1
|
Kotey SK, Tan X, Kinser AL, Liu L, Cheng Y. Host Long Noncoding RNAs as Key Players in Mycobacteria-Host Interactions. Microorganisms 2024; 12:2656. [PMID: 39770858 PMCID: PMC11728548 DOI: 10.3390/microorganisms12122656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Mycobacterial infections, caused by various species within the Mycobacterium genus, remain one of the main challenges to global health across the world. Understanding the complex interplay between the host and mycobacterial pathogens is essential for developing effective diagnostic and therapeutic strategies. Host long noncoding RNAs (lncRNAs) have emerged as key regulators in cellular response to bacterial infections within host cells. This review provides an overview of the intricate relationship between mycobacterial infections and host lncRNAs in the context of Mycobacterium tuberculosis and non-tuberculous mycobacterium (NTM) infections. Accumulation of evidence indicates that host lncRNAs play a critical role in regulating cellular response to mycobacterial infection within host cells, such as macrophages, the primary host cells for mycobacterial intracellular survival. The expression of specific host lncRNAs has been implicated in the pathogenesis of mycobacterial infections, providing potential targets for the development of novel host-directed therapies and biomarkers for TB diagnosis. In summary, this review aims to highlight the current state of knowledge regarding the involvement of host lncRNAs in mycobacterial infections. It also emphasizes their potential application as novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Stephen K. Kotey
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Xuejuan Tan
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Audrey L. Kinser
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Yong Cheng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| |
Collapse
|
2
|
Ferreira DA, Medeiros ABA, Soares MM, Lima ÉDA, de Oliveira GCSL, Leite MBDS, Machado MV, Villar JAFP, Barbosa LA, Scavone C, Moura MT, Rodrigues-Mascarenhas S. Evaluation of Anti-Inflammatory Activity of the New Cardiotonic Steroid γ-Benzylidene Digoxin 8 (BD-8) in Mice. Cells 2024; 13:1568. [PMID: 39329752 PMCID: PMC11430542 DOI: 10.3390/cells13181568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Cardiotonic steroids are known to bind to Na+/K+-ATPase and regulate several biological processes, including the immune response. The synthetic cardiotonic steroid γ-Benzylidene Digoxin 8 (BD-8) is emerging as a promising immunomodulatory molecule, although it has remained largely unexplored. Therefore, we tested the immunomodulatory potential of BD-8 both in vitro and in vivo. Hence, primary mouse macrophages were incubated with combinations of BD-8 and the pro-inflammatory fungal protein zymosan (ZYM). Nitric oxide (NO) production was determined by Griess reagent and cytokines production was assessed by enzyme-linked immunosorbent assay. Inducible nitric oxide synthase (iNOS), reactive oxygen species (ROS), p-nuclear factor kappa B p65 (NF-κB p65), p-extracellular signal-regulated kinase (p-ERK), and p-p38 were evaluated by flow cytometry. Macrophages exposed to BD-8 displayed reduced phagocytic activity, NO levels, and production of the proinflammatory cytokine IL-1β induced by ZYM. Furthermore, BD-8 diminished the expression of iNOS and phosphorylation of NF-κB p65, ERK, and p38. Additionally, BD-8 exhibited anti-inflammatory capacity in vivo in a carrageenan-induced mouse paw edema model. Taken together, these findings demonstrate the anti-inflammatory activity of BD-8 and further reinforce the potential of cardiotonic steroids and their derivatives as immunomodulatory molecules.
Collapse
Affiliation(s)
- Davi Azevedo Ferreira
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Anna Beatriz Araujo Medeiros
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Mariana Mendonça Soares
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Éssia de Almeida Lima
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Gabriela Carolina Santos Lima de Oliveira
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Mateus Bernardo da Silva Leite
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| | - Matheus Vieira Machado
- Laboratory of Cellular Biochemistry, Campus Centro-Oeste Dona Lindú, Federal University of São João del-Rei, Divinópolis 35.501-296, MG, Brazil; (M.V.M.); (J.A.F.P.V.); (L.A.B.)
| | - José Augusto Ferreira Perez Villar
- Laboratory of Cellular Biochemistry, Campus Centro-Oeste Dona Lindú, Federal University of São João del-Rei, Divinópolis 35.501-296, MG, Brazil; (M.V.M.); (J.A.F.P.V.); (L.A.B.)
| | - Leandro Augusto Barbosa
- Laboratory of Cellular Biochemistry, Campus Centro-Oeste Dona Lindú, Federal University of São João del-Rei, Divinópolis 35.501-296, MG, Brazil; (M.V.M.); (J.A.F.P.V.); (L.A.B.)
| | - Cristoforo Scavone
- Laboratory of Neuropharmacology Research, Department of Pharmacology, Institute of Biomedical Sciences ICB-1, University of São Paulo, São Paulo 05.508-900, SP, Brazil;
| | - Marcelo Tigre Moura
- Laboratory of Cellular Reprogramming, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil;
| | - Sandra Rodrigues-Mascarenhas
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraiba, João Pessoa 58.051-900, PB, Brazil; (D.A.F.); (A.B.A.M.); (M.M.S.); (É.d.A.L.); (G.C.S.L.d.O.); (M.B.d.S.L.)
| |
Collapse
|
3
|
Cassiano BA, Silveira ALPA, Kim YJ, do Amaral JB, da Silva Nali LH, Bachi ALL, Resende LD, Fonseca FAH, de Oliveira Izar MC, Tuleta ID, Victor JR, Pallos D, França CN. Role of circulating microparticles and cytokines in periodontitis associated with diabetes. Front Med (Lausanne) 2024; 11:1394300. [PMID: 39253540 PMCID: PMC11381390 DOI: 10.3389/fmed.2024.1394300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Background Periodontitis is a chronic inflammatory condition that affects the supporting tissues of the teeth, and can lead to serious complications such as tooth loss and systemic health problems, including diabetes, which have a bidirectional relationship with periodontitis. Circulating microparticles originate from different cell types after stimuli such as activation or apoptosis. Interleukins are related to processes in the regulation of the immune response, inflammation, and cell growth. This study aimed to evaluate circulating microparticles as well as interleukins in the plasma, at baseline and 1 month after the end of the non-surgical periodontal treatment. Methods Samples were collected from 45 patients, with moderate to severe periodontitis with diabetes (N = 25) and without diabetes (N = 20). Microparticles were evaluated in the platelet-poor plasma by flow cytometer. Cytokine levels were evaluated by the enzyme immunoabsorption assay (ELISA). Results Higher levels of the pro-inflammatory cytokines were found in the group with diabetes compared to the non-diabetic group both at baseline and 1 month after the end of the treatment. A higher IL-6/IL-10 ratio was found in patients with diabetes compared to the group without diabetes at T0 and T1, whereas an increased IFN-γ/IL-10 ratio was only found at T1 in patients with diabetes in comparison to the group without diabetes. In the group with diabetes, it was verified positive correlations between IL-10 and IL-6 or IFN-γ and a negative correlation between IL-6 and PMP, at T0; in contrast, in the T1, negative correlations were found between TNF-α and IL-10 or PMP. Besides, at T0, it was evidenced positive correlations both between circulating TNF-α and IL-6, and IL-10 and EMP, as well as a negative correlation between IL-10 and PMP in the group with diabetes. In addition, it was observed in T1 positive correlations between levels of TNF-α and IL-6, IFN-γ, or IL-10, and between PMP and IFN-γ, and between EMP and IL-6, TNF-α and IFN-γ in this group. Conclusion The results suggest a modulatory effect of the periodontitis associated with diabetes, as well as the periodontal treatment, in the systemic inflammatory status of the participants of the study.
Collapse
Affiliation(s)
| | | | - Yeon Jung Kim
- Odontology Post Graduation, Santo Amaro University, São Paulo, Brazil
| | - Jônatas Bussador do Amaral
- ENT Research Laboratory, Otorhinolaryngology-Head and Neck Surgery Department, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | - Izabela Dorota Tuleta
- Department of Medicine-Cardiology, Albert Einstein College of Medicine, New York, NY, United States
| | | | - Débora Pallos
- Odontology Post Graduation, Santo Amaro University, São Paulo, Brazil
| | | |
Collapse
|
4
|
Gnanagobal H, Chakraborty S, Vasquez I, Chukwu-Osazuwa J, Cao T, Hossain A, Dang M, Valderrama K, Kumar S, Bindea G, Hill S, Boyce D, Hall JR, Santander J. Transcriptome profiling of lumpfish (Cyclopterus lumpus) head kidney to Renibacterium salmoninarum at early and chronic infection stages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 156:105165. [PMID: 38499166 DOI: 10.1016/j.dci.2024.105165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Renibacterium salmoninarum causes Bacterial Kidney Disease (BKD) in several fish species. Atlantic lumpfish, a cleaner fish, is susceptible to R. salmoninarum. To profile the transcriptome response of lumpfish to R. salmoninarum at early and chronic infection stages, fish were intraperitoneally injected with either a high dose of R. salmoninarum (1 × 109 cells dose-1) or PBS (control). Head kidney tissue samples were collected at 28- and 98-days post-infection (dpi) for RNA sequencing. Transcriptomic profiling identified 1971 and 139 differentially expressed genes (DEGs) in infected compared with control samples at 28 and 98 dpi, respectively. At 28 dpi, R. salmoninarum-induced genes (n = 434) mainly involved in innate and adaptive immune response-related pathways, whereas R. salmoninarum-suppressed genes (n = 1537) were largely connected to amino acid metabolism and cellular processes. Cell-mediated immunity-related genes showed dysregulation at 98 dpi. Several immune-signalling pathways were dysregulated in response to R. salmoninarum, including apoptosis, alternative complement, JAK-STAT signalling, and MHC-I dependent pathways. In summary, R. salmoninarum causes immune suppression at early infection, whereas lumpfish induce a cell-mediated immune response at chronic infection. This study provides a complete depiction of diverse immune mechanisms dysregulated by R. salmoninarum in lumpfish and opens new avenues to develop immune prophylactic tools to prevent BKD.
Collapse
Affiliation(s)
- Hajarooba Gnanagobal
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada.
| | - Setu Chakraborty
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ignacio Vasquez
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Joy Chukwu-Osazuwa
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Trung Cao
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ahmed Hossain
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - My Dang
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Katherine Valderrama
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Surendra Kumar
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada; Ocean Frontier Institute, Ocean Sciences Centre, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Gabriela Bindea
- INSERM, Laboratory of Integrative Cancer Immunology, 75006, Paris, France; Equipe Labellisée Ligue Contre Le Cancer, 75013, Paris, France; Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Stephen Hill
- Cold-Ocean Deep-Sea Research Facility, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, A1C 5S7, Canada
| | - Danny Boyce
- The Dr. Joe Brown Aquatic Research Building (JBARB), Ocean Sciences Centre, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jennifer R Hall
- Aquatic Research Cluster, CREAIT Network, Ocean Sciences Centre, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Javier Santander
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
5
|
Zhang Q, Yang G, Luo Y, Jiang L, Chi H, Tian G. Neuroinflammation in Alzheimer's disease: insights from peripheral immune cells. Immun Ageing 2024; 21:38. [PMID: 38877498 PMCID: PMC11177389 DOI: 10.1186/s12979-024-00445-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Alzheimer's disease (AD) is a serious brain disorder characterized by the presence of beta-amyloid plaques, tau pathology, inflammation, neurodegeneration, and cerebrovascular dysfunction. The presence of chronic neuroinflammation, breaches in the blood-brain barrier (BBB), and increased levels of inflammatory mediators are central to the pathogenesis of AD. These factors promote the penetration of immune cells into the brain, potentially exacerbating clinical symptoms and neuronal death in AD patients. While microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in AD, recent evidence suggests the infiltration of cerebral vessels and parenchyma by peripheral immune cells, including neutrophils, T lymphocytes, B lymphocytes, NK cells, and monocytes in AD. These cells participate in the regulation of immunity and inflammation, which is expected to play a huge role in future immunotherapy. Given the crucial role of peripheral immune cells in AD, this article seeks to offer a comprehensive overview of their contributions to neuroinflammation in the disease. Understanding the role of these cells in the neuroinflammatory response is vital for developing new diagnostic markers and therapeutic targets to enhance the diagnosis and treatment of AD patients.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, USA
| | - Yuan Luo
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China.
| | - Gang Tian
- Department of Laboratory Medicine, Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, China.
| |
Collapse
|
6
|
Huo M, Rai SK, Nakatsu K, Deng Y, Jijiwa M. Subverting the Canon: Novel Cancer-Promoting Functions and Mechanisms for snoRNAs. Int J Mol Sci 2024; 25:2923. [PMID: 38474168 PMCID: PMC10932220 DOI: 10.3390/ijms25052923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Small nucleolar RNAs (snoRNAs) constitute a class of intron-derived non-coding RNAs ranging from 60 to 300 nucleotides. Canonically localized in the nucleolus, snoRNAs play a pivotal role in RNA modifications and pre-ribosomal RNA processing. Based on the types of modifications they involve, such as methylation and pseudouridylation, they are classified into two main families-box C/D and H/ACA snoRNAs. Recent investigations have revealed the unconventional synthesis and biogenesis strategies of snoRNAs, indicating their more profound roles in pathogenesis than previously envisioned. This review consolidates recent discoveries surrounding snoRNAs and provides insights into their mechanistic roles in cancer. It explores the intricate interactions of snoRNAs within signaling pathways and speculates on potential therapeutic solutions emerging from snoRNA research. In addition, it presents recent findings on the long non-coding small nucleolar RNA host gene (lncSNHG), a subset of long non-coding RNAs (lncRNAs), which are the transcripts of parental SNHGs that generate snoRNA. The nucleolus, the functional epicenter of snoRNAs, is also discussed. Through a deconstruction of the pathways driving snoRNA-induced oncogenesis, this review aims to serve as a roadmap to guide future research in the nuanced field of snoRNA-cancer interactions and inspire potential snoRNA-related cancer therapies.
Collapse
Affiliation(s)
- Matthew Huo
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA;
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (S.K.R.); (K.N.)
| | - Sudhir Kumar Rai
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (S.K.R.); (K.N.)
| | - Ken Nakatsu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (S.K.R.); (K.N.)
- Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (S.K.R.); (K.N.)
| | - Mayumi Jijiwa
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (S.K.R.); (K.N.)
| |
Collapse
|
7
|
Giannuzzi D, Capra E, Bisutti V, Vanzin A, Marsan PA, Cecchinato A, Pegolo S. Methylome-wide analysis of milk somatic cells upon subclinical mastitis in dairy cattle. J Dairy Sci 2024; 107:1805-1820. [PMID: 37939836 DOI: 10.3168/jds.2023-23821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023]
Abstract
Better understanding of the molecular mechanisms behind bovine mastitis is fundamental for improving the management of this disease, which continues to be of major concern for the dairy industry, especially in its subclinical form. Disease severity and progression depend on numerous aspects, such as livestock genetics, and the interaction between the causative agent, the host, and the environment. In this context, epigenetic mechanisms have proven to have a role in controlling the response of the animal to inflammation. Therefore, in this study we aimed to explore genome-wide DNA methylation of milk somatic cells (SC) in healthy cows (n = 15) and cows affected by naturally occurring subclinical mastitis by Streptococcus agalactiae (n = 12) and Prototheca spp. (n = 11), to better understand the role of SC methylome in the host response to disease. Differentially methylated regions (DMR) were evaluated comparing: (1) Strep. agalactiae-infected versus healthy; (2) Prototheca-infected versus healthy, and (3) mastitis versus healthy and (4) Strep. agalactiae-infected versus Prototheca-infected. The functional analysis was performed at 2 levels. To begin with, we extracted differentially methylated genes (DMG) from promoter DMR, which were analyzed using the Cytoscape ClueGO plug-in. Coupled with this DMG-driven approach, all the genes associated with promoter-methylated regions were fed to the Pathifier algorithm. From the DMR analysis, we identified 1,081 hypermethylated and 361 hypomethylated promoter regions in Strep. agalactiae-infected animals, while 1,514 hypermethylated and 358 hypomethylated promoter regions were identified in Prototheca-infected animals, when compared with the healthy controls. When considering infected animals as a whole group (regardless of the pathogen), we found 1,576 hypermethylated and 460 hypomethylated promoter regions. Both pathogens were associated with methylation differences in genes involved in pathways related to meiosis, reproduction and tissue remodeling. Exploring the whole methylome, in subclinically infected cows we observed a strong deregulation of immune-related pathways, such as nuclear factor kB and toll-like receptors signaling pathways, and of energy-related pathways such as the tricarboxylic acid cycle and unsaturated fatty acid biosynthesis. In conclusion, no evident pathogen-specific SC methylome signature was detected in the present study. Overall, we observed a clear regulation of host immune response driven by DNA methylation upon subclinical mastitis. Further studies on a larger cohort of animals are needed to validate our results and to possibly identify a unique SC methylome that signifies pathogen-specific alterations.
Collapse
Affiliation(s)
- D Giannuzzi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| | - E Capra
- Institute of Agricultural Biology and Biotechnology, National Research Council (IBBA CNR), 26900, Lodi, Italy
| | - V Bisutti
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy.
| | - A Vanzin
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| | - P Ajmone Marsan
- Department of Animal Science, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - A Cecchinato
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| | - S Pegolo
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020, Legnaro, Italy
| |
Collapse
|
8
|
Minucci SB, Heise RL, Reynolds AM. Agent-based vs. equation-based multi-scale modeling for macrophage polarization. PLoS One 2024; 19:e0270779. [PMID: 38271449 PMCID: PMC10810539 DOI: 10.1371/journal.pone.0270779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/29/2023] [Indexed: 01/27/2024] Open
Abstract
Macrophages show high plasticity and result in heterogenic subpopulations or polarized states identified by specific cellular markers. These immune cells are typically characterized as pro-inflammatory, or classically activated M1, and anti-inflammatory, or alternatively activated M2. However, a more precise definition places them along a spectrum of activation where they may exhibit a number of pro- or anti-inflammatory roles. To understand M1-M2 dynamics in the context of a localized response and explore the results of different mathematical modeling approaches based on the same biology, we utilized two different modeling techniques, ordinary differential equation (ODE) modeling and agent-based modeling (ABM), to simulate the spectrum of macrophage activation to general pro- and anti-inflammatory stimuli on an individual and multi-cell level. The ODE model includes two hallmark pro- and anti-inflammatory signaling pathways and the ABM incorporates similar M1-M2 dynamics but in a spatio-temporal platform. Both models link molecular signaling with cellular-level dynamics. We then performed simulations with various initial conditions to replicate different experimental setups. Similar results were observed in both models after tuning to a common calibrating experiment. Comparing the two models' results sheds light on the important features of each modeling approach. When more data is available these features can be considered when choosing techniques to best fit the needs of the modeler and application.
Collapse
Affiliation(s)
- Sarah B. Minucci
- Department of Mathematics & Applied Mathematics, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Rebecca L. Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States of America
| | - Angela M. Reynolds
- Department of Mathematics & Applied Mathematics, Virginia Commonwealth University, Richmond, VA, United States of America
| |
Collapse
|
9
|
Hammersen J, Birndt S, Döhner K, Reuken P, Stallmach A, Sauerbrey P, La Rosée F, Pfirrmann M, Fabisch C, Weiss M, Träger K, Bremer H, Russo S, Illerhaus G, Drömann D, Schneider S, La Rosée P, Hochhaus A. The JAK1/2 inhibitor ruxolitinib in patients with COVID-19 triggered hyperinflammation: the RuxCoFlam trial. Leukemia 2023; 37:1879-1886. [PMID: 37507425 PMCID: PMC10457200 DOI: 10.1038/s41375-023-01979-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Dysregulated hyperinflammatory response is key in the pathogenesis in patients with severe COVID-19 leading to acute respiratory distress syndrome and multiorgan failure. Whilst immunosuppression has been proven to be effective, potential biological targets and optimal timing of treatment are still conflicting. We sought to evaluate efficacy and safety of the Janus Kinase 1/2 inhibitor ruxolitinib, employing the previously developed COVID-19 Inflammation Score (CIS) in a prospective multicenter open label phase II trial (NCT04338958). Primary objective was reversal of hyperinflammation (CIS reduction of ≥25% at day 7 in ≥20% of patients). In 184 patients with a CIS of ≥10 (median 12) ruxolitinib was commenced at an initial dose of 10 mg twice daily and applied over a median of 14 days (range, 2-31). On day 7, median CIS declined to 6 (range, 1-13); 71% of patients (CI 64-77%) achieved a ≥25% CIS reduction accompanied by a reduction of markers of inflammation. Median cumulative dose was 272.5 mg/d. Treatment was well tolerated without any grade 3-5 adverse events related to ruxolitinib. Forty-four patients (23.9%) died, all without reported association to study drug. In conclusion, ruxolitinib proved to be safe and effective in a cohort of COVID-19 patients with defined hyperinflammation.
Collapse
Affiliation(s)
- J Hammersen
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany
| | - S Birndt
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany
| | - K Döhner
- Universitätsklinikum Ulm, Klinik für Innere Medizin III, Hämatologie, Onkologie, Palliativmedizin, Rheumatologie und Infektionskrankheiten, Ulm, Germany
| | - P Reuken
- Universitätsklinikum Jena, Klinik für Innere Medizin IV, Gastroenterologie, Hepatologie, Infektiologie, Interdisziplinäre Endoskopie, Jena, Germany
| | - A Stallmach
- Universitätsklinikum Jena, Klinik für Innere Medizin IV, Gastroenterologie, Hepatologie, Infektiologie, Interdisziplinäre Endoskopie, Jena, Germany
| | - P Sauerbrey
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany
| | - F La Rosée
- Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - M Pfirrmann
- Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie (IBE), Medizinische Fakultät, Ludwig-Maximilians-Universität München, München, Germany
| | - C Fabisch
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany
| | - M Weiss
- Universitätsklinikum Ulm, Klinik für Anästhesiologie und Intensivmedizin, Ulm, Germany
| | - K Träger
- Universitätsklinikum Ulm, Klinik für Anästhesiologie und Intensivmedizin, Ulm, Germany
| | - H Bremer
- Schwarzwald-Baar Klinikum, Lungenzentrum Donaueschingen, Donaueschingen, Germany
| | - S Russo
- Schwarzwald-Baar Klinikum, Klinik für Anästhesiologie, Intensiv-, Notfall- und Schmerzmedizin, Villingen-Schwenningen, Germany
| | - G Illerhaus
- Klinikum Stuttgart, Klinik für Hämatologie, Onkologie, Stammzelltransplantation und Palliativmedizin, Stuttgart, Germany
| | - D Drömann
- Universitätsklinikum Schleswig-Holstein, Medizinische Klinik III, Pulmologie, Lübeck, Germany
| | - S Schneider
- SRH Klinikum Gera, Klinik für Pneumologie/Infektiologie, Hämatologie/Onkologie, Rheumatologie, Gera, Germany
| | - P La Rosée
- Schwarzwald-Baar Klinikum, Klinik für Innere Medizin II, Hämatologie, Onkologie, Immunologie, Infektiologie und Palliativmedizin, Villingen-Schwenningen, Germany
| | - A Hochhaus
- Universitätsklinikum Jena, Klinik für Innere Medizin II, Hämatologie und Internistische Onkologie, Jena, Germany.
| |
Collapse
|
10
|
Wohlrab J, Kegel T, Große R, Eichner A. Handlungsempfehlungen zur Risikominimierung beim Einsatz von Januskinase-Inhibitoren zur Therapie chronisch-entzündlicher Hauterkrankungen. J Dtsch Dermatol Ges 2023; 21:845-852. [PMID: 37574686 DOI: 10.1111/ddg.15136_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/25/2023] [Indexed: 08/15/2023]
Abstract
ZusammenfassungDer Ausschuss für Risikobewertung (PRAC) der Europäischen Arzneimittelagentur (EMA) hat gemäß Artikel 20 der Verordnung (EG) Nr. 726/2004 die Sicherheit für Januskinase‐Inhibitoren für die Behandlung von Entzündungserkrankungen neu bewertet und von den bisherigen Hinweisen in den jeweiligen Fachinformationen der betreffenden Präparate abweichende Sicherheitsangaben formuliert. Diese beziehen sich arzneistoff‐ und indikationsübergreifend auf die Beachtung eines möglicherweise erhöhten Risikos für venöse thromboembolische oder schwere kardiovaskuläre Ereignisse, eine erhöhte Infektionsrate sowie eine Erhöhung der Prävalenz von Hautkrebs. Deshalb wird empfohlen, bei Patienten mit unabhängigen Risikofaktoren (Alter ab 65 Jahre, Raucher oder ehemalige Raucher, Patientinnen mit oraler Kontrazeption beziehungsweise Hormonersatztherapie sowie anderen Risikofaktoren) Januskinase‐Inhibitoren nur dann therapeutisch einzusetzen, wenn es keine geeigneten Behandlungsalternativen gibt. Um im klinischen Alltag eine pragmatische und sorgfältige Erfassung von Risikopatienten zu ermöglichen, wurde interdisziplinär eine Checkliste erarbeitet, die aus der Perspektive des Dermatologen als Arbeitsmittel geeignet ist.
Collapse
Affiliation(s)
- Johannes Wohlrab
- Universitätsklinik für Dermatologie und Venerologie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale)
- Institut für angewandte Dermatopharmazie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale)
| | - Thomas Kegel
- Universitätsklinik für Innere Medizin IV (Hämatologie und Onkologie), Martin-Luther-Universität Halle-Wittenberg, Halle (Saale)
| | - Regina Große
- Universitätsklinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale)
| | - Adina Eichner
- Universitätsklinik für Dermatologie und Venerologie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale)
- Institut für angewandte Dermatopharmazie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale)
| |
Collapse
|
11
|
Wohlrab J, Kegel T, Große R, Eichner A. Recommendations for risk minimization when using Janus kinase inhibitors for the treatment of chronic inflammatory skin diseases. J Dtsch Dermatol Ges 2023; 21:845-851. [PMID: 37345890 DOI: 10.1111/ddg.15136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/25/2023] [Indexed: 06/23/2023]
Abstract
In accordance with article 20 of Regulation (EC) No 726/2004, the Pharmacovigilance Risk Assessment Committee (PRAC) of the European Medicines Agency (EMA) has re-evaluated the safety of Janus kinase inhibitors for the treatment of inflammatory diseases and formulated safety information deviating from the previous indications in the respective summary of product characteristics of the products concerned. These refer to the consideration of a possibly increased risk of venous thromboembolic or severe cardiovascular events, an increased infection rate and an increase in the prevalence of skin cancer across drugs and indications. Therefore, in patients with independent risk factors (age 65 years and older, smokers or former smokers, patients with oral contraception or hormone replacement therapy and other risk factors), it is recommended to use Janus kinase inhibitors therapeutically only if there are no suitable treatment alternatives. To facilitate a pragmatic and thorough detection of high-risk patients in everyday clinical practice, an interdisciplinary checklist was developed that is suitable as a working tool from the perspective of the dermatologist.
Collapse
Affiliation(s)
- Johannes Wohlrab
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Institute for Applied Dermatopharmaceutics, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Thomas Kegel
- Department of Internal Medicine IV (Hematology and Oncology), Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Regina Große
- Deparment of Gynecology, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Adina Eichner
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Institute for Applied Dermatopharmaceutics, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
12
|
Fletcher P, O'Donnell KL, Doratt BM, Malherbe DC, Clancy CS, Rhoderick JF, Feldmann F, Hanley PW, Ksiazek TG, Geisbert TW, Messaoudi I, Marzi A. Single-dose VSV-based vaccine protects cynomolgus macaques from disease after Taï Forest virus infection. Emerg Microbes Infect 2023:2239950. [PMID: 37470396 PMCID: PMC10392270 DOI: 10.1080/22221751.2023.2239950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Taï Forest virus (TAFV) is a lesser-known ebolavirus that causes lethal infections in chimpanzees and is responsible for a single human case. Limited research has been done on this human pathogen; however, with the recent emergence of filoviruses in West Africa, further investigation and countermeasure development against this virus is warranted.We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the TAFV glycoprotein as the viral antigen and assessed it for protective efficacy in nonhuman primates (NHPs). Following a single high-dose vaccination, NHPs developed antigen-specific binding and neutralizing antibodies as well as modest T cell responses. Importantly, all vaccinated NHPs were uniformly protected from disease after lethal TAFV challenge while the naïve control group succumbed to the disease. Histopathologic lesions consistent with filovirus disease were present in control NHPs but were not observed in vaccinated NHPs. Transcriptional analysis of whole blood samples obtained after vaccination and challenge was performed to gain insight into molecular underpinnings conferring protection. Differentially expressed genes (DEG) detected 7 days post-vaccination were enriched to processes associated with innate immunity and antiviral responses. Only a small number of DEG was detected in vaccinated NHPs post-challenge while over 1,000 DEG were detected in control NHPs at end-stage disease which mapped to gene ontology terms indicative of defense responses and inflammation. Taken together, this data demonstrates the effective single-dose protection of the VSV-TAFV vaccine, and its potential for use in outbreaks.
Collapse
Affiliation(s)
- Paige Fletcher
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kyle L O'Donnell
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Brianna M Doratt
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Delphine C Malherbe
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Chad S Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Joseph F Rhoderick
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Patrick W Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Thomas G Ksiazek
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
13
|
Laux J, Martorelli M, Späth N, Maier F, Burnet M, Laufer SA. Selective Inhibitors of Janus Kinase 3 Modify Responses to Lipopolysaccharides by Increasing the Interleukin-10-to-Tumor Necrosis Factor α Ratio. ACS Pharmacol Transl Sci 2023; 6:892-906. [PMID: 37325444 PMCID: PMC10262334 DOI: 10.1021/acsptsci.3c00043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Indexed: 06/17/2023]
Abstract
Janus kinase (JAK) inhibitors act at low doses (e.g., tofacitinib, 0.2-0.4 μmol/kg bid) in clinical use, suggesting an efficient underlying mode of action. We hypothesized that their effectiveness is due to their ability to raise the ratio of IL-10 to TNFα. Unlike other JAK isoforms, JAK3 is expressed mainly in hematopoietic cells and is essential for immune function. We used JAK3 selective inhibitors with preferential distribution to immune cells. Inhibition of JAK3 in human leukocytes reduced TNFα and IL-6 but maintained levels of IL-10, while pan-JAK inhibitors increased TNFα, IL-6, and IL-10. JAK1 is required for IL-10 receptor signaling, which suggests that, at exposure above the IC50 (55 nM for tofacitinib on JAK1), there is less feedback control of TNFα levels. This leads to self-limiting effects of JAK1 inhibitors and could place an upper limit on appropriate doses. In vivo, treating mice with JAK3 inhibitors before LPS administration decreased plasma TNFα and increased IL-10 above vehicle levels, suggesting that JAK3 inhibition may limit TNFα release by increasing IL-10 while leaving the IL-10 receptor functional. This mechanism should have general utility in controlling autoimmune diseases and can be conveniently observed by measuring the ratio of IL-10 to TNFα. In summary, our targeted, "leukotropic" inhibitors more effectively increased IL-10/TNFα ratios than unselective control compounds and could, therefore, be ideal for autoimmune therapy.
Collapse
Affiliation(s)
- Julian Laux
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
| | - Mariella Martorelli
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
| | - Nadja Späth
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Florian Maier
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Michael Burnet
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Stefan A. Laufer
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany
- Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| |
Collapse
|
14
|
Manuck TA, Eaves LA, Rager JE, Sheffield-abdullah K, Fry RC. Nitric oxide-related gene and microRNA expression in peripheral blood in pregnancy vary by self-reported race. Epigenetics 2022; 17:731-745. [PMID: 34308756 PMCID: PMC9336489 DOI: 10.1080/15592294.2021.1957576] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Adverse pregnancy outcomes disproportionately affect non-Hispanic (NH) Black patients in the United States. Structural racism has been associated with increased psychosocial distress and inflammation and may trigger oxidative stress. Thus, the nitric oxide (NO) pathway (involved in the regulation of inflammation and oxidative stress) may partly explain the underlying disparities in obstetric outcomes.Cohort study of 154 pregnant patients with high-risk obstetric histories; n = 212 mRNAs and n = 108 microRNAs (miRNAs) in the NO pathway were evaluated in circulating white blood cells. NO pathway mRNA and miRNA transcript counts were compared by self-reported race; NH Black patients were compared with women of other races/ethnicities. Finally, miRNA-mRNA expression levels were correlated.Twenty-two genes (q < 0.10) were differentially expressed in self-identified NH Black individuals. Superoxide dismutase 1 (SOD1), interleukin-8 (IL-8), dynein light chain LC8-type 1 (DYNLL1), glutathione peroxidase 4 (GPX4), and glutathione peroxidase 1 (GPX1) were the five most differentially expressed genes among NH Black patients compared to other patients. There were 63 significantly correlated miRNA-mRNA pairs (q < 0.10) demonstrating potential miRNA regulation of associated target mRNA expression. Ten miRNAs that were identified as members of significant miRNA-mRNA pairs were also differentially expressed among NH Black patients (q < 0.10).These findings support an association between NO pathway and inflammation and infection-related mRNA and miRNA expression in blood drawn during pregnancy and patient race/ethnicity. These findings may reflect key differences in the biology of inflammatory gene dysregulation that occurs in response to the stress of systemic racism and that underlies disparities in pregnancy outcomes.
Collapse
Affiliation(s)
- Tracy A. Manuck
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, United States
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Lauren A. Eaves
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Julia E Rager
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | | | - Rebecca C. Fry
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC
| |
Collapse
|
15
|
Abbasi B, Shamsasenjan K, Ahmadi M, Beheshti SA, Saleh M. Mesenchymal stem cells and natural killer cells interaction mechanisms and potential clinical applications. Stem Cell Res Ther 2022; 13:97. [PMID: 35255980 PMCID: PMC8900412 DOI: 10.1186/s13287-022-02777-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/09/2021] [Indexed: 12/29/2022] Open
Abstract
Natural killer cells (NK cells) are innate immune cells that are activated to fight tumor cells and virus-infected cells. NK cells also play an important role in the graft versus leukemia response. However, they can over-develop inflammatory reactions by secreting inflammatory cytokines and increasing Th1 differentiation, eventually leading to tissue damage. Today, researchers have attributed some autoimmune diseases and GVHD to NK cells. On the other hand, it has been shown that mesenchymal stem cells (MSCs) can modulate the activity of NK cells, while some researchers have shown that NK cells can cause MSCs to lysis. Therefore, we considered it is necessary to investigate the effect of these two cells and their signaling pathway in contact with each other, also their clinical applications.
Collapse
Affiliation(s)
- Batol Abbasi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedeh Ameneh Beheshti
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahshid Saleh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Chu X, Di C, Chang P, Li L, Feng Z, Xiao S, Yan X, Xu X, Li H, Qi R, Gong H, Zhao Y, Xiao F, Chang Z. Lactylated Histone H3K18 as a Potential Biomarker for the Diagnosis and Predicting the Severity of Septic Shock. Front Immunol 2022; 12:786666. [PMID: 35069560 PMCID: PMC8773995 DOI: 10.3389/fimmu.2021.786666] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/16/2021] [Indexed: 12/28/2022] Open
Abstract
Objective To date, there are no studies regarding the lactylation profile and its role in critically ill patients. Thus, we aimed to examine expression of histone H3 lysine 18 (H3K18) lactylation and its role in patients with septic shock. Methods Thirteen healthy volunteers and 35 critically ill patients from the Department of Surgical Intensive Care Medicine, Beijing Hospital were enrolled in our study. Baseline information and clinical outcomes were obtained prospectively. Lactylation levels of all proteins and H3K18 from peripheral blood mononuclear (PBMC) were determined by western blotting and serum levels of inflammatory cytokines by flow cytometry. Arginase-1 (Arg1) and Krüppel-like factor-4 (Klf4) mRNA expression was evaluated by quantitative real-time PCR (qRT-PCR). Results Lactylation was found to be an all-protein post-translational modification and was detected in PBMCs from both healthy volunteers and critically ill patients, with a significantly higher relative density in shock patients (t=2.172, P=0.045). H3K18la was expressed in all subjects, including healthy volunteers, with the highest level in septic shock patients (compared with non-septic shock patients, critically ill without shock patients and healthy volunteers P=0.033, 0.000 and 0.000, respectively). Furthermore, H3K18la protein expression correlated positively with APACHE II scores, SOFA scores on day 1, ICU stay, mechanical ventilation time and serum lactate (ρ=0.42, 0.63, 0.39, 0.51 and 0.48, respectively, ρ=0.012, 0.000, 0.019, 0.003 and 0.003, respectively). When we matched patients with septic shock and with non-septic shock according to severity, we found higher H3K18la levels in the former group (t=-2.208, P =0.040). Moreover, H3K18la exhibited a close correlation with procalcitonin levels (ρ=0.71, P=0.010). Patients with high H3K18la expression showed higher IL-2, IL-5, IL-6, IL-8, IL-10, IL-17, IFN-α levels (ρ=0.33, 0.37, 0.62, 0.55, 0.65, 0.49 and 0.374 respectively, P=0.024, 0.011, 0.000, 0.000, 0.000 and 0.000 respectively). H3K18la expression also displayed a positive correlation with the level of Arg1 mRNA (ρ=0.561, P=0.005). Conclusions Lactylation is an all-protein post-translational modification occurring in both healthy subjects and critically ill patients. H3K18la may reflect the severity of critical illness and the presence of infection. H3K18la might mediate inflammatory cytokine expression and Arg1 overexpression and stimulate the anti-inflammatory function of macrophages in sepsis.
Collapse
Affiliation(s)
- Xin Chu
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Chenyi Di
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Panpan Chang
- Trauma Center, Department of Orthopaedics and Traumatology, Peking University People's Hospital, Beijing, China
| | - Lina Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhe Feng
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shirou Xiao
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyu Yan
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaodong Xu
- Department of Haematology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hexin Li
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruomei Qi
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Huan Gong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Yanyang Zhao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Fei Xiao
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Chang
- Department of Surgical Intensive Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Jin K, Parreau S, Warrington KJ, Koster MJ, Berry GJ, Goronzy JJ, Weyand CM. Regulatory T Cells in Autoimmune Vasculitis. Front Immunol 2022; 13:844300. [PMID: 35296082 PMCID: PMC8918523 DOI: 10.3389/fimmu.2022.844300] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
Blood vessels are indispensable for host survival and are protected from inappropriate inflammation by immune privilege. This protection is lost in patients with autoimmune vasculitides, a heterogeneous group of diseases causing damage to arteries, arterioles, and capillaries. Vasculitis leads to vascular wall destruction and/or luminal occlusion, resulting in hemorrhage and tissue ischemia. Failure in the quantity and quality of immunosuppressive regulatory T cells (Treg) has been implicated in the breakdown of the vascular immune privilege. Emerging data suggest that Treg deficiencies are disease-specific, affecting distinct pathways in distinct vasculitides. Mechanistic studies have identified faulty CD8+ Tregs in Giant Cell Arteritis (GCA), a vasculitis of the aorta and the large aortic branch vessels. Specifically, aberrant signaling through the NOTCH4 receptor expressed on CD8+ Treg cells leads to rerouting of intracellular vesicle trafficking and failure in the release of immunosuppressive exosomes, ultimately boosting inflammatory attack to medium and large arteries. In Kawasaki’s disease, a medium vessel vasculitis targeting the coronary arteries, aberrant expression of miR-155 and dysregulated STAT5 signaling have been implicated in undermining CD4+ Treg function. Explorations of mechanisms leading to insufficient immunosuppression and uncontrolled vascular inflammation hold the promise to discover novel therapeutic interventions that could potentially restore the immune privilege of blood vessels and pave the way for urgently needed innovations in vasculitis management.
Collapse
Affiliation(s)
- Ke Jin
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Simon Parreau
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Kenneth J. Warrington
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Matthew J. Koster
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
| | - Gerald J. Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Jörg J. Goronzy
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cornelia M. Weyand
- Department of Medicine, Mayo College of Medicine and Science, Rochester, MN, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
- *Correspondence: Cornelia M. Weyand,
| |
Collapse
|
18
|
Zhou Q, Ren Q, Jiao L, Huang J, Yi J, Chen J, Lai J, Ji G, Zheng T. The potential roles of JAK/STAT signaling in the progression of osteoarthritis. Front Endocrinol (Lausanne) 2022; 13:1069057. [PMID: 36506076 PMCID: PMC9729341 DOI: 10.3389/fendo.2022.1069057] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
Osteoarthritis (OA) is an age-related chronic progressive degenerative disease that induces persistent pain and disabilities. The development of OA is a complex process, and the risk factors are various, including aging, genetics, trauma and altered biomechanics. Inflammation and immunity play an important role in the pathogenesis of OA. JAK/STAT pathway is one of the most prominent intracellular signaling pathways, regulating cell proliferation, differentiation, and apoptosis. Inflammatory factors can act as the initiators of JAK/STAT pathway, which is implicated in the pathophysiological activity of chondrocyte. In this article, we provide a review on the importance of JAK/STAT pathway in the pathological development of OA. Potentially, JAK/STAT pathway becomes a therapeutic target for managing OA.
Collapse
Affiliation(s)
- Qingluo Zhou
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qun Ren
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Linhui Jiao
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Jishang Huang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jun Yi
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jincai Chen
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jinliang Lai
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Guanglin Ji
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Guanglin Ji, ; Tiansheng Zheng,
| | - Tiansheng Zheng
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Guanglin Ji, ; Tiansheng Zheng,
| |
Collapse
|
19
|
Interactions of Muscovy duck reovirus, gut microbiota, and host innate immunity: Transcriptome and gut microbiota analysis. Vet Microbiol 2021; 264:109286. [PMID: 34856425 DOI: 10.1016/j.vetmic.2021.109286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 11/20/2022]
Abstract
It has been shown that Muscovy duck reovirus (MDRV) infection causes severe intestinal barrier damage and intestinal mucosal immune suppression. The health and balance of gut microbes is essential for the progression of intestinal infectious diseases. To investigate the interaction of MDRV, intestinal bacteria with host intestinal innate immunity, an MDRV contact-infection model was established in this study. High-throughput sequencing technology was used to sequence 16S rDNA and transcripts in ileal samples from experimental Muscovy ducklings. Our results suggest that intestinal opportunistic pathogens such as Streptococcus and Corynebacterium proliferated massively in MDRV-infected Muscovy ducklings. The body initiates antiviral and antibacterial immunity and actively fights the infection of gut microbes. The synthesis of peptidoglycan, lipopolysaccharide, and flagellin by intestinal bacteria activates the Toll-like receptor signaling pathway resulting in increased secretion of IFN-β, IL-1β, and IL-8. The RIG-I-like receptor signaling pathway is an important signaling pathway for the interaction between MDRV and the host. At the same time, we also observed that multiple genes in the JAK-STAT signaling pathway were significantly different. These genes are important targets for studying the immunosuppression caused by MDRV. In conclusion, we analyzed the interaction of MDRV, intestinal flora and host immune system during MDRV infection, which provides a basis for the further study on the mechanism of intestinal immunosuppression caused by MDRV.
Collapse
|
20
|
Wang J, Du L, Tang H. Suppression of Interferon-α Treatment Response by Host Negative Factors in Hepatitis B Virus Infection. Front Med (Lausanne) 2021; 8:784172. [PMID: 34901094 PMCID: PMC8651562 DOI: 10.3389/fmed.2021.784172] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/03/2021] [Indexed: 02/05/2023] Open
Abstract
Chronic hepatitis B virus (CHB) infection remains a major global public health issue for which there is still lacking effective curative treatment. Interferon-α (IFN-α) and its pegylated form have been approved as an anti-HBV drug with the advantage of antiviral activity and host immunity against HBV infection enhancement, however, IFN-α treatment failure in CHB patients is a challenging obstacle with 70% of CHB patients respond poorly to exogenous IFN-α treatment. The IFN-α treatment response is negatively regulated by both viral and host factors, and the role of viral factors has been extensively illustrated, while much less attention has been paid to host negative factors. Here, we summarized evidence of host negative regulators and parameters involved in IFN-α therapy failure, review the mechanisms responsible for these effects, and discuss the possible improvement of IFN-based therapy and the rationale of combining the inhibitors of negative regulators in achieving an HBV cure.
Collapse
Affiliation(s)
- Jiayi Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Icariin improves cutaneous wound healing in streptozotocin-induced diabetic rats. J Tissue Viability 2021; 31:197-206. [PMID: 34565677 DOI: 10.1016/j.jtv.2021.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 12/27/2022]
Abstract
In diabetes, wound healing gets delayed due to various factors. Icariin, a flavonoid obtained from the plants of the Epimedium genus, exhibited anti-inflammatory, angiogenic, and matrix metalloproteinase-inhibiting effects to heal skin wounds in non-diabetic rats on topical application. Hence, we designed the present study to explore icariin's potential to heal cutaneous diabetic wounds in rats. Diabetes was induced by streptozotocin in male Wistar rats and they were divided into two groups after creating a skin wound of approximately 4 cm2 area. Simple ointment base and 0.04% icariin ointment were applied twice daily for 19 days in the control and the treatment group, respectively. The healing of the wound was assessed based on wound closure, the expression patterns of NF-κB, TNF-α, IL-10, CD31, MMP-2 and -9 activities, and collagen deposition on predetermined days after wound creation. Wounds treated with icariin showed a marked increase in per cent wound closure on different post-wounding days than diabetic control. Upregulation of IL-10 and decreased expressions of NF-κB and TNF-α were revealed in western blots, indicating an anti-inflammatory effect of icariin. Western blot, as well as immunohistochemistry, showed increased expression of CD31 on all days confirming the angiogenic effect of icariin in healing. Icariin treatment reduced MMP-2 and -9 activities and increased deposition of well-organized collagen. Results demonstrate that icariin reduced inflammation and improved angiogenesis and thus, it appears to possess the potential to enhance the healing of diabetic wounds.
Collapse
|
22
|
Dowarah J, Marak BN, Yadav UCS, Singh VP. Potential drug development and therapeutic approaches for clinical intervention in COVID-19. Bioorg Chem 2021; 114:105016. [PMID: 34144277 PMCID: PMC8143914 DOI: 10.1016/j.bioorg.2021.105016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/20/2021] [Indexed: 01/25/2023]
Abstract
While the vaccination is now available to many countries and will slowly dissipate to others, effective therapeutics for COVID-19 is still illusive. The SARS-CoV-2 pandemic has posed an unprecedented challenge to researchers, scientists, and clinicians and affected the wellbeing of millions of people worldwide. Since the beginning of the pandemic, a multitude of existing anti-viral, antibiotic, antimalarial, and anticancer drugs have been tested, and some have shown potency in the treatment and management of COVID-19, albeit others failed to leave any positive impact and a few also became controversial as they showed mixed clinical outcomes. In the present article, we have brought together some of the candidate therapeutic drugs being repurposed or used in the clinical trials and discussed their clinical efficacy and safety for COVID-19.
Collapse
Affiliation(s)
- Jayanta Dowarah
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | - Brilliant N Marak
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | | | - Ved Prakash Singh
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India; Department of Industrial Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India.
| |
Collapse
|
23
|
Xiong D, Luo S, Wu K, Yu Y, Sun J, Wang Y, Hu J, Hu W. Transcriptional profiling of Microtus fortis responses to S. japonicum: New sight into Mf-Hsp90α resistance mechanism. Parasite Immunol 2021; 43:e12842. [PMID: 33959966 PMCID: PMC8365665 DOI: 10.1111/pim.12842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 12/22/2022]
Abstract
AIMS Schistosomiasis is a parasitic disease with a chronic debilitating character caused by parasitic flatworms of the genus Schistosoma. The main disease-causing species of Schistosoma in China is S. japonicum. M fortis has been proved to be a nonpermissive host of S. japonicum. Mf-HSP90α (Microtus fortis heat shock protein 90alpha), the homologue of HSP90α, display anti-schistosome effect in vitro and in vivo. In the current study, in order to investigate the mechanism of anti-schistosome effect of Mf-HSP90α, we conducted RNA-Seq to obtain the transcriptome profile of M. fortis liver infected with S. japonicum at different time points. METHODS AND RESULTS By mapping the differential expressed genes (DEGs) to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), we found that the JAK2/STAT1 pathway was highly enriched with an elevated level of IL-10 and HSP90α. We then checked the IL-10-JAK2/STAT1-HSP90α pathway, and found that this pathway was activated in the infected mice with S. japonicum. The expression of the molecules in this pathway was elevated on the 10th day after infection and gradually decreased on the 20th day. CONCLUSIONS The IL-10-JAK2/STAT1-HSP90α axis was associated with the anti-schistosome effect of Mf-HSP90α, and targeting IL-10-JAK2/STAT1-HSP90α axis might be a novel therapeutic strategy for developing resistance to S. japonicum infection.
Collapse
Affiliation(s)
- Dehui Xiong
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Saiqun Luo
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Kunlu Wu
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Yuanjing Yu
- Department of Laboratory Animal, Central South University, Changsha, China
| | - Jiameng Sun
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Yanpeng Wang
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Jingping Hu
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Weixin Hu
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| |
Collapse
|
24
|
Manuck TA, Eaves LA, Rager JE, Fry RC. Mid-pregnancy maternal blood nitric oxide-related gene and miRNA expression are associated with preterm birth. Epigenomics 2021; 13:667-682. [PMID: 33890487 DOI: 10.2217/epi-2020-0346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The nitric oxide (NO) pathway modulates inflammation and may influence birth timing. Patients & methods: Case-control analysis of 136 pregnant women with RNA obtained <28 weeks; n = 212 mRNAs and n = 108 miRNAs in the NO pathway were evaluated. NO-pathway mRNA and miRNA transcript counts in women delivering preterm versus at term were compared, miRNA-mRNA expression levels correlated and prediction models generated. Results: Fourteen genes were differentially expressed in women delivering <37 weeks; 13/14 were also differentially expressed in those delivering <34 weeks (q <0.10) versus term births. Multiple miRNA-mRNA pairs were correlated. Models with gene expression better predicted prematurity than models with only clinical or nongenomic predictors. Conclusion: Maternal blood NO pathway-related mRNA and miRNA expression is associated with prematurity.
Collapse
Affiliation(s)
- Tracy A Manuck
- Department of Obstetrics & Gynecology, Division of Maternal Fetal Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.,Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lauren A Eaves
- Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Julia E Rager
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rebecca C Fry
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Environmental Sciences & Engineering, Gillings School of Global Public Health, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
25
|
Zhou S, Qi F, Gong Y, Zhang J, Zhu B. Biological Therapies for Atopic Dermatitis: A Systematic Review. Dermatology 2021; 237:542-552. [PMID: 33735876 DOI: 10.1159/000514535] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/17/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a widely acquired, relapsing inflammatory skin disease. Biologics are now widely used in patients with moderate-to-severe AD. OBJECTIVE This work aims to summarize both label and off-label biologics on AD treatment in phase II and phase III stages, and compile evidence on the efficacy of the most-studied biologics. METHODS We conducted a comprehensive literature search through PubMed, EMBASE, and ClinicalTrials.gov to identify all documented biological therapies for AD. The criteria were further refined to focus on those treatments with the highest evidence level for AD with at least one randomized clinical trial supporting their use. Only studies or articles published in English were enrolled in this study. FINDINGS Primary searches identified 525 relevant articles and 27 trials. Duplicated articles and papers without a full text were excluded. Only completed trials were enrolled. We included 28 randomized controlled trials, 4 unpublished trials, 2 observational studies, and 1 meta-analysis. Eight kinds of biologics, including IL-4/IL-13 inhibitors, JAK inhibitors, anti-IL-13 antibodies, anti-IL-22 antibodies, anti-IL-33 antibodies, thymic stromal lymphopoietin inhibitor (TSLP), OX40 antibodies, and H4R-antagonists were included in this work. Dupliumab, as the most widely used and investigated biologic, was reported in 1 meta-analysis and 4 trials exploring its long-term use and application in both adults and pediatric patients. Besides dupilumab, four other IL-4/IL-13 inhibitors recruited were all randomized, clinical trials at phase 2-3 stage. Six different kinds of JAK inhibitors were summarized with strong evidence revealing their significant therapeutic effects on AD. There were 3 trials for nemolizumab, an anti-IL-13 antibody, all of which were in the phase 2 clinical trial stage. Results showed nemolizumab could be another alternative therapy for moderate-to-severe AD with long-term efficiency and safety. CONCLUSION The biological therapies with the most robust evidence on efficacy and long-term safety for AD treatment include dupilumab, barcitinib, abrocitinib, and delgocitinib. Most of the biologics mentioned in this review were still at the exploratory stage. This review will help practitioners advise patients seeking suitable biological therapies and offer experimental study directions for treatment.
Collapse
Affiliation(s)
- Shuying Zhou
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| | - Fei Qi
- Capital Medical University Affiliated with Beijing Chaoyang Hospital, Beijing, China,
| | - Yue Gong
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| | - Jinping Zhang
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| | - Binghua Zhu
- Department of Dermatology, The 305 Hospital of PLA, Beijing, China
| |
Collapse
|
26
|
García-Baos A, Alegre-Zurano L, Cantacorps L, Martín-Sánchez A, Valverde O. Role of cannabinoids in alcohol-induced neuroinflammation. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110054. [PMID: 32758518 DOI: 10.1016/j.pnpbp.2020.110054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Alcohol is a psychoactive substance highly used worldwide, whose harmful use might cause a broad range of mental and behavioural disorders. Underlying brain impact, the neuroinflammatory response induced by alcohol is recognised as a key contributing factor in the progression of other neuropathological processes, such as neurodegeneration. These sequels are determined by multiple factors, including age of exposure. Strikingly, it seems that the endocannabinoid system modulation could regulate the alcohol-induced neuroinflammation. Although direct CB1 activation can worsen alcohol consequences, targeting other components of the expanded endocannabinoid system may counterbalance the pro-inflammatory response. Indeed, specific modulations of the expanded endocannabinoid system have been proved to exert anti-inflammatory effects, primarily through the CB2 and PPARγ signalling. Among them, some endo- and exogeneous cannabinoids can block certain pro-inflammatory mediators, such as NF-κB, thereby neutralizing the neuroinflammatory intracellular cascades. Furthermore, a number of cannabinoids are able to activate complementary anti-inflammatory pathways, which are necessary for the transition from chronically overactivated microglia to a regenerative microglial phenotype. Thus, cannabinoid modulation provides cooperative anti-inflammatory mechanisms that may be advantageous to resolve a pathological neuroinflammation in an alcohol-dependent context.
Collapse
Affiliation(s)
- Alba García-Baos
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laia Alegre-Zurano
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lídia Cantacorps
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Martín-Sánchez
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Programme, IMIM-Hospital del Mar Research Institute, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Neuroscience Research Programme, IMIM-Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
27
|
Borcherding N, Jethava Y, Vikas P. Repurposing Anti-Cancer Drugs for COVID-19 Treatment. Drug Des Devel Ther 2020; 14:5045-5058. [PMID: 33239864 PMCID: PMC7680713 DOI: 10.2147/dddt.s282252] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic has caused catastrophic damage to human life across the globe along with social and financial hardships. According to the Johns Hopkins University Coronavirus Resource Center, more than 41.3 million people worldwide have been infected, and more than 1,133,000 people have died as of October 22, 2020. At present, there is no available vaccine and a scarcity of efficacious therapies. However, there is tremendous ongoing effort towards identifying effective drugs and developing novel vaccines. Early data from Adaptive COVID-19 Treatment Trials (ACTT) sponsored by the National Institute of Allergy and Infectious Diseases (NIAID) and compassionate use study have shown promise for remdesivir, leading to emergency authorization by the Food and Drug Administration (FDA) for treatment of hospitalized COVID-19 patients. However, several randomized studies have now shown no benefit or increased adverse events associated with remdesivir treatment. Drug development is a time-intensive process and requires extensive safety and efficacy evaluations. In contrast, drug repurposing is a time-saving and cost-effective drug discovery strategy geared towards using existing drugs instead of de novo drug discovery. Treatments for cancer and COVID-19 often have similar goals of controlling inflammation, inhibiting cell division, and modulating the host microenvironment to control the disease. In this review, we focus on anti-cancer drugs that can potentially be repurposed for COVID-19 and are currently being tested in clinical trials.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA, USA
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA, USA
| | - Yogesh Jethava
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, IA, USA
| | - Praveen Vikas
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, IA, USA
| |
Collapse
|
28
|
Lee YM, Shin DW, Lim BO. Chlorogenic Acid Improves Symptoms of Inflammatory Bowel Disease in Interleukin-10 Knockout Mice. J Med Food 2020; 23:1043-1053. [PMID: 33054539 DOI: 10.1089/jmf.2019.4621] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammation that is highly prevalent worldwide. Interleukin (IL)-10 can effectively inhibit negative cascades such as the production of inflammatory mediators (inducible nitric oxide synthase [iNOS], cyclooxygenase-2), accumulation of inflammatory infiltrates (macrophages, eosinophils, neutrophils), toxicity (lower T cell subsets), and secretion of pro-inflammatory cytokines (IL-1β, TNF-α) in tissues such as the spleen, mesenteric lymph nodes (MLN), Peyer's patch (PP), and colon. In this study, we investigated whether chlorogenic acid (CHA) can regulate inflammation in IL-10 knockout (KO) mice used as an IBD animal model. CHA significantly increased the ratio of CD4+/CD8+, T cell subsets in PP, and MLN of IL-10 KO mice. In addition, CHA also morphologically attenuated colon inflammation in IL-10 KO mice. We demonstrated that CHA significantly reduced the expression levels of iNOS, IL-1β, TNF-α, which were highly expressed in IL-10 KO mice. Therefore, CHA may provide beneficial effects for improving IBD by decreasing inflammations.
Collapse
Affiliation(s)
- Young Min Lee
- College of Biomedical and Health Science, Konkuk University, Chungju, Korea
| | - Dong Wook Shin
- College of Biomedical and Health Science, Konkuk University, Chungju, Korea
| | - Beong Ou Lim
- College of Biomedical and Health Science, Konkuk University, Chungju, Korea.,Research Institute of Inflammatory Diseases, Konkuk University, Chungju, Korea
| |
Collapse
|
29
|
Huang C, Zhu J, Wang L, Chu A, Yin Y, Vali K, Garmendia A, Tang Y. Cryptotanshinone protects porcine alveolar macrophages from infection with porcine reproductive and respiratory syndrome virus. Antiviral Res 2020; 183:104937. [PMID: 32961199 DOI: 10.1016/j.antiviral.2020.104937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/31/2020] [Accepted: 09/17/2020] [Indexed: 10/23/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS), caused by PRRS virus (PRRSV) infection, imposes enormous economic impact to the world pork industry. Currently there is no effective treatment to prevent PRRSV infection in swine. We report that the natural compound cryptotanshinone (Cpt) effectively inhibits the infection of various strains of PRRSV to porcine alveolar macrophages (PAMs), the primary cell target of PRRSV in vivo. Mechanistically, Cpt inhibits the activation of signal transducer and activator of transcription 3 (STAT3), and blocks the interleukin 10 (IL-10) stimulated as well as the basal level CD163 expression in PAMs. Cpt-treatment of PAMs is effective when applied either before or after PRRSV infection, with the combined pre- and post-PRRSV infection treatment resulting in the most significant, dose-dependent inhibition of PRRSV infection. Cpt inhibited both type I/II PRRSV infection in PAMs. Our study identified a new approach to prevent/treat PRRSV infection of pigs with natural compounds.
Collapse
Affiliation(s)
- Chang Huang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA
| | - Jiaqi Zhu
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA
| | - Ling Wang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA
| | - Alexander Chu
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA
| | - Yexuan Yin
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA
| | - Kaneha Vali
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA
| | - Antonio Garmendia
- Department of Pathobiology and Veterinary Sciences, University of Connecticut, 61 North Eagleville Road, Storrs, CT, 06269, USA.
| | - Young Tang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, 1390 Storrs Rd, Storrs, CT, 06269, USA.
| |
Collapse
|
30
|
Duncan SA, Sahu R, Dixit S, Singh SR, Dennis VA. Suppressors of Cytokine Signaling (SOCS)1 and SOCS3 Proteins Are Mediators of Interleukin-10 Modulation of Inflammatory Responses Induced by Chlamydia muridarum and Its Major Outer Membrane Protein (MOMP) in Mouse J774 Macrophages. Mediators Inflamm 2020; 2020:7461742. [PMID: 32684836 PMCID: PMC7333066 DOI: 10.1155/2020/7461742] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022] Open
Abstract
The immunopathology of chlamydial diseases is exacerbated by a broad-spectrum of inflammatory mediators, which we reported are inhibited by IL-10 in macrophages. However, the chlamydial protein moiety that induces the inflammatory mediators and the mechanisms by which IL-10 inhibits them are unknown. We hypothesized that Chlamydia major outer membrane protein (MOMP) mediates its disease pathogenesis, and the suppressor of cytokine signaling (SOCS)1 and SOCS3 proteins are mediators of the IL-10 inhibitory actions. Our hypothesis was tested by exposing mouse J774 macrophages to chlamydial stimulants (live Chlamydia muridarum and MOMP) with and without IL-10. MOMP significantly induced several inflammatory mediators (IL-6, IL-12p40, CCL5, CXCL10), which were dose-dependently inhibited by IL-10. Chlamydial stimulants induced the mRNA gene transcripts and protein expression of SOCS1 and SOCS3, with more SOCS3 expression. Notably, IL-10 reciprocally regulated their expression by reducing SOCS1 and increasing SOCS3. Specific inhibitions of MAPK pathways revealed that p38, JNK, and MEK1/2 are required for inducing inflammatory mediators as well as SOCS1 and SOCS3. Chlamydial stimulants triggered an M1 pro-inflammatory phenotype evidently by an enhanced nos2 (M1 marker) expression, which was skewed by IL-10 towards a more M2 anti-inflammatory phenotype by the increased expression of mrc1 and arg1 (M2 markers) and the reduced SOCS1/SOCS3 ratios. Neutralization of endogenously produced IL-10 augmented the secretion of inflammatory mediators, reduced SOCS3 expression, and skewed the chlamydial M1 to an M2 phenotype. Inhibition of proteasome degradation increased TNF but decreased IL-10, CCL5, and CXCL10 secretion by suppressing SOCS1 and SOCS3 expressions and dysregulating their STAT1 and STAT3 transcription factors. Our data show that SOCS1 and SOCS3 are regulators of IL-10 inhibitory actions, and underscore SOCS proteins as therapeutic targets for IL-10 control of inflammation for Chlamydia and other bacterial inflammatory diseases.
Collapse
Affiliation(s)
- Skyla A. Duncan
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Rajnish Sahu
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Saurabh Dixit
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Shree R. Singh
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Vida A. Dennis
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| |
Collapse
|
31
|
Barone M, Catani L, Ricci F, Romano M, Forte D, Auteri G, Bartoletti D, Ottaviani E, Tazzari PL, Vianelli N, Cavo M, Palandri F. The role of circulating monocytes and JAK inhibition in the infectious-driven inflammatory response of myelofibrosis. Oncoimmunology 2020; 9:1782575. [PMID: 32923146 PMCID: PMC7458658 DOI: 10.1080/2162402x.2020.1782575] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myelofibrosis (MF) is characterized by chronic inflammation and hyper-activation of the JAK-STAT pathway. Infections are one of the main causes of morbidity/mortality. Therapy with Ruxolitinib (RUX), a JAK1/2 inhibitor, may further increase the infectious risk. Monocytes are critical players in inflammation/immunity through cytokine production and release of bioactive extracellular vesicles. However, the functional behavior of MF monocytes, particularly during RUX therapy, is still unclear. In this study, we found that monocytes from JAK2V617F-mutated MF patients show an altered expression of chemokine (CCR2, CXCR3, CCR5) and cytokine (TNF-α-R, IL10-R, IL1β-R, IL6-R) receptors. Furthermore, their ability to produce and secrete free and extracellular vesicles-linked cytokines (IL1β, TNF-α, IL6, IL10) under lipopolysaccharides (LPS) stimulation is severely impaired. Interestingly, monocytes from RUX-treated patients show normal level of chemokine, IL10, IL1β, and IL6 receptors together with a restored ability to produce intracellular and to secrete extracellular vesicles-linked cytokines after LPS stimulation. Conversely, RUX therapy does not normalize TNF-R1/2 receptors expression and the LPS-driven secretion of free pro/anti-inflammatory cytokines. Accordingly, upon LPS stimulation, in vitro RUX treatment of monocytes from MF patients increases their secretion of extracellular vesicles-linked cytokines but inhibits the secretion of free pro/anti-inflammatory cytokines. In conclusion, we demonstrated that in MF the infection-driven response of circulating monocytes is defective. Importantly, RUX promotes their infection-driven cytokine production suggesting that infections following RUX therapy may not be due to monocyte failure. These findings contribute to better interpreting the immune vulnerability of MF and to envisaging strategies to improve the infection-driven immune response.
Collapse
Affiliation(s)
- Martina Barone
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lucia Catani
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ricci
- Immunohematology and Blood Bank, Azienda Ospedaliero-Universitaria S. Orsola-Malpighi di Bologna, Bologna, Italy
| | - Marco Romano
- School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Dorian Forte
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giuseppe Auteri
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Daniela Bartoletti
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Emanuela Ottaviani
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pier Luigi Tazzari
- Immunohematology and Blood Bank, Azienda Ospedaliero-Universitaria S. Orsola-Malpighi di Bologna, Bologna, Italy
| | - Nicola Vianelli
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michele Cavo
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Palandri
- Department of Experimental, Institute of Hematology "L. E A. "Seràgnoli", Diagnostic and Specialty Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
32
|
Khordadmehr M, Ghaderi S, Mesgari-Abbasi M, Jigari-Asl F, Nofouzi K, Tayefi-Nasrabadi H, McIntyre G. The Beneficial Effects of Actinomycetales Immune Modulators in the Pancreas of Diabetic Rats. Adv Pharm Bull 2020; 11:371-377. [PMID: 33880360 PMCID: PMC8046390 DOI: 10.34172/apb.2021.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 11/09/2022] Open
Abstract
Purpose: Type 1 diabetes mellitus (T1DM) has dramatically increased in recent years, especially in young people, and limits the life quality of the patients involved. Thus, many researchers are performing extensive studies to find alternative treatments for DM. Methods: Here, we evaluated the improvement effects of the heat-killed Actinomycetales species, including Gordonia bronchialis, and Tsukamurella inchonensis in streptozotocin (STZ)- diabetic rats by biochemical, immunological, and histopathological examinations. Results: The present findings exhibited a dramatic and progressive alteration in the serum levels of interleukin-6 (IL-6), IL-10 and tumor necrosis factor-α (TNF-α) in the diabetic group, which were related to the blood glucose and insulin levels, oxidative stress defense (evaluated by TAC and MDA activities), and the pancreas biochemical indicators (such as amylase and lipase). More importantly, the present results were consistent with the histopathological findings, which included cellular degeneration, vascular congestion, hemorrhage, focal necrosis associated with mononuclear cell infiltration. Interestingly, all of the diabetic changes in the blood serum and tissues improved remarkably in the treated groups by Actinomycetales species. Conclusion: Surprisingly, most of the current diabetic complications effectively attenuated after oral administration of both Actinomycetales species, particularly with a high dose of T. inchonensis. Thus, it is concluded that the heat-killed Actinomycetales species can prevent and improve the progression of T1DM and its various complications profoundly.
Collapse
Affiliation(s)
- Monireh Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Solin Ghaderi
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | | | - Farinaz Jigari-Asl
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Katayoon Nofouzi
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | | | - Graham McIntyre
- Center for Infectious Diseases and International Health, Windeyer Institute for Medical Sciences, University College London, UK
| |
Collapse
|
33
|
Rapid Bladder Interleukin-10 Synthesis in Response to Uropathogenic Escherichia coli Is Part of a Defense Strategy Triggered by the Major Bacterial Flagellar Filament FliC and Contingent on TLR5. mSphere 2019; 4:4/6/e00545-19. [PMID: 31776239 PMCID: PMC6881718 DOI: 10.1128/msphere.00545-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interleukin-10 is part of the immune response to urinary tract infection (UTI) due to E. coli, and it is important in the early control of infection in the bladder. Defining the mechanism of engagement of the immune system by the bacteria that enables the protective IL-10 response is critical to exploring how we might exploit this mechanism for new infection control strategies. In this study, we reveal part of the bacterial flagellar apparatus (FliC) is an important component that is sensed by and responsible for induction of IL-10 in the response to UPEC. We show this response occurs in a TLR5-dependent manner. Using infection prevention and control trials in mice infected with E. coli, this study also provides evidence that purified FliC might be of value in novel approaches for the treatment of UTI or in preventing infection by exploiting the FliC-triggered bladder transcriptome. Urinary tract infection (UTI) caused by uropathogenic Escherichia coli (UPEC) engages interleukin-10 (IL-10) as an early innate immune response to regulate inflammation and promote the control of bladder infection. However, the mechanism of engagement of innate immunity by UPEC that leads to elicitation of IL-10 in the bladder is unknown. Here, we identify the major UPEC flagellar filament, FliC, as a key bacterial component sensed by the bladder innate immune system responsible for the induction of IL-10 synthesis. IL-10 responses of human as well as mouse bladder epithelial cell-monocyte cocultures were triggered by flagella of three major UPEC representative strains, CFT073, UTI89, and EC958. FliC purified to homogeneity induced IL-10 in vitro and in vivo as well as other functionally related cytokines, including IL-6. The genome-wide innate immunological context of FliC-induced IL-10 in the bladder was defined using RNA sequencing that revealed a network of transcriptional and antibacterial defenses comprising 1,400 genes that were induced by FliC. Of the FliC-responsive bladder transcriptome, altered expression of il10 and 808 additional genes were dependent on Toll-like receptor 5 (TLR5), according to analysis of TLR5-deficient mice. Examination of the potential of FliC and associated innate immune signature in the bladder to boost host defense, based on prophylactic or therapeutic administration to mice, revealed significant benefits for the control of UPEC. We conclude that detection of FliC through TLR5 triggers rapid IL-10 synthesis in the bladder, and FliC represents a potential immune modulator that might offer benefit for the treatment or prevention of UPEC UTI. IMPORTANCE Interleukin-10 is part of the immune response to urinary tract infection (UTI) due to E. coli, and it is important in the early control of infection in the bladder. Defining the mechanism of engagement of the immune system by the bacteria that enables the protective IL-10 response is critical to exploring how we might exploit this mechanism for new infection control strategies. In this study, we reveal part of the bacterial flagellar apparatus (FliC) is an important component that is sensed by and responsible for induction of IL-10 in the response to UPEC. We show this response occurs in a TLR5-dependent manner. Using infection prevention and control trials in mice infected with E. coli, this study also provides evidence that purified FliC might be of value in novel approaches for the treatment of UTI or in preventing infection by exploiting the FliC-triggered bladder transcriptome.
Collapse
|
34
|
Cytokine Effects on the Entry of Filovirus Envelope Pseudotyped Virus-Like Particles into Primary Human Macrophages. Viruses 2019; 11:v11100889. [PMID: 31547585 PMCID: PMC6832363 DOI: 10.3390/v11100889] [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: 07/17/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 12/25/2022] Open
Abstract
Macrophages are one of the first and also a major site of filovirus replication and, in addition, are a source of multiple cytokines, presumed to play a critical role in the pathogenesis of the viral infection. Some of these cytokines are known to induce macrophage phenotypic changes in vitro, but how macrophage polarization may affect the cell susceptibility to filovirus entry remains largely unstudied. We generated different macrophage subsets using cytokine pre-treatment and subsequently tested their ability to fuse with beta-lactamase containing virus-like particles (VLP), pseudotyped with the surface glycoprotein of Ebola virus (EBOV) or the glycoproteins of other clinically relevant filovirus species. We found that pre-incubation of primary human monocyte-derived macrophages (MDM) with interleukin-10 (IL-10) significantly enhanced filovirus entry into cells obtained from multiple healthy donors, and the IL-10 effect was preserved in the presence of pro-inflammatory cytokines found to be elevated during EBOV disease. In contrast, fusion of IL-10-treated macrophages with influenza hemagglutinin/neuraminidase pseudotyped VLPs was unchanged or slightly reduced. Importantly, our in vitro data showing enhanced virus entry are consistent with the correlation established between elevated serum IL-10 and increased mortality in filovirus infected patients and also reveal a novel mechanism that may account for the IL-10-mediated increase in filovirus pathogenicity.
Collapse
|
35
|
Bare Y, Marhendra APW, Sasase T, Fatchiyah F. Differential Expression of IL-10 Gene and Protein in Target Tissues of Rattus Norvegicus Strain Wistar Model Type 2 Diabetes Mellitus (T2DM). Acta Inform Med 2018; 26:87-92. [PMID: 30061777 PMCID: PMC6029922 DOI: 10.5455/aim.2018.26.87-92] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Introduction: Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease caused by insulin resistance. Insulin resistance leads to hyperglycaemia that causes complication such as microangiopathy and macroangiopathy. The immune system of T2DM will be produce IL-10 as an anti-inflammatory cytokine role immune-stimulator and immunosuppressant in the organ system. This present study investigated of IL-10 gene profile and protein expression in the rat organ (Rattus norvegicus) strain Wistar model T2DM. Material and Methods: This research was used three of male rats group T2DM and three of male of normal rat as a control. The DNA tissues were isolated, amplified and sequenced by using IL-10 gene primer. The IL-10 protein profile and expression of rat tissues was analyzed using Experion-Pro260 gel and dot blotting using IL-10 antibody. Results: This study showed the differential expression of IL-10 gene profile among tissues among normal and T2DM groups. The IL-10 gene sequences, we found eight mutations in brain and twenty-seven mutations on gastric of T2DM group compare with control group, meanwhile there are no mutation in other tissues of both groups. The protein profile of all tissues in both groups was completely diverse as proper. Moreover, the level expression of IL-10 of heart, lung, gastric and kidney of T2DM group was lower than other tissues of both groups. Conclusion: This study concludes that T2DM animal model triggering mutation of IL-10 gene sequences of brain and gastric and induced the increasing level expression of IL-10 of ileum, brain and liver.
Collapse
Affiliation(s)
- Yohanes Bare
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, East Java, Indonesia.,Research Center of Smart Molecule of Natural Genetics Resources UB, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, East Java, Indonesia
| | - Agung Pramana Warih Marhendra
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, East Java, Indonesia
| | - Tomohiko Sasase
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc, Osaka, Japan
| | - Fatchiyah Fatchiyah
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, East Java, Indonesia.,Research Center of Smart Molecule of Natural Genetics Resources UB, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, East Java, Indonesia
| |
Collapse
|
36
|
MERS-CoV infection in humans is associated with a pro-inflammatory Th1 and Th17 cytokine profile. Cytokine 2018; 104:8-13. [PMID: 29414327 PMCID: PMC7129230 DOI: 10.1016/j.cyto.2018.01.025] [Citation(s) in RCA: 441] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 11/24/2022]
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) has been recognized as a highly pathogenic virus to humans that infects the respiratory tract and is associated with high morbidity and mortality. Studies in animal models suggest that MERS-CoV infection induces a strong inflammatory response, which may be related to the severity of disease. Data showing the cytokine profiles in humans during the acute phase of MERS-CoV infection are limited. In this study, we have analyzed the profile of cytokine responses in plasma samples from patients with confirmed MERS-CoV infections (n = 7) compared to healthy controls (n = 13). The cytokine profiles, including T helper (Th) 1, Th2 and Th17 responses, were analyzed using cytometric bead array (CBA). A prominent pro-inflammatory Th1 and Th17 response was clearly seen in patients with MERS-CoV infection, with markedly increased concentrations of IFN-γ, TNF-α, IL-15 and IL-17 compared to controls. IL-12 expression levels showed no difference between patients with MERS-CoV infection and the healthy controls despite the significantly increased levels of IFN-α2 and IFN-γ (P < .01). No changes were observed in the levels of IL-2, IL-4, IL-5, IL-13, and TGF-α (P > .05). Our results demonstrate a marked pro-inflammatory cytokine response during the acute phase of MERS-CoV infection in humans.
Collapse
|
37
|
Chamoun MN, Blumenthal A, Sullivan MJ, Schembri MA, Ulett GC. Bacterial pathogenesis and interleukin-17: interconnecting mechanisms of immune regulation, host genetics, and microbial virulence that influence severity of infection. Crit Rev Microbiol 2018; 44:465-486. [PMID: 29345518 DOI: 10.1080/1040841x.2018.1426556] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine involved in the control of many different disorders, including autoimmune, oncogenic, and diverse infectious diseases. In the context of infectious diseases, IL-17 protects the host against various classes of microorganisms but, intriguingly, can also exacerbate the severity of some infections. The regulation of IL-17 expression stems, in part, from the activity of Interleukin-23 (IL-23), which drives the maturation of different classes of IL-17-producing cells that can alter the course of infection. In this review, we analyze IL-17/IL-23 signalling in bacterial infection, and examine the interconnecting mechanisms that link immune regulation, host genetics, and microbial virulence in the context of bacterial pathogenesis. We consider the roles of IL-17 in both acute and chronic bacterial infections, with a focus on mouse models of human bacterial disease that involve infection of mucosal surfaces in the lungs, urogenital, and gastrointestinal tracts. Polymorphisms in IL-17-encoding genes in humans, which have been associated with heightened host susceptibility to some bacterial pathogens, are discussed. Finally, we examine the implications of IL-17 biology in infectious diseases for the development of novel therapeutic strategies targeted at preventing bacterial infection.
Collapse
Affiliation(s)
- Michelle N Chamoun
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| | - Antje Blumenthal
- b The University of Queensland Diamantina Institute, Translational Research Institute , Brisbane , Australia
| | - Matthew J Sullivan
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| | - Mark A Schembri
- c School of Chemistry and Molecular Biosciences, and Australian Infectious Disease Research Centre , The University of Queensland , Brisbane , Australia
| | - Glen C Ulett
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| |
Collapse
|
38
|
Kong Y, Zhang Y, Zhao X, Wang G, Liu Q. Carboxymethyl-chitosan attenuates inducible nitric oxide synthase and promotes interleukin-10 production in rat chondrocytes. Exp Ther Med 2017; 14:5641-5646. [PMID: 29285104 DOI: 10.3892/etm.2017.5258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/10/2017] [Indexed: 11/06/2022] Open
Abstract
Osteoarthritis (OA) is a common age-related degenerative joint disease, which is caused by the breakdown of joint cartilage and the underlying bone. Carboxymethyl (CM)-chitosan is a soluble derivative of chitosan that has similar physicochemical properties to the extracellular proteoglycans identified in hyaline cartilage. Previous studies have demonstrated that CM-chitosan serves a protective role in a rabbit OA model. The aim of the present study was to investigate the effect of CM-chitosan on NO production and inflammation through its upregulation of interleukin (IL)-10, and the activation of the janus kinase (JAK)/signal transducer and activator of transcription (STAT)/suppressor of cytokine signaling (SOCS) signaling pathway. In the present study primary rat chondrocytes were induced to inflammation with 2 µg/ml lipopolysaccharide. The cells were subsequently subjected to increasing concentrations of CM-chitosan (50, 100 and 200 µg/ml) and the relative mRNA and protein expression of inducible nitric oxide synthase (iNOS), IL-10, JAK1, STAT3 and SOCS3 were measured by RT-qPCR and western blot analysis respectively. The results revealed that CM-chitosan attenuated inflammation by significantly reducing iNOS expression and upregulating the anti-inflammatory cytokine IL-10 in a dose-dependent manner (P<0.05). The expression of JAK1, STAT3 and SOCS3 were also significantly upregulated by CM-chitosan (all P<0.05). The protective role of CM-chitosan against NO production was due to its upregulation of IL-10 and its activation of the JAK/STAT/SOCS signaling pathway.
Collapse
Affiliation(s)
- Ying Kong
- Department of Bone and Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Yuanmin Zhang
- Department of Bone and Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Xiaowei Zhao
- Department of Bone and Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Guodong Wang
- Department of Bone and Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Qingkuan Liu
- Department of Bone and Joint Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
39
|
Acharya AB, Thakur S, Muddapur MV, Kulkarni RD. Cytokine ratios in chronic periodontitis and type 2 diabetes mellitus. Diabetes Metab Syndr 2017; 11:277-278. [PMID: 27989515 DOI: 10.1016/j.dsx.2016.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 12/12/2016] [Indexed: 01/22/2023]
Abstract
AIM Chronic periodontitis may influence systemic cytokines in type 2 diabetes. This study aimed to evaluate the cytokine ratios in type 2 diabetes with, and without chronic periodontitis. MATERIALS & METHODS Gingival status, periodontal, glycemic parameters and serum cytokines were evaluated in participants grouped as healthy, chronic periodontitis, and type 2 diabetes with, and without chronic periodontitis. RESULTS AND CONCLUSIONS Cytokine ratios showed significant differences in type 2 diabetes and chronic periodontitis, were highest in participants having both type 2 diabetes and chronic periodontitis, with a statistically significant cut-off point and area under curve by receiver operating characteristic.
Collapse
Affiliation(s)
- Anirudh B Acharya
- Department of Periodontics, S.D.M. College of Dental Sciences & Hospital, Dharwad-580009, Karnataka, India.
| | - Srinath Thakur
- S.D.M. College of Dental Sciences & Hospital, Dharwad-580009, Karnataka, India
| | - M V Muddapur
- Faculty of Statistics, S.D.M. College of Dental Sciences & Hospital, Dharwad-580009, Karnataka, India
| | - Raghavendra D Kulkarni
- Department of Microbiology, S.D.M. College of Medical Sciences & Hospital, Dharwad-580009, Karnataka, India
| |
Collapse
|
40
|
Zou R, Li D, Wang G, Zhang M, Zhao Y, Yang Z. TAZ Activator Is Involved in IL-10-Mediated Muscle Responses in an Animal Model of Traumatic Brain Injury. Inflammation 2017; 40:100-105. [PMID: 27718096 DOI: 10.1007/s10753-016-0457-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The transcriptional coactivator with PDZ-binding motif (TAZ) functions as a downstream regulatory target in the Hippo signaling pathway that plays various roles. We previously developed a cell-based assay and identified the TAZ activator IBS008738 as a potential therapeutic target for glucocorticoid-induced atrophy. To further explore the application of IBS008738 in various muscle-related diseases, we examined the function of IBS008738 in inflammatory cytokine-mediated mouse muscle responses after traumatic brain injury (TBI). Preliminary screening suggested that IBS008738 treatments increased the levels of IL-10 in C2C12 cells. In TBI and sham control mice, we compared the effect of IBS008738 treatments on TNF α, IL-6, and IL-10 mRNA levels, muscle morphologic changes, and macrophage phenotype changes. Our findings support that the TAZ activator IBS008738 decreases muscle wasting by upregulating IL-10 and inhibiting TNF α and IL-6, and this process is implemented by changing the macrophage phenotypes. These results indicate a new mechanism of the TAZ activator as a potential therapy for atrophy.
Collapse
Affiliation(s)
- Ruyi Zou
- Department of Neurosurgery, General Hospital of Benxi Iron and Steel CO. LTD, Benxi, 117000, China
| | - Da Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Gang Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Mo Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yili Zhao
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, 23507, VA, USA
| | - Zeyu Yang
- Department of Ultrasound, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
41
|
Boulassel MR, Al-Ghonimi M, Al-Balushi B, Al-Naamani A, Al-Qarni Z, Wali Y, Elshinawy M, Al-Shezawi M, Khan H, Nazir H, Khater D, Pathare A, Al-Kindi S. Regulatory B Cells Are Functionally Impaired in Patients Having Hemophilia A With Inhibitors. Clin Appl Thromb Hemost 2017; 24:618-624. [PMID: 28393618 DOI: 10.1177/1076029617702244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Development of inhibitors remains a major clinical complication in patients with hemophilia A receiving replacement therapy with factor VIII (FVIII). Understanding the immune mechanisms involved in the development of inhibitors can provide valuable information about pathways to human tolerance. Recent evidence indicates that B regulatory (Breg) cells play a pivotal role in controlling the production of antibodies (Abs) while promoting follicular T helper (Tfh) cells and monocytes, expressing the low-density lipoprotein receptor-related protein (LRP/CD91), which is involved in FVIII intake from the circulation. We studied circulating levels of Breg cells along with Tfh cells and the expression of LRP/CD91 on monocytes in patients with hemophilia A using 8-color flow cytometry and cell culture. Compared to healthy controls, patients with hemophilia A with inhibitors showed a severe reduction in levels of Breg cells and produced less interleukin-10 when activated via the CD40 signaling pathway. In addition, patients with hemophilia A with inhibitors exhibited an overexpression of LPR/CD91 on monocytes and normal levels of Tfh cells. Levels of Breg cells were not significantly related to LPR/CD91 although negative associations were evidenced. Collectively, these results provide new insights into the role of Breg cells and LPR/CD91 in the development of inhibitors in patients with hemophilia A.
Collapse
Affiliation(s)
- Mohamed-Rachid Boulassel
- 1 Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman.,2 Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Maryam Al-Ghonimi
- 1 Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Badriya Al-Balushi
- 2 Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Amal Al-Naamani
- 1 Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Zahra Al-Qarni
- 1 Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Yasser Wali
- 3 Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman.,4 Faculty of Medicine, Pediatric Department, Alexandria University, Alexandria Governorate, Egypt
| | - Mohamed Elshinawy
- 3 Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman.,4 Faculty of Medicine, Pediatric Department, Alexandria University, Alexandria Governorate, Egypt
| | - Maryam Al-Shezawi
- 3 Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Hamad Khan
- 2 Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Hanan Nazir
- 3 Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman.,4 Faculty of Medicine, Pediatric Department, Alexandria University, Alexandria Governorate, Egypt
| | - Doaa Khater
- 3 Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman.,4 Faculty of Medicine, Pediatric Department, Alexandria University, Alexandria Governorate, Egypt
| | - Anil Pathare
- 2 Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Salam Al-Kindi
- 2 Department of Haematology, College of Medicine and Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
42
|
Regulatory T Cells: Molecular Actions on Effector Cells in Immune Regulation. J Immunol Res 2016; 2016:1720827. [PMID: 27298831 PMCID: PMC4889823 DOI: 10.1155/2016/1720827] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/27/2016] [Indexed: 01/11/2023] Open
Abstract
T regulatory cells play a key role in the control of the immune response, both in health and during illness. While the mechanisms through which T regulatory cells exert their function have been extensively described, their molecular effects on effector cells have received little attention. Thus, this revision is aimed at summarizing our current knowledge on those regulation mechanisms on the target cells from a molecular perspective.
Collapse
|
43
|
In Vivo Molecular Dissection of the Effects of HIV-1 in Active Tuberculosis. PLoS Pathog 2016; 12:e1005469. [PMID: 26986567 PMCID: PMC4795555 DOI: 10.1371/journal.ppat.1005469] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/02/2016] [Indexed: 12/18/2022] Open
Abstract
Increased risk of tuberculosis (TB) associated with HIV-1 infection is primarily attributed to deficient T helper (Th)1 immune responses, but most people with active TB have robust Th1 responses, indicating that these are not sufficient to protect against disease. Recent findings suggest that favourable outcomes following Mycobacterium tuberculosis infection arise from finely balanced inflammatory and regulatory pathways, achieving pathogen control without immunopathology. We hypothesised that HIV-1 and antiretroviral therapy (ART) exert widespread changes to cell mediated immunity, which may compromise the optimal host protective response to TB and provide novel insights into the correlates of immune protection and pathogenesis. We sought to define these effects in patients with active TB by transcriptional profiling of tuberculin skin tests (TST) to make comprehensive molecular level assessments of in vivo human immune responses at the site of a standardised mycobacterial challenge. We showed that the TST transcriptome accurately reflects the molecular pathology at the site of human pulmonary TB, and used this approach to investigate immune dysregulation in HIV-1/TB co-infected patients with distinct clinical phenotypes associated with TST reactivity or anergy and unmasking TB immune reconstitution inflammatory syndrome (IRIS) after initiation of ART. HIV-1 infected patients with positive TSTs exhibited preserved Th1 responses but deficient immunoregulatory IL10-inducible responses. Those with clinically negative TSTs revealed profound anergy of innate as well as adaptive immune responses, except for preservation of type 1 interferon activity, implicated in impaired anti-mycobacterial immunity. Patients with unmasking TB IRIS showed recovery of Th1 immunity to normal levels, but exaggerated Th2-associated responses specifically. These mechanisms of immune dysregulation were localised to the tissue microenvironment and not evident in peripheral blood. TST molecular profiling categorised different mechanisms of immunological dysfunction in HIV-1 infection beyond the effects on CD4 T cells, each associated with increased risk of TB disease and amenable to host-directed therapies.
Collapse
|
44
|
Acharya AB, Thakur S, Muddapur MV. Evaluation of serum interleukin-10 levels as a predictor of glycemic alteration in chronic periodontitis and type 2 diabetes mellitus. J Indian Soc Periodontol 2015; 19:388-92. [PMID: 26392685 PMCID: PMC4555794 DOI: 10.4103/0972-124x.150876] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/24/2014] [Indexed: 12/13/2022] Open
Abstract
Aim: Chronic periodontal disease (CPD) and type 2 diabetes mellitus (T2DM) share common pathogenic pathways. This study aimed to estimate levels of serum interleukin (IL-10), an anti-inflammatory cytokine also associated with T2DM and evaluate its association with hyperglycemia. Materials and Methods: This investigation involved sixty participants divided into four groups comprising 15 participants each: Group 1 (healthy controls), Group 2 (CPD patients), Group 3 (T2DM patients with CPD) and Group 4 (T2DM patients). Plaque index, gingival index, probing pocket depths (PPD), clinical attachment loss, bleeding on probing, random blood sugar, glycosylated hemoglobin (HbA1c), and serum IL-10 was measured. Results: Interleukin-10 was detected in all four groups. Statistically significant (P < 0.05) differences were observed in most of the variables in all groups. IL-10 correlated significantly with PPD in Group 1 and with HbA1c in Group 4. IL-10 regressed with PPD in Group 1 and with HbA1c in Group 4. IL-10 levels were lower in Group 3 when compared with Group 4 and was lowest in Group 2. Conclusion: Low IL-10 levels associated with high HbA1c. Pathogenic mechanisms of CPD seem to regulate IL-10. Serum IL-10 levels may be one of the predictors of glycemia.
Collapse
Affiliation(s)
| | - Srinath Thakur
- Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, Dharwad, Karnataka, India
| | | |
Collapse
|
45
|
Interleukin-10 modulates antigen presentation by dendritic cells through regulation of NLRP3 inflammasome assembly during Chlamydia infection. Infect Immun 2015; 83:4662-72. [PMID: 26371131 DOI: 10.1128/iai.00993-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/11/2015] [Indexed: 01/12/2023] Open
Abstract
Interleukin-10 (IL-10) has been implicated in susceptibility to genital chlamydial infection and the development of tubal pathologies. IL-10 limitation also resulted in the rapid elicitation of immune responses against Chlamydia, and decreased levels of IL-10 correlated with protective anti-Chlamydia immunity. To investigate the molecular basis for these effects, we compared the reproductive pathologies and fertility rates in Chlamydia-infected wild-type (WT) and IL-10-knockout (IL-10(-/-)) mice; we also analyzed the expression of the Toll-like receptor (TLR)/interleukin-1 receptor (IL-1R) superfamily, IL-1β production, NLRP3 inflammasome assembly and activation, and the immunostimulatory capacity and apoptotic predilection of Chlamydia-exposed dendritic cells (DCs) from WT and IL-10(-/-) mice. Our results revealed that, in addition to the rapid clearance of infection, genitally infected IL-10(-/-) mice were protected from tubal pathologies and infertility, whereas WT (IL-10(+/+)) mice were not. Chlamydia-pulsed IL-10(-/-) DCs expressed larger numbers of TLR4/IL-1R molecules and had enhanced IL-1β production. In addition, NLRP3 inflammasome assembly was suppressed in IL-10(-/-) DCs through the inhibition of the P2X purinoceptor 7 (P2X7) receptor (P2X7R), an ATP-gated ion channel, and a decrease in intracellular Ca(2+) levels, which inhibited DC apoptosis. Thus, the potent immunostimulatory capacity of IL-10-deficient DCs is due, at least in part, to the suppression of the intracellular inflammasome assembly, which prevents DC apoptosis, allowing efficient antigen presentation. The results indicate that IL-10 deficiency enables efficient antigen presentation by DCs for rapid and enhanced immune activation against Chlamydia, which results in rapid microbial clearance, which prevents tubal pathologies during infection. Our finding has important implications for the induction of protective immunity against Chlamydia and other infectious and noninfectious diseases by vaccines.
Collapse
|
46
|
Moreira ML, Dorneles EMS, Soares RP, Magalhães CP, Costa-Pereira C, Lage AP, Teixeira-Carvalho A, Martins-Filho OA, Araújo MSS. Cross-reactivity of commercially available anti-human monoclonal antibodies with canine cytokines: establishment of a reliable panel to detect the functional profile of peripheral blood lymphocytes by intracytoplasmic staining. Acta Vet Scand 2015; 57:51. [PMID: 26362860 PMCID: PMC4566394 DOI: 10.1186/s13028-015-0142-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/27/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The process for obtaining monoclonal antibodies against a specific antigen is very laborious, involves sophisticated technologies and it is not available in most research laboratories. Considering that most cytokines remain partially conserved among species during evolution, the search for antibody cross-reactivity is an important strategy for immunological studies in veterinary medicine. In this context, the amino acid sequence from human and canine cytokines have demonstrated 49-96 % homology, suggesting high probability of cross-reactivity amongst monoclonal antibodies. For this, 17 commercially available anti-human monoclonal antibodies [IL-1α, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8 (#1, #2), IL-10, IL-12, IL-13, IL-17A, IFN-γ (#1, #2), TNF-α (#1, #2) and TGF-β], were evaluated in vitro for intracellular cytokine detection in a stimulated canine blood culture by flow cytometry and confocal microscopy. Lymphocytes from peripheral blood of healthy and two unhealthy dogs were analyzed. RESULTS Eleven anti-human mAbs [IL-1α, IL-4, IL-5, IL-6, IL-8 (#1, #2), IL-12, IL-17A, TNF-α (#1, #2) and TGF-β] cross-reacted against canine intracellular cytokines. The specificity of the assays was not affected after Fc-blocking. Three anti-human cytokine mAbs [IL-4, IL-8 (#2) and TGF-β] when evaluated by confocal microscopy also cross-reacted with intracellular canine cytokines. The identification of human mAbs that cross-reacted with canine cytokines may support their use as immunological biomarkers in veterinary medicine studies. CONCLUSION The identification of these 11 anti-human cytokine mAbs that cross-reacted with canine cytokines will be useful immunological biomarkers for pathological conditions by flow cytometry and fluorescence microscopy in dogs.
Collapse
|
47
|
Genomic analysis of LPS-stimulated myeloid cells identifies a common pro-inflammatory response but divergent IL-10 anti-inflammatory responses. Sci Rep 2015; 5:9100. [PMID: 25765318 PMCID: PMC4650320 DOI: 10.1038/srep09100] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/12/2015] [Indexed: 01/02/2023] Open
Abstract
Inflammation is an essential physiological response to infection and injury that must be kept within strict bounds. The IL-10/STAT3 anti-inflammatory response (AIR) is indispensable for controlling the extent of inflammation, although the complete mechanisms downstream of STAT3 have not yet been elucidated. The AIR is widely known to extend to other myeloid cells, but it has best been characterized in macrophages. Here we set out to characterize the LPS-mediated pro-inflammatory response and the AIR across a range of myeloid cells. We found that whereas the LPS-induced pro-inflammatory response is broadly similar among macrophages, dendritic cells, neutrophils, mast cells and eosinophils, the AIR is drastically different across all myeloid cell types that respond to IL-10 (all bar eosinophils). We propose a model whereby the IL-10/STAT3 AIR works by selectively inhibiting specific pathways in distinct cell types: in macrophages the AIR most likely works through the inhibition of NF-κB target genes; in DCs and mast cells through indirect IRF disruption; and in neutrophils through IRF disruption and possibly also indirect NF-κB inhibition. In summary, no conserved IL-10/STAT3 AIR effectors were identified; instead a cell type-specific model of the AIR is proposed.
Collapse
|
48
|
Mittal SK, Roche PA. Suppression of antigen presentation by IL-10. Curr Opin Immunol 2015; 34:22-7. [PMID: 25597442 DOI: 10.1016/j.coi.2014.12.009] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 12/13/2022]
Abstract
Regulated antigen presentation to immune cells determines the effectiveness of an immune response. IL-10 is an immunosuppressive cytokine that regulates immune responses by inhibiting the ability of APCs to present antigens to T cells in a variety of ways. The mechanisms of IL-10-mediated immunosuppression include interference in TLR-mediated or IFNγ-mediated dendritic cell (DC) and macrophage activation as well as direct induction of genes that suppress APC function. In this review we will discuss current studies exploring the molecular mechanisms by which IL-10 suppresses APC function.
Collapse
Affiliation(s)
- Sharad K Mittal
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
49
|
Qu H, Xu Y, Sun H, Lin J, Yu J, Tang Z, Shen J, Liang C, Li S, Chen W, Li X, Wu Z, Huang Y, Yu X. Systemic and local mucosal immune responses induced by orally delivered Bacillus subtilis spore expressing leucine aminopeptidase 2 of Clonorchis sinensis. Parasitol Res 2014; 113:3095-103. [PMID: 24906994 DOI: 10.1007/s00436-014-3975-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 05/25/2014] [Indexed: 11/25/2022]
Abstract
Human clonorchiasis caused by Clonorchis sinensis (C. sinensis) has been increasingly prevalent in recent years so that an effective measure is essential and urgent to control the infectious disease. Oral delivery of antigens from C. sinensis may be an important approach to effectively induce both systemic and local immune responses to anti-infection of the parasite. In the current study, we used Bacillus subtilis (B. subtilis) spores as a delivery vehicle to introduce leucine aminopeptidase 2 of C. sinensis (CsLAP2), an excretory/secretory antigen with high immunogenicity, expressing on their surface. SDS-PAGE, western blotting, and flow cytometry indicated that CsLAP2 was successfully expressed on the surface of B. subtilis spores (CotC-CsLAP2 spores). BALB/c mice were treated with spores intragastrically. On day 31 after the treatment, we found that mice intragastrically treated with CotC-CsLAP2 spores exhibited higher IgG, IgG1, IgG2a, and IgA level in sera as well as higher sIgA level in bile and intestinal lavage fluid compared to mice orally administrated with spores not expressing CsLAP2 (CotC spores) and naïve mice. The peak titer of IgG/IgA presented on day 31/49 after oral administration. IgG1 level was lower than IgG2a in group administrated with CotC-CsLAP2 spores. sIgA-secreting cells were obviously observed in intestinal epithelium of mice orally treated with CotC-CsLAP2 spores. After incubated with CotC-CsLAP2, the levels of IFN-γ, IL-6, IL-10, IL-17A, and TNF significantly increased in the supernatant of splenocytes isolated from mice orally treated with CotC-CsLAP2 spores, while there was no statistically significant difference of IL-4 level representing Th2 response among the groups. Our study demonstrated that oral administration of CsLAP2 delivered by B. subtilis spore elicited obvious systemic and local mucosal immunity. Secretory IgA and Th1-Th17 cellular immunity might involved in mechanisms of the immune response.
Collapse
Affiliation(s)
- Hongling Qu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|