 2017; 7:12222-34. [PMID: 26933817 PMCID: PMC4914280 DOI: 10.18632/oncotarget.7731] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/05/2016] [Indexed: 01/05/2023] Open
Abstract
Aflatoxin B1 (AFB1) is a potent immunosuppressive agent in endotherms, which can be related to the up-regulated apoptosis of immune organs. In this study, we investigated the roles of the mitochondrial, death receptor, and endoplasmic reticulum pathways in Aflatoxin B1 induced thymocytes apoptosis. Chickens were fed an aflatoxin B1 containing diet (0.6 mg/kg AFB1) for 3 weeks. Our results showed that (1) AFB1 diet induced the decrease of T-cell subsets, morphological changes, and excessive apoptosis of thymus. (2) The excessive apoptosis involved the mitochondrial pathway (up-regulation of Bax, Bak, cytC and down-regulation of Bcl-2 and Bcl-xL) and death receptor pathway (up-regulation of FasL, Fas and FADD). (3) Oxidative stress, an apoptosis inducer, was confirmed in the thymus. In conclusion, this is the first study to demonstrate that mitochondrial and death receptor pathways involved in AFB1 induced thymocytes apoptosis in broilers.
Collapse
|
99989
|
Efimov GA, Raats JMH, Chirivi RGS, van Rosmalen JWG, Nedospasov SA. Humanization of Murine Monoclonal anti-hTNF Antibody: The F10 Story. Mol Biol 2017. [DOI: 10.1134/s0026893317060061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
99990
|
Guan X, Chaffey PK, Chen H, Feng W, Wei X, Yang LM, Ruan Y, Wang X, Li Y, Barosh KB, Tran AH, Zhu J, Liang W, Zheng YT, Wang X, Tan Z. O-GalNAcylation of RANTES Improves Its Properties as a Human Immunodeficiency Virus Type 1 Entry Inhibitor. Biochemistry 2017; 57:136-148. [PMID: 29202246 DOI: 10.1021/acs.biochem.7b00875] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Many human proteins have the potential to be developed as therapeutic agents. However, side effects caused by direct administration of natural proteins have significantly slowed expansion of protein therapeutics into the clinic. Post-translational modifications (PTMs) can improve protein properties, but because of significant knowledge gaps, we are considerably limited in our ability to apply PTMs to generate better protein therapeutics. Here, we seek to fill the gaps by studying the PTMs of a small representative chemotactic cytokine, RANTES. RANTES can inhibit HIV-1 infection by competing with it for binding to receptor CCR5 and stimulating CCR5 endocytosis. Unfortunately, RANTES can induce strong signaling, leading to severe inflammatory side effects. We apply a chemical biology approach to explore the potential of post-translationally modified RANTES as safe inhibitors of HIV-1 infection. We synthesized and systematically tested a library of RANTES isoforms for their ability to inhibit inflammatory signaling and prevent HIV-1 infection of primary human cells. Through this research, we revealed that most of the glycosylated variants have decreased inflammation-associated properties and identified one particular glyco variant, a truncated RANTES containing a Galβ1-3GalNAc disaccharide α-linked to Ser4, which stands out as having the best overall properties: relatively high HIV-1 inhibition potency but also weak inflammatory properties. Moreover, our results provided a structural basis for the observed changes in the properties of RANTES. Taken together, this work highlights the potential importance of glycosylation as an alternative strategy for developing CCR5 inhibitors to treat HIV-1 infection and, more generally, for reducing or eliminating unwanted properties of therapeutic proteins.
Collapse
Affiliation(s)
- Xiaoyang Guan
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Patrick K Chaffey
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Huan Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223, China
| | - Wei Feng
- Department of Chemistry & Biochemistry, Arizona State University , Tempe, Arizona 85287, United States
| | - Xiuli Wei
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Liu-Meng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223, China
| | - Yuan Ruan
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Xinfeng Wang
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Yaohao Li
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Kimberly B Barosh
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Amy H Tran
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Jaimie Zhu
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| | - Wei Liang
- Protein & Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223, China
| | - Xu Wang
- Department of Chemistry & Biochemistry, Arizona State University , Tempe, Arizona 85287, United States
| | - Zhongping Tan
- Department of Chemistry and Biochemistry and BioFrontiers Institute, University of Colorado , Boulder, Colorado 80303, United States
| |
Collapse
|
99991
|
Wang N, Wang J, Jiang R. Effects of IL-10 on OX62, MHC-II and CD86 in bone marrow DCs in rats with organophosphate poisoning. Exp Ther Med 2017; 15:1906-1909. [PMID: 29434782 PMCID: PMC5776626 DOI: 10.3892/etm.2017.5629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022] Open
Abstract
This study investigated the effect of interleukin (IL)-10 on the expression of OX62, MHC-II and CD86 in bone marrow dendritic cells (DCs) in rats with organophosphorus poisoning. Sixty adult SD rats were randomly divided into normal control group (group A) (20 rats), 20 rats with organophosphorus pesticide poisoning (group B), 20 rats with organophosphorus poisoning, and IL-10 treated (group C). Group A was not treated with special treatment. Group B was treated with 4% omethoate by gavage to establish the model of organophosphate poisoning. Group C was treated with omethoate to establish the model of organophosphate poisoning, then the rats were given intraperitoneal injection of IL-10 for 3 continuous days. Rats were sacrificed after 3 days, bone marrow lymphocytes were extracted, DCs were collected and cultured for 7 days, the expression of DC surface antigen OX62, MHC-II, CD86 and related proteins was detected by flow cytometry and western blotting after cell maturation. The expression of DC surface antigen and corresponding protein increased in group B, and decreased in group C, the difference was statistically significant (P<0.05). The results showed that the expression of OX62, MHC-II and CD86 in bone marrow DCs is enhanced and the cellular immune function is enhanced after organophosphate poisoning. IL-10 can down-regulate the antigen presenting function of DCs, achieve anti-inflammatory effect and assist the treatment of organophosphorus pesticide poisoning.
Collapse
Affiliation(s)
- Ning Wang
- Department of Electrophysiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Jinfeng Wang
- Department of Electrophysiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Ronggang Jiang
- Department of Electrophysiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
99992
|
Han SS, Yang SH, Jo HA, Oh YJ, Park M, Kim JY, Lee H, Lee JP, Lee SH, Joo KW, Lim CS, Kim YS, Kim DK. BAFF and APRIL expression as an autoimmune signature of membranous nephropathy. Oncotarget 2017; 9:3292-3302. [PMID: 29423047 PMCID: PMC5790464 DOI: 10.18632/oncotarget.23232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022] Open
Abstract
Background Based on the fact that B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) have a regulatory role in B cell biology, excessive levels of these cytokines can promote autoimmune pathogenesis. However, the expression and implication remain unresolved in cases of membranous nephropathy (MN). Results The plasma BAFF levels of the primary MN patients were higher than those of healthy controls but lower than those of secondary MN patients, whereas the APRIL levels were similar between the MN patients and healthy controls. The BAFF levels were higher in relapse cases, whereas the APRIL levels were higher in the patients who did not experience remission compared with the counterpart patients. The ectopic expression of BAFF and APRIL was observed in the glomeruli or circulating B cells of MN patients, and this high expression trend was similar to that of lupus patients. Conclusions Expression profile of BAFF and APRIL in MN is similar to that of other autoimmune disease, which affects the kidney outcomes. Methods Plasma BAFF and APRIL levels were measured upon kidney biopsy in patients with primary (n = 89) and secondary MN (n = 13), and the results were compared with the levels in healthy controls (n = 111). The kidney outcomes (e.g., remission and relapse) were traced for the median of 3 years. Aberrant expression of the cytokines was evaluated in the kidney and circulating B cells using immunohistochemistry and flow cytometry analyses, respectively.
Collapse
Affiliation(s)
- Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hyung Ah Jo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Jung Oh
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Minkyoung Park
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Joo Young Kim
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Sang-Ho Lee
- Department of Internal Medicine, College of medicine, Kyung Hee University, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
99993
|
Wu X, Dao Thi VL, Huang Y, Billerbeck E, Saha D, Hoffmann HH, Wang Y, Silva LAV, Sarbanes S, Sun T, Andrus L, Yu Y, Quirk C, Li M, MacDonald MR, Schneider WM, An X, Rosenberg BR, Rice CM. Intrinsic Immunity Shapes Viral Resistance of Stem Cells. Cell 2017; 172:423-438.e25. [PMID: 29249360 DOI: 10.1016/j.cell.2017.11.018] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/11/2017] [Accepted: 11/09/2017] [Indexed: 12/26/2022]
Abstract
Stem cells are highly resistant to viral infection compared to their differentiated progeny; however, the mechanism is mysterious. Here, we analyzed gene expression in mammalian stem cells and cells at various stages of differentiation. We find that, conserved across species, stem cells express a subset of genes previously classified as interferon (IFN) stimulated genes (ISGs) but that expression is intrinsic, as stem cells are refractory to interferon. This intrinsic ISG expression varies in a cell-type-specific manner, and many ISGs decrease upon differentiation, at which time cells become IFN responsive, allowing induction of a broad spectrum of ISGs by IFN signaling. Importantly, we show that intrinsically expressed ISGs protect stem cells against viral infection. We demonstrate the in vivo importance of intrinsic ISG expression for protecting stem cells and their differentiation potential during viral infection. These findings have intriguing implications for understanding stem cell biology and the evolution of pathogen resistance.
Collapse
Affiliation(s)
- Xianfang Wu
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Viet Loan Dao Thi
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yumin Huang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; Department of Hematology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Eva Billerbeck
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Debjani Saha
- Program in Immunogenomics, The Rockefeller University, New York, NY 10065, USA
| | - Hans-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yaomei Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | | | - Stephanie Sarbanes
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Tony Sun
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Linda Andrus
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yingpu Yu
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Corrine Quirk
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Melody Li
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - William M Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Brad R Rosenberg
- Program in Immunogenomics, The Rockefeller University, New York, NY 10065, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
99994
|
Nowlin BT, Wang J, Schafer JL, Autissier P, Burdo TH, Williams KC. Monocyte subsets exhibit transcriptional plasticity and a shared response to interferon in SIV-infected rhesus macaques. J Leukoc Biol 2017; 103:141-155. [PMID: 29345061 DOI: 10.1002/jlb.4a0217-047r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/26/2017] [Accepted: 10/16/2017] [Indexed: 12/24/2022] Open
Abstract
The progression to AIDS is influenced by changes in the biology of heterogeneous monocyte subsets. Classical (CD14++CD16-), intermediate (CD14++CD16+), and nonclassical (CD14+CD16++) monocytes may represent progressive stages of monocyte maturation or disparate myeloid lineages with different turnover rates and function. To investigate the relationship between monocyte subsets and the response to SIV infection, we performed microarray analysis of monocyte subsets in rhesus macaques at three time points: prior to SIV infection, 26 days postinfection, and necropsy with AIDS. Genes with a 2-fold change between monocyte subsets (2023 genes) or infection time points (424 genes) were selected. We identify 172 genes differentially expressed among monocyte subsets in both uninfected and SIV-infected animals. Classical monocytes express genes associated with inflammatory responses and cell proliferation. Nonclassical monocytes express genes associated with activation, immune effector functions, and cell cycle inhibition. The classical and intermediate subsets are most similar at all time points, and transcriptional similarity between intermediate and nonclassical monocytes increases with AIDS. Cytosolic sensors of nucleic acids, restriction factors, and IFN-stimulated genes are induced in all three subsets with AIDS. We conclude that SIV infection alters the transcriptional relationship between monocyte subsets and that the innate immune response to SIV infection is conserved across monocyte subsets.
Collapse
Affiliation(s)
- Brian T Nowlin
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - John Wang
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Jamie L Schafer
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Patrick Autissier
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Tricia H Burdo
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Kenneth C Williams
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
99995
|
Miyake T, Miyake T, Sakaguchi M, Nankai H, Nakazawa T, Morishita R. Prevention of Asthma Exacerbation in a Mouse Model by Simultaneous Inhibition of NF-κB and STAT6 Activation Using a Chimeric Decoy Strategy. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:159-169. [PMID: 29499930 PMCID: PMC5751966 DOI: 10.1016/j.omtn.2017.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 11/24/2022]
Abstract
Transactivation of inflammatory and immune mediators in asthma is tightly regulated by nuclear factor κB (NF-κB) and signal transducer and activator of transcription 6 (STAT6). Therefore, we investigated the efficacy of simultaneous inhibition of NF-κB and STAT6 using a chimeric decoy strategy to prevent asthma exacerbation. The effects of decoy oligodeoxynucleotides were evaluated using an ovalbumin-induced mouse asthma model. Ovalbumin-sensitized mice received intratracheal administration of decoy oligodeoxynucleotides 3 days before ovalbumin challenge. Fluorescent-dye-labeled decoy oligodeoxynucleotides could be detected in lymphocytes and macrophages in the lung, and activation of NF-κB and STAT6 was inhibited by chimeric decoy oligodeoxynucleotide transfer. Consequently, treatment with chimeric or NF-κB decoy oligodeoxynucleotides protected against methacholine-induced airway hyperresponsiveness, whereas the effect of chimeric decoy oligodeoxynucleotides was significantly greater than that of NF-κB decoy oligodeoxynucleotides. Treatment with chimeric decoy oligodeoxynucleotides suppressed airway inflammation through inhibition of overexpression of interleukin-4 (IL-4), IL-5, and IL-13 and inflammatory infiltrates. Histamine levels in the lung were reduced via suppression of mast cell accumulation. A significant reduction in mucin secretion was observed due to suppression of MUC5AC gene expression. Interestingly, the inhibitory effects on IL-5, IL-13, and histamine secretion were achieved by transfer of chimeric decoy oligodeoxynucleotides only. This novel therapeutic approach could be useful to treat patients with various types of asthma.
Collapse
Affiliation(s)
- Tetsuo Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Takashi Miyake
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| | | | | | | | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
99996
|
Costea PI, Coelho LP, Sunagawa S, Munch R, Huerta-Cepas J, Forslund K, Hildebrand F, Kushugulova A, Zeller G, Bork P. Subspecies in the global human gut microbiome. Mol Syst Biol 2017; 13:960. [PMID: 29242367 PMCID: PMC5740502 DOI: 10.15252/msb.20177589] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 02/06/2023] Open
Abstract
Population genomics of prokaryotes has been studied in depth in only a small number of primarily pathogenic bacteria, as genome sequences of isolates of diverse origin are lacking for most species. Here, we conducted a large-scale survey of population structure in prevalent human gut microbial species, sampled from their natural environment, with a culture-independent metagenomic approach. We examined the variation landscape of 71 species in 2,144 human fecal metagenomes and found that in 44 of these, accounting for 72% of the total assigned microbial abundance, single-nucleotide variation clearly indicates the existence of sub-populations (here termed subspecies). A single subspecies (per species) usually dominates within each host, as expected from ecological theory. At the global scale, geographic distributions of subspecies differ between phyla, with Firmicutes subspecies being significantly more geographically restricted. To investigate the functional significance of the delineated subspecies, we identified genes that consistently distinguish them in a manner that is independent of reference genomes. We further associated these subspecies-specific genes with properties of the microbial community and the host. For example, two of the three Eubacterium rectale subspecies consistently harbor an accessory pro-inflammatory flagellum operon that is associated with lower gut community diversity, higher host BMI, and higher blood fasting insulin levels. Using an additional 676 human oral samples, we further demonstrate the existence of niche specialized subspecies in the different parts of the oral cavity. Taken together, we provide evidence for subspecies in the majority of abundant gut prokaryotes, leading to a better functional and ecological understanding of the human gut microbiome in conjunction with its host.
Collapse
Affiliation(s)
- Paul I Costea
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Luis Pedro Coelho
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Shinichi Sunagawa
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Robin Munch
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jaime Huerta-Cepas
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Kristoffer Forslund
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Falk Hildebrand
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max-Delbrück-Centre for Molecular Medicine, Berlin, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
99997
|
Hou R, Han Y, Fei Q, Gao Y, Qi R, Cai R, Qi Y. Dietary Flavone Tectochrysin Exerts Anti-Inflammatory Action by Directly Inhibiting MEK1/2 in LPS-Primed Macrophages. Mol Nutr Food Res 2017; 62. [DOI: 10.1002/mnfr.201700288] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/28/2017] [Indexed: 01/25/2023]
Affiliation(s)
- Rui Hou
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Yixin Han
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Qiaoling Fei
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Yuan Gao
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Ruijuan Qi
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Runlan Cai
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Yun Qi
- Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| |
Collapse
|
99998
|
He S, Liu Y, Meng L, Sun H, Wang Y, Ji Y, Purushe J, Chen P, Li C, Madzo J, Issa JP, Soboloff J, Reshef R, Moore B, Gattinoni L, Zhang Y. Ezh2 phosphorylation state determines its capacity to maintain CD8 + T memory precursors for antitumor immunity. Nat Commun 2017; 8:2125. [PMID: 29242551 PMCID: PMC5730609 DOI: 10.1038/s41467-017-02187-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022] Open
Abstract
Memory T cells sustain effector T-cell production while self-renewing in reaction to persistent antigen; yet, excessive expansion reduces memory potential and impairs antitumor immunity. Epigenetic mechanisms are thought to be important for balancing effector and memory differentiation; however, the epigenetic regulator(s) underpinning this process remains unknown. Herein, we show that the histone methyltransferase Ezh2 controls CD8+ T memory precursor formation and antitumor activity. Ezh2 activates Id3 while silencing Id2, Prdm1 and Eomes, promoting the expansion of memory precursor cells and their differentiation into functional memory cells. Akt activation phosphorylates Ezh2 and decreases its control of these transcriptional programs, causing enhanced effector differentiation at the expense of T memory precursors. Engineering T cells with an Akt-insensitive Ezh2 mutant markedly improves their memory potential and capability of controlling tumor growth compared to transiently inhibiting Akt. These findings establish Akt-mediated phosphorylation of Ezh2 as a critical target to potentiate antitumor immunotherapeutic strategies. During an immune response naive CD8+ T cells can differentiate into either effector or memory T cells. Here the authors show that Akt-mediated phosphorylation of the epigenetic regulator Ezh2 is critical for the generation of an anti-tumor CD8 T cell response and promotes the expansion of memory-precursors.
Collapse
Affiliation(s)
- Shan He
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA.
| | - Yongnian Liu
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Lijun Meng
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Hongxing Sun
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Yun Ji
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Janaki Purushe
- Department of Microbiology & Immunology, Temple University, Philadelphia, PA, 19140, USA
| | - Pan Chen
- The Division of Endocrinology and the Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Changhong Li
- The Division of Endocrinology and the Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jozef Madzo
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Jean-Pierre Issa
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Ran Reshef
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Bethany Moore
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Luca Gattinoni
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA. .,Department of Microbiology & Immunology, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
99999
|
Interleukins 12 and 15 induce cytotoxicity and early NK-cell differentiation in type 3 innate lymphoid cells. Blood Adv 2017; 1:2679-2691. [PMID: 29296921 DOI: 10.1182/bloodadvances.2017008839] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 11/19/2017] [Indexed: 02/06/2023] Open
Abstract
Type 3 innate lymphoid cells (ILC3s) fulfill protective functions at mucosal surfaces via cytokine production. Although their plasticity to become ILC1s, the innate counterparts of type 1 helper T cells, has been described previously, we report that they can differentiate into cytotoxic lymphocytes with many characteristics of early differentiated natural killer (NK) cells. This transition is promoted by the proinflammatory cytokines interleukin 12 (IL-12) and IL-15, and correlates with expression of the master transcription factor of cytotoxicity, eomesodermin (Eomes). As revealed by transcriptome analysis and flow cytometric profiling, differentiated ILC3s express CD94, NKG2A, NKG2C, CD56, and CD16 among other NK-cell receptors, and possess all components of the cytotoxic machinery. These characteristics allow them to recognize and kill leukemic cells with perforin and granzymes. Therefore, ILC3s can be harnessed for cytotoxic responses via differentiation under the influence of proinflammatory cytokines.
Collapse
|
100000
|
Wei S. Yin-yang regulating effects of cancer-associated genes, proteins, and cells: An ancient Chinese concept in vogue in modern cancer research. Biosci Trends 2017; 11:612-618. [PMID: 29238002 DOI: 10.5582/bst.2017.01259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Great achievements have been made in human cancer research, but most of this research is focused on conditions at the microscopic rather than the systemic level. Recent studies have increasingly cited the ancient Chinese theory of yin-yang in an effort to expand beyond the microscopic level. Various cancer-associated genes and proteins such as mitogen-activated protein kinase (MAPK), p38, p53, c-Myc, tumor necrosis factor (TNF)-α, NF-κB, Cyclin D1, and cyclin-dependent kinase (CDK) and cells such as T cells, B cells, macrophages, neutrophils, and fibroblasts have been reported to regulate various types of cancers in a yin-yang manner. These studies have brought the theory of yin-yang into vogue in cancer research worldwide.
Collapse
Affiliation(s)
- Shuyong Wei
- College of Animal Science, Southwest University
| |
Collapse
|