1
|
Gaggioli MR, Jones AG, Panagi I, Washington EJ, Loney RE, Muench JH, Foster MW, Brennan RG, Thurston TLM, Ko DC. A single amino acid in the Salmonella effector SarA/SteE triggers supraphysiological activation of STAT3 for anti-inflammatory gene expression. Cell Rep 2025; 44:115530. [PMID: 40188438 PMCID: PMC12014907 DOI: 10.1016/j.celrep.2025.115530] [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: 02/20/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/08/2025] Open
Abstract
Salmonella causes ∼1 million cases of gastroenteritis annually in the United States. Critical to virulence are secreted effectors that reprogram host functions. We previously discovered the effector SarA facilitates phosphorylation of STAT3, inducing expression of the anti-inflammatory cytokine interleukin-10 (IL-10). This STAT3 activation requires a region of homology with the host cytokine receptor gp130. Here, we demonstrate that a single amino acid difference is critical for the anti-inflammatory bias of SarA-STAT3 signaling. An isoleucine at pY+1 of the YxxQ motif in SarA (which binds the STAT3 SH2 domain) causes increased STAT3 recruitment and phosphorylation, biasing toward anti-inflammatory targets. This isoleucine renders SarA a better substrate for tyrosine phosphorylation by GSK-3. GSK-3 is canonically a serine/threonine kinase that nonetheless undergoes tyrosine autophosphorylation at a motif with isoleucine at the pY+1 position. Our results provide a molecular basis for how a Salmonella effector achieves supraphysiological levels of STAT3 activation to control host genes.
Collapse
Affiliation(s)
- Margaret R Gaggioli
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Angela G Jones
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Ioanna Panagi
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Erica J Washington
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Rachel E Loney
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | | | - Matthew W Foster
- Duke Proteomics and Metabolomics Core Facility, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Richard G Brennan
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Teresa L M Thurston
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Feng A, Gonzalez MV, Kalaycioglu M, Yin X, Mumau M, Shyamsundar S, Bustamante MS, Chang SE, Dhingra S, Dodig-Crnkovic T, Schwenk JM, Garg T, Yoshizaki K, van Rhee F, Fajgenbaum DC, Utz PJ. Common connective tissue disorder and anti-cytokine autoantibodies are enriched in idiopathic multicentric castleman disease patients. Front Immunol 2025; 16:1528465. [PMID: 40181993 PMCID: PMC11966032 DOI: 10.3389/fimmu.2025.1528465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Idiopathic Multicentric Castleman Disease (iMCD) is a polyclonal lymphoproliferative disorder involving cytokine storms that can lead to organ failure and death. The cause of iMCD is unknown, but some clinical evidence suggests an autoimmune etiology. For example, connective tissue disorders (CTDs) and iMCD share many clinical features, and autoantibodies have been anecdotally reported in individual iMCD patients. This study investigates whether common autoantibodies are shared across iMCD patients. Methods We assembled custom bead-based protein arrays consisting of 52 autoantigens traditionally associated with CTDs and 38 full-length cytokines and screened serum samples from 101 iMCD patients for IgG autoantibodies. We also screened samples with a 1,103-plex array of recombinant human protein fragments to identify additional autoantibody targets. Finally, we performed receptor blocking assays on select samples with anti-cytokine autoantibodies (ACAs) identified by array. Results We found that an increased proportion of iMCD patients (47%) tested positive for at least one CTD-associated autoantibody compared to healthy controls (HC) (17%). Commonly detected CTD-associated autoantibodies were associated with myositis and overlap syndromes as well as systemic lupus erythematosus (SLE) and Sjögren's Syndrome (SS). ACAs were also detected in a greater proportion of iMCD patients (38%) compared to HC (10%), while the protein fragment array identified a variety of other autoantibody targets. One iMCD sample tested positive for receptor blocking against interferon-ω (IFNω). Discussion IgG autoantibodies binding autoantigens associated with common CTDs and cytokines are elevated in iMCD patients compared to HC, suggesting that autoimmunity may be involved in iMCD pathogenesis.
Collapse
Affiliation(s)
- Allan Feng
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael V. Gonzalez
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Muge Kalaycioglu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Xihui Yin
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Melanie Mumau
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Saishravan Shyamsundar
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Mateo Sarmiento Bustamante
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah E. Chang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Shaurya Dhingra
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Tea Dodig-Crnkovic
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jochen M. Schwenk
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tarun Garg
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Kazuyuki Yoshizaki
- Department of Biomolecular Science and Regulation, The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Frits van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA, United States
| | - Paul J. Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
3
|
Wang J, Wu N, Zhang J, Li X, Hu Y, Dai J, Shen C, Chen X. Ciliary neurotrophic factor attenuates myocardial infarction-induced oxidative stress and ferroptosis via PI3K/Akt signaling. J Mol Histol 2025; 56:90. [PMID: 39954087 DOI: 10.1007/s10735-025-10359-w] [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: 11/21/2023] [Accepted: 01/19/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND As a member of the interleukin-6 family, ciliary neurotrophic factor (CNTF) regulates inflammation, oxidative stress, and other processes to exhibit neurotrophic and differentiating effects over cells in the central nervous system. It has not yet been documented, therefore, if CNTF influences the cardiac remodeling brought on by myocardial infarction (MI). The purpose of the current investigation was to identify the function and underlying mechanisms of CNTF in cardiac remodeling brought on by MI. METHODS Using an adeno-associated virus 9 (AAV9) system and tail vein injection, we overexpressed CNTF in the hearts. To create a model of MI, C57BL/6 mice underwent left anterior descending (LAD) ligation. The following techniques were employed to assess the impact of CNTF overexpression and the underlying mechanisms: quantitative real-time PCR, western blotting, histological analysis, immunofluorescence and immunohistochemistry analysis, and echocardiography. We used H9c2 cells to confirm CNTF's in vitro effects. RESULTS In MI mice, overexpression of CNTF prevents cardiac hypertrophy and cardiac fibrosis. Furthermore, oxidative stress and ferroptosis in response to MI damage were markedly reduced by CNTF overexpression. Mechanistically, overexpression of CNTF in both in vivo and in vitro markedly enhanced PI3K/Akt signaling. However, blocking this pathway effectively negated the beneficial impact of CNTF overexpression. CONCLUSIONS Our research indicates that via initiating the PI3K/Akt signaling pathway, CNTF controls myocardial dysfunction, oxidative stress, and ferroptosis in MI-induced cardiac remodeling. CNTF may have therapeutic potential in treating MI-induced cardiac remodeling.
Collapse
Affiliation(s)
- Jian Wang
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Nan Wu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Jie Zhang
- Department of Intensive Care Unit, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Xiaojing Li
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Yingchu Hu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Jiating Dai
- Health Science Center, Ningbo University, Ningbo, 315000, China
| | - Caijie Shen
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China.
- , 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China.
| | - Xiaomin Chen
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China.
- , 59 Liuting Street, Haishu District, Ningbo, Zhejiang, China.
| |
Collapse
|
4
|
Niu L, Gao M, Li Y, Wang C, Zhang C, Duan H, Li H, Wang F, Ge J. Effects of the stress hormone norepinephrine on the probiotic properties of Levilactobacillus: antibacterial colonization, anti-inflammation, and antioxidation. Front Microbiol 2025; 16:1526362. [PMID: 39996081 PMCID: PMC11849050 DOI: 10.3389/fmicb.2025.1526362] [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: 11/12/2024] [Accepted: 01/16/2025] [Indexed: 02/26/2025] Open
Abstract
Probiotics as antibiotic alternatives are unstable for use under stress in clinical applications. To explore the influence of catecholamine hormones on probiotic bacterial inhibition and antimicrobial activity, we tested the effects of norepinephrine (NE) on Levilactobacillus in vitro and in a mouse model. The in vitro results showed that in the presence of NE, 80% of Levilactobacillus strains showed increased growth rate and more than 80% of the strains indicated lower antimicrobial activity at 22 h. Furthermore, in the mouse model, NE weakens the protective effect of L. brevis 23,017 on Escherichia coli infection, which is shown by the decreased ability of antibacterial colonization, antioxidation, and anti-inflammation, and downregulating the expression of antioxidant genes and intestinal mucosal barrier-related genes. At the same time, the addition of NE modulates the bacterial microbiota richness and diversity in the intestine, disrupting the balance of intestinal probiotics. These findings provide evidence that NE reduces the probiotic ability of Levilactobacillus and illustrates the plasticity of the probiotics in response to the intestinal microenvironment under stress.
Collapse
Affiliation(s)
- Lingdi Niu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingchun Gao
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yifan Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chaonan Wang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chuankun Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Haoyuan Duan
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hai Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Fang Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Junwei Ge
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
5
|
Remali J, Aizat WM. Medicinal plants and plant-based traditional medicine: Alternative treatments for depression and their potential mechanisms of action. Heliyon 2024; 10:e38986. [PMID: 39640650 PMCID: PMC11620067 DOI: 10.1016/j.heliyon.2024.e38986] [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: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
Background Clinical depression is a serious public health issue that affects 4.7 % of the world's population and can lead to suicide tendencies. Although drug medications are available, only 60 % of the depressed patients respond positively to the treatments, while the rest experience side effects that resulted in the discontinuation of their medication. Thus, there is an urgent need for developing a new anti-depressant with a distinct mode of action and manageable side effects. One of the options is using medicinal plants or plant-based traditional medicine as alternative therapies for psychiatric disorders. Objectives Therefore, the objective of this review was twofold; to identify and critically evaluate anti-depressant properties of medicinal plants or those incorporated in traditional medicine; and to discuss their possible mechanism of action as well as challenges and way forward for this alternative treatment approach. Methods Relevant research articles were retrieved from various databases, including Scopus, PubMed, and Web of Science, for the period from 2018 to 2020, and the search was updated in September 2024. The inclusion criterion was relevance to antidepressants, while the exclusion criteria included duplicates, lack of full-text availability, and non-English publications. Results Through an extensive literature review, more than 40 medicinal plant species with antidepressant effects were identified, some of which are part of traditional medicine. The list of the said plant species included Albizia zygia (DC.) J.F.Macbr., Calculus bovis Sativus, Celastrus paniculatus Willd., Cinnamomum sp., Erythrina velutina Willd., Ficus platyphylla Delile, Garcinia mangostana Linn., Hyptis martiusii Benth, and Polygonum multiflorum Thunb. Anti-depressant mechanisms associated with those plants were further characterised based on their modes of action such as anti-oxidation system, anti-inflammation action, modulation of various neurotransmitters, neuroprotective effect, the regulation of hypothalamic-pituitary-adrenal (HPA) axis and anti-depressant mechanism. The challenges and future outlook of this alternative and complementary medicine are also explored and discussed. Conclusion This pool of identified plant species is hoped to offer health care professionals the best possible alternatives of anti-depressants from natural phytocompounds that are efficacious, safe and affordable for applications in future clinical settings.
Collapse
Affiliation(s)
- Juwairiah Remali
- Department of Pathology, Hospital Pulau Pinang, Jalan Residensi, 10450, George Town, Pulau Pinang, Malaysia
| | - Wan Mohd Aizat
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia (UKM), 43600, Bangi, Selangor, Malaysia
| |
Collapse
|
6
|
Roy A, Paul I, Luharuka S, Ray S. An in-silico scaffold- hopping approach to design novel inhibitors against gp130: A potential therapeutic application in cancer and Covid-19. Mol Divers 2024; 28:3129-3151. [PMID: 37934366 DOI: 10.1007/s11030-023-10737-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 09/25/2023] [Indexed: 11/08/2023]
Abstract
An upregulation of the gp130-signalling cascade has been reported in multiple cancers, making gp130 an attractive target for the development of anticancer drugs. An inverted-funnel-like approach was utilised along with various structure-based drug designing strategies to discover and optimise novel potential inhibitors of gp130. The study resulted in the discovery of 2 ligands- 435 and 510, both of which exhibit a very high-binding affinity towards the gp130 D1 domain which controls cytokine recognition and interaction thus being involved in complexation. The two resulting complexes remained stable over time with the ligands maintaining a steady interaction with the target. This inference is drawn from their RMSD, Rg, SASA and RMSF analysis. We also tested the protein folding patterns based on their principal component analysis, energy of surface and landscape. The leads also displayed a more favourable ADMET profile than their parent compounds. The two lead candidates show a better therapeutic profile in comparison to the two existing drugs- bazedoxifene and raloxifene. Both these potential leads can be addressed for their activity in-vitro and can be used as a potential anti-cancer treatment as well as to combat Covid-19 related cytokine storm.
Collapse
Affiliation(s)
- Alankar Roy
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Ishani Paul
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Shreya Luharuka
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Sujay Ray
- Amity Institute of Biotechnology, Amity University, Kolkata, India.
| |
Collapse
|
7
|
Shao J, Qiu X, Zhang L, Li S, Xue S, Si Y, Li Y, Jiang J, Wu Y, Xiong Q, Wang Y, Chen Q, Gao T, Zhu L, Wang H, Xie M. Multi-layered computational gene networks by engineered tristate logics. Cell 2024; 187:5064-5080.e14. [PMID: 39089254 DOI: 10.1016/j.cell.2024.07.001] [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: 10/30/2023] [Revised: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/03/2024]
Abstract
So far, biocomputation strictly follows traditional design principles of digital electronics, which could reach their limits when assembling gene circuits of higher complexity. Here, by creating genetic variants of tristate buffers instead of using conventional logic gates as basic signal processing units, we introduce a tristate-based logic synthesis (TriLoS) framework for resource-efficient design of multi-layered gene networks capable of performing complex Boolean calculus within single-cell populations. This sets the stage for simple, modular, and low-interference mapping of various arithmetic logics of interest and an effectively enlarged engineering space within single cells. We not only construct computational gene networks running full adder and full subtractor operations at a cellular level but also describe a treatment paradigm building on programmable cell-based therapeutics, allowing for adjustable and disease-specific drug secretion logics in vivo. This work could foster the evolution of modern biocomputers to progress toward unexplored applications in precision medicine.
Collapse
Affiliation(s)
- Jiawei Shao
- Department of Pharmacy, Center for Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China.
| | - Xinyuan Qiu
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China; College of Computer Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Lihang Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; Research Center for Life Sciences Computing, Zhejiang Laboratory, Hangzhou, Zhejiang 311100, China
| | - Shichao Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shuai Xue
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Yaqing Si
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yilin Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jian Jiang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yuhang Wu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Qiqi Xiong
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yukai Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China; School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Qidi Chen
- Department of Pharmacy, Center for Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang 322000, China
| | - Ting Gao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China.
| | - Hui Wang
- Research Center for Life Sciences Computing, Zhejiang Laboratory, Hangzhou, Zhejiang 311100, China.
| | - Mingqi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China; School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China.
| |
Collapse
|
8
|
Sawoo R, Bishayi B. TLR4/TNFR1 blockade suppresses STAT1/STAT3 expression and increases SOCS3 expression in modulation of LPS-induced macrophage responses. Immunobiology 2024; 229:152840. [PMID: 39126792 DOI: 10.1016/j.imbio.2024.152840] [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: 12/15/2023] [Revised: 07/15/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024]
Abstract
Due to the urgent need to create appropriate treatment techniques, which are currently unavailable, LPS-induced sepsis has become a serious concern on a global scale. The primary active component in the pathophysiology of inflammatory diseases such as sepsis is the Gram-negative bacterial lipopolysaccharide (LPS). LPS interacts with cell surface TLR4 in macrophages, causing the formation of reactive oxygen species (ROS), TNF-α, IL-1β and oxidative stress. It also significantly activates the MAPKs and NF-κB pathway. Excessive production of pro-inflammatory cytokines is one of the primary characteristic features in the onset and progression of inflammation. Cytokines mainly signal through the JAK/STAT pathway. We hypothesize that blocking of TLR4 along with TNFR1 might be beneficial in suppressing the effects of STAT1/STAT3 due to the stimulation of SOCS3 proteins. Prior to the LPS challenge, the macrophages were treated with antibodies against TLR4 and TNFR1 either individually or in combination. On analysis of the macrophage populations by flowcytometry, it was seen that receptor blockade facilitated the phenotypic shift of the M1 macrophages towards M2 resulting in lowered oxidative stress. Blocking of TLR4/TNFR1 upregulated the SOCS3 and mTOR expressions that enabled the transition of inflammatory M1 macrophages towards the anti-inflammatory M2 phenotype, which might be crucial in curbing the inflammatory responses. Also the reduction in the production of inflammatory cytokines such as IL-6, IL-1β due to the reduction in the activation of the STAT1 and STAT3 molecules was observed in our combination treatment group. All these results indicated that neutralization of both TLR4 and TNFR1 might provide new insights in establishing an alternative therapeutic strategy for LPS-sepsis.
Collapse
Affiliation(s)
- Ritasha Sawoo
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| |
Collapse
|
9
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [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: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
10
|
Fedorka CE, Scoggin KE, El-Sheikh Ali H, Troedsson MHT. Evaluating the IL-6 Family of Cytokines Throughout Equine Gestation. Am J Reprod Immunol 2024; 92:e13910. [PMID: 39072818 DOI: 10.1111/aji.13910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
INTRODUCTION The interleukin (IL)-6 family of cytokines is grouped by a common receptor subunit (gp130), but functions in distinct but overlapping physiological activities, including regulation of acute phase reaction and the balance between effector and regulatory T cell populations-both of which play a role in successful pregnancy maturation. METHODS Here, we aim to assess the expression profiles of members of the IL-6 cytokine family throughout equine gestation. To do so, RNA Sequencing was performed on chorioallantois and endometrium of mares at 120, 180, 300, and 330 days of gestation (n = 4/stage), as well as 45-day chorioallantois (n = 4) and diestrus endometrium (n = 3). Expression levels of members of the IL-6 cytokine family including ciliary neurotrophic factor (CNTF), cardiotrophin 1 (CT-1), cardiotrophin-like cytokine factor 1 (CLCF1), galectin-10, oncostatin M (OSM), and IL-6, -11, and -27 were evaluated in addition to the receptors for IL-6 (IL-6R) and the common receptor subunit gp130. Additionally, peripheral concentration of IL-6 was assessed. RESULTS In the chorioallantois, differential expression of IL-6, IL-11, CNTF, CLCF1, OSM, and CT-1 was noted. In the endometrium, the gestational age of pregnancy impacted the expression of IL-11, CNTF, and CT-1. Circulatory IL-6 concentrations reached their highest concentrations at 120 days, with lesser concentrations noted at 45, 180, 300, and 330 days. Both IL-6R and gp130 altered in expression throughout equine gestation. CONCLUSION In conclusion, members of the IL-6 cytokine family appear to fluctuate constantly throughout equine pregnancy, with varying expression profiles noted when comparing individual members. Additionally, different expression profiles were noted when comparing chorioallantois, endometrium, and circulation, indicating that the function of the cytokine is tissue-specific.
Collapse
Affiliation(s)
- Carleigh E Fedorka
- Department of Animal Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Kirsten E Scoggin
- Department of Veterinary Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Hossam El-Sheikh Ali
- Department of Veterinary Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Mats H T Troedsson
- Department of Veterinary Sciences, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
11
|
Bharti V, Kumar A, Wang Y, Roychowdhury N, de Lima Bellan D, Kassaye BB, Watkins R, Capece M, Chung CG, Hilinski G, Vilgelm AE. TTK inhibitor OSU13 promotes immunotherapy responses by activating tumor STING. JCI Insight 2024; 9:e177523. [PMID: 38900577 PMCID: PMC11383830 DOI: 10.1172/jci.insight.177523] [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: 11/13/2023] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
TTK spindle assembly checkpoint kinase is an emerging cancer target. This preclinical study explored the antitumor mechanism of TTK inhibitor OSU13 to define a strategy for clinical development. We observed prominent antitumor activity of OSU13 in melanoma, colon and breast cancer cells, organoids derived from patients with melanoma, and mice bearing colon tumors associated with G2 cell cycle arrest, senescence, and apoptosis. OSU13-treated cells displayed DNA damage and micronuclei that triggered the cytosolic DNA-sensing cGAS/STING pathway. STING was required for the induction of several proteins involved in T cell recruitment and activity. Tumors from OSU13-treated mice showed an increased proportion of T and NK cells and evidence of PD-1/PD-L1 immune checkpoint activation. Combining a low-toxicity dose of OSU13 with anti-PD-1 checkpoint blockade resulted in prominent STING- and CD8+ T cell-dependent tumor inhibition and improved survival. These findings provide a rationale for utilizing TTK inhibitors in combination with immunotherapy in STING-proficient tumors.
Collapse
Affiliation(s)
- Vijaya Bharti
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Amrendra Kumar
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Yinchong Wang
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
- Molecular Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Nikhil Roychowdhury
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Daniel de Lima Bellan
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Reese Watkins
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Marina Capece
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| | | | - Gerard Hilinski
- Drug Development Institute, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | - Anna E Vilgelm
- Department of Pathology
- Pelotonia Institute for Immunooncology, and
- Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
12
|
Gober IG, Russell AL, Shick TJ, Vagni VA, Carlson JC, Kochanek PM, Wagner AK. Exploratory assessment of the effect of systemic administration of soluble glycoprotein 130 on cognitive performance and chemokine levels in a mouse model of experimental traumatic brain injury. J Neuroinflammation 2024; 21:149. [PMID: 38840141 PMCID: PMC11155101 DOI: 10.1186/s12974-024-03129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Uncontrolled neuroinflammation mediates traumatic brain injury (TBI) pathology and impairs recovery. Interleukin-6 (IL-6), a pleiotropic inflammatory regulator, is associated with poor clinical TBI outcomes. IL-6 operates via classical-signaling through membrane-bound IL-6 receptor (IL-6R) and trans-signaling through soluble IL-6 receptor (s)IL-6R. IL-6 trans-signaling specifically contributes to neuropathology, making it a potential precision therapeutic TBI target. Soluble glycoprotein 130 (sgp130) prevents IL-6 trans-signaling, sparing classical signaling, thus is a possible treatment. Mice received either controlled cortical impact (CCI) (6.0 ± 0.2 m/s; 2 mm; 50-60ms) or sham procedures. Vehicle (VEH) or sgp130-Fc was subcutaneously administered to sham (VEH or 1 µg) and CCI (VEH, 0.25 µg or 1 µg) mice on days 1, 4, 7, 10 and 13 post-surgery to assess effects on cognition [Morris Water Maze (MWM)] and ipsilateral hemisphere IL-6 related biomarkers (day 21 post-surgery). CCI + sgp130-Fc groups (0.25 µg and 1 µg) were combined for analysis given similar behavior/biomarker outcomes. CCI + VEH mice had longer latencies and path lengths to the platform and increased peripheral zone time versus Sham + VEH and Sham + sgp130-Fc mice, suggesting injury-induced impairments in learning and anxiety. CCI + sgp130-Fc mice had shorter platform latencies and path lengths and had decreased peripheral zone time, indicating a therapeutic benefit of sgp130-Fc after injury on learning and anxiety. Interestingly, Sham + sgp130-Fc mice had shorter platform latencies, path lengths and peripheral zone times than Sham + VEH mice, suggesting a beneficial effect of sgp130-Fc, independent of injury. CCI + VEH mice had increased brain IL-6 and decreased sgp130 levels versus Sham + VEH and Sham + sgp130-Fc mice. There was no treatment effect on IL-6, sIL6-R or sgp130 in Sham + VEH versus Sham + sgp130-Fc mice. There was also no treatment effect on IL-6 in CCI + VEH versus CCI + sgp130-Fc mice. However, CCI + sgp130-Fc mice had increased sIL-6R and sgp130 versus CCI + VEH mice, demonstrating sgp130-Fc treatment effects on brain biomarkers. Inflammatory chemokines (MIP-1β, IP-10, MIG) were increased in CCI + VEH mice versus Sham + VEH and Sham + sgp130-Fc mice. However, CCI + sgp130-Fc mice had decreased chemokine levels versus CCI + VEH mice. IL-6 positively correlated, while sgp130 negatively correlated, with chemokine levels. Overall, we found that systemic sgp130-Fc treatment after CCI improved learning, decreased anxiety and reduced CCI-induced brain chemokines. Future studies will explore sex-specific dosing and treatment mechanisms for sgp130-Fc therapy.
Collapse
Affiliation(s)
- Ian G Gober
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Ashley L Russell
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Tyler J Shick
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Vincent A Vagni
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jenna C Carlson
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA, 15213, USA.
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA.
- Center for Neuroscience, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Zhang F, Clair AJ, Dankert JF, Lee YJ, Campbell KA, Kirsch T. Cytokine Receptor-like Factor 1 (CRLF1) and Its Role in Osteochondral Repair. Cells 2024; 13:757. [PMID: 38727293 PMCID: PMC11083199 DOI: 10.3390/cells13090757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Since cytokine receptor-like factor 1 (CRLF1) has been implicated in tissue regeneration, we hypothesized that CRLF1 released by mesenchymal stem cells can promote the repair of osteochondral defects. METHODS The degree of a femoral osteochondral defect repair in rabbits after intra-articular injections of bone marrow-derived mesenchymal stem cells (BMSCs) that were transduced with empty adeno-associated virus (AAV) or AAV containing CRLF1 was determined by morphological, histological, and micro computer tomography (CT) analyses. The effects of CRLF1 on chondrogenic differentiation of BMSCs or catabolic events of interleukin-1beta-treated chondrocyte cell line TC28a2 were determined by alcian blue staining, gene expression levels of cartilage and catabolic marker genes using real-time PCR analysis, and immunoblot analysis of Smad2/3 and STAT3 signaling. RESULTS Intra-articular injections of BMSCs overexpressing CRLF1 markedly improved repair of a rabbit femoral osteochondral defect. Overexpression of CRLF1 in BMSCs resulted in the release of a homodimeric CRLF1 complex that stimulated chondrogenic differentiation of BMSCs via enhancing Smad2/3 signaling, whereas the suppression of CRLF1 expression inhibited chondrogenic differentiation. In addition, CRLF1 inhibited catabolic events in TC28a2 cells cultured in an inflammatory environment, while a heterodimeric complex of CRLF1 and cardiotrophin-like Cytokine (CLC) stimulated catabolic events via STAT3 activation. CONCLUSION A homodimeric CRLF1 complex released by BMSCs enhanced the repair of osteochondral defects via the inhibition of catabolic events in chondrocytes and the stimulation of chondrogenic differentiation of precursor cells.
Collapse
Affiliation(s)
- Fenglin Zhang
- Department of Urology, New York University Grossman School of Medicine, New York, NY 10010, USA;
| | | | - John F. Dankert
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10010, USA; (J.F.D.); (Y.J.L.); (K.A.C.)
| | - You Jin Lee
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10010, USA; (J.F.D.); (Y.J.L.); (K.A.C.)
| | - Kirk A. Campbell
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10010, USA; (J.F.D.); (Y.J.L.); (K.A.C.)
| | - Thorsten Kirsch
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY 10010, USA; (J.F.D.); (Y.J.L.); (K.A.C.)
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, NY 10010, USA
| |
Collapse
|
14
|
Aarstad HH, Moe SEE, Lybak S, Bruserud Ø, Tvedt THA, Aarstad HJ. Plasma IL-1 and IL-6 Family Cytokines with Soluble Receptor Levels at Diagnosis in Head and Neck Squamous Cell Carcinoma: High Levels Predict Decreased Five-Year Disease-Specific and Overall Survival. Cancers (Basel) 2024; 16:1484. [PMID: 38672565 PMCID: PMC11048558 DOI: 10.3390/cancers16081484] [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: 03/21/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Activation of the acute-phase cascade (APC) has been correlated with outcomes in various cancers, including head and neck squamous cell carcinoma (HNSCC). Primary drivers of the APC are the cytokines within the interleukin-6 (IL-6) and IL-1 families. Plasma levels of IL-6 family cytokines/soluble receptors (IL-6, IL-27, IL-31, OSM, CNTF, soluble (s-)gp130, s-IL-6Rα) and IL-1 family members (IL-1RA, s-IL-33Rα) were determined at diagnosis for 87 human papillomavirus (HPV)-negative (-) HNSCC patients. We then studied the 5-year Disease-Specific Survival (DSS) and Overall Survival (OS). Increased plasma levels of IL-6 (p < 0.001/p < 0.001) (DSS/OS), IL-31 (p = 0.044/p = 0.07), IL-1RA (p = 0.004/p = 0.035), soluble (s)-IL-6Rα p = 0.022/p = 0.035), and s-gp130 (p = 0.007/p = 0.003) at diagnosis were predictors of both OS and DSS from HPV(-) HNSCC patients. The cytokine DSS/OS predictions were associated with TNM stage and smoking history, whereas the soluble receptors IL-6Rα, gp130, and IL33Rα more uniquely predicted DSS/OS. Clinically, IL-6 levels above 2.5 pg/mL yielded 75% specificity and 70% sensitivity for DSS. In conclusion, high plasma levels of IL-6, IL-31, and IL-1RA, as well as the soluble receptors IL-6Rα, gp130, and IL33Rα, predicted clinical outcome. This shows their potential as candidates for both general therapy and immune therapy stratification, as well as being future platforms for the development of new immunotherapy.
Collapse
Affiliation(s)
- Helene Hersvik Aarstad
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
- Department of Surgery, Haraldsplass Deaconal Hospital, 5009 Bergen, Norway
| | - Svein Erik Emblem Moe
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
| | - Stein Lybak
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5007 Bergen, Norway
| | - Øystein Bruserud
- Section for Haematology, Department of Medicine, Haukeland University Hospital, 5009 Bergen, Norway
| | | | - Hans Jørgen Aarstad
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5009 Bergen, Norway; (H.H.A.); (S.E.E.M.); (S.L.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5007 Bergen, Norway
| |
Collapse
|
15
|
Gardner S, Jin Y, Fyfe PK, Voisin TB, Bellón JS, Pohler E, Piehler J, Moraga I, Bubeck D. Structural insights into IL-11-mediated signalling and human IL6ST variant-associated immunodeficiency. Nat Commun 2024; 15:2071. [PMID: 38453915 PMCID: PMC10920896 DOI: 10.1038/s41467-024-46235-6] [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: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
IL-11 and IL-6 activate signalling via assembly of the cell surface receptor gp130; however, it is unclear how signals are transmitted across the membrane to instruct cellular responses. Here we solve the cryoEM structure of the IL-11 receptor recognition complex to discover how differences in gp130-binding interfaces may drive signalling outcomes. We explore how mutations in the IL6ST gene encoding for gp130, which cause severe immune deficiencies in humans, impair signalling without blocking cytokine binding. We use cryoEM to solve structures of both IL-11 and IL-6 complexes with a mutant form of gp130 associated with human disease. Together with molecular dynamics simulations, we show that the disease-associated variant led to an increase in flexibility including motion within the cytokine-binding core and increased distance between extracellular domains. However, these distances are minimized as the transmembrane helix exits the membrane, suggesting a stringency in geometry for signalling and dimmer switch mode of action.
Collapse
Affiliation(s)
- Scott Gardner
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK
| | - Yibo Jin
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK
| | - Paul K Fyfe
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Tomas B Voisin
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK
| | - Junel Sotolongo Bellón
- Department of Biology/Chemistry and Centre for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Elizabeth Pohler
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Jacob Piehler
- Department of Biology/Chemistry and Centre for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Ignacio Moraga
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Doryen Bubeck
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
16
|
Gaggioli MR, Jones AG, Panagi I, Washington EJ, Loney RE, Muench JH, Brennan RG, Thurston TLM, Ko DC. A single amino acid in the Salmonella effector SarA/SteE triggers supraphysiological activation of STAT3 for anti-inflammatory target gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580367. [PMID: 38405869 PMCID: PMC10888966 DOI: 10.1101/2024.02.14.580367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Non-typhoidal Salmonella enterica cause an estimated 1 million cases of gastroenteritis annually in the United States. These serovars use secreted protein effectors to mimic and reprogram host cellular functions. We previously discovered that the secreted effector SarA (Salmonella anti-inflammatory response activator; also known as SteE) was required for increased intracellular replication of S. Typhimurium and production of the anti-inflammatory cytokine interleukin-10 (IL-10). SarA facilitates phosphorylation of STAT3 through a region of homology with the host cytokine receptor gp130. Here, we demonstrate that a single amino acid difference between SarA and gp130 is critical for the anti-inflammatory bias of SarA-STAT3 signaling. An isoleucine at the pY+1 position of the YxxQ motif in SarA (which binds the SH2 domain in STAT3) causes increased STAT3 phosphorylation and expression of anti-inflammatory target genes. This isoleucine, completely conserved in ~4000 Salmonella isolates, renders SarA a better substrate for tyrosine phosphorylation by GSK-3. GSK-3 is canonically a serine/threonine kinase that nonetheless undergoes tyrosine autophosphorylation at a motif that has an invariant isoleucine at the pY+1 position. Our results provide a molecular basis for how a Salmonella secreted effector achieves supraphysiological levels of STAT3 activation to control host genes during infection.
Collapse
Affiliation(s)
- Margaret R. Gaggioli
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Angela G. Jones
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Ioanna Panagi
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Erica J. Washington
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Rachel E. Loney
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | | | - Richard G. Brennan
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Teresa L. M. Thurston
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, UK
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
- Lead contact
| |
Collapse
|
17
|
Zaporowska-Stachowiak I, Springer M, Stachowiak K, Oduah M, Sopata M, Wieczorowska-Tobis K, Bryl W. Interleukin-6 Family of Cytokines in Cancers. J Interferon Cytokine Res 2024; 44:45-59. [PMID: 38232478 DOI: 10.1089/jir.2023.0103] [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] [Indexed: 01/19/2024] Open
Abstract
Nine soluble ligands [interleukin-6 (IL-6), interleukin-11 (IL-11), leukemia inhibitory factor (LIF), oncostatin M (OSM), ciliary neurotrophic factor (CNTF), cardiotrophin-1 (CT-1), cardiotrophin-like cytokine, interleukin-27 (IL-27), and interleukin-31] share the ubiquitously expressed transmembrane protein-glycoprotein-130 beta-subunit (gp130) and thus form IL-6 family cytokines. Proteins that may be important for cancerogenesis, CT-1, IL-11, IL-27, LIF, OSM, and CNTF, belong to the superfamily of IL-6. Cytokines such as IL-6, IL-11, and IL-27 are better investigated in comparison with other members of the same family of cytokines, eg, CT-1. Gp130 is one of the main receptors through which these cytokines exert their effects. The clinical implication of understanding the pathways of these cytokines in oncology is that targeted therapy to inhibit or potentiate cytokine activity may lead to remission in some cases.
Collapse
Affiliation(s)
- Iwona Zaporowska-Stachowiak
- Department and Clinic of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Palliative Medicine In-Patient Unit, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Springer
- Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Mary Oduah
- English Students' Research Association, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Sopata
- Department and Clinic of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Palliative Medicine In-Patient Unit, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Wieczorowska-Tobis
- Department and Clinic of Palliative Medicine, Poznan University of Medical Sciences, Poznan, Poland
- Palliative Medicine In-Patient Unit, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, Poznan, Poland
| | - Wiesław Bryl
- Department of Internal Diseases, Metabolic Disorders and Arterial Hypertension, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
18
|
Widjaja AA, Cook SA. Nonspecific Inhibition of IL6 Family Cytokine Signalling by Soluble gp130. Int J Mol Sci 2024; 25:1363. [PMID: 38338642 PMCID: PMC10855816 DOI: 10.3390/ijms25031363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
IL6 is a proinflammatory cytokine that binds to membrane-bound IL6 receptor (IL6R) or soluble IL6R to signal via gp130 in cis or trans, respectively. We tested the hypothesis that sgp130Fc, which is believed to be a selective IL6 trans-signalling inhibitor, is in fact a non-specific inhibitor of gp130 signalling. In human cancer and primary cells, sgp130Fc inhibited IL6, IL11, OSM and CT1 cis-signalling. The IC50 values of sgp130Fc for IL6 and OSM cis-signalling were markedly (20- to 200-fold) lower than the concentrations of sgp130Fc used in mouse studies and clinical trials. sgp130 inhibited IL6 and OSM signalling in the presence of an ADAM10/17 inhibitor and the absence of soluble IL6R or OSMR, with effects that were indistinguishable from those of a gp130 neutralising antibody. These data show that sgp130Fc does not exclusively block IL6 trans-signalling and reveal instead that broad inhibition of gp130 signalling likely underlies its therapeutic effects. This proposes global or modular inhibition of gp130 as a therapeutic approach for treating human disease.
Collapse
Affiliation(s)
- Anissa A. Widjaja
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
| | - Stuart A. Cook
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857, Singapore
- National Heart Centre Singapore, National Heart Research Institute Singapore, Singapore 169609, Singapore
- MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London W6 8RF, UK
| |
Collapse
|
19
|
Ventelä J, Alanko A, Auvinen A, Lohi O, Nikkilä A. Dual direction associations between common autoimmune diseases and leukemia among children and young adults: A systematic review. Cancer Epidemiol 2023; 86:102411. [PMID: 37423102 DOI: 10.1016/j.canep.2023.102411] [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: 05/17/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Childhood leukemia and many autoimmune (AI) diseases are severe pediatric conditions with lifelong consequences. AI diseases form a heterogeneous disease group affecting about 5 % of children worldwide, while leukemia is the most common malignancy among children aged 0-14 years. The timing and similarities in suggested inflammatory and infectious triggers of AI disease and leukemia have raised a question whether the diseases share common etiological origins. We conducted a systematic review to evaluate the evidence linking childhood leukemia and AI diseases. DATA SOURCES In the systematic literature search CINAHL (from 1970), Cochrane Library (form 1981), PubMed (from 1926) and Scopus (from 1948) were queried in June 2023. REVIEW METHODS We included studies covering the association between any AI disease and acute leukemia, limiting it to children and adolescents under 25 years old. The studies were reviewed independently by two researchers and the risk of bias was assessed. RESULTS A total of 2119 articles were screened and 253 studies were selected for detailed evaluation. Nine studies met the inclusion criteria, of which eight were cohort studies and one was a systematic review. The diseases covered were type 1 diabetes mellitus, inflammatory bowel diseases and juvenile arthritis alongside acute leukemia. Five cohort studies were suitable for more detailed analysis: a rate ratio for leukemia diagnosis after any AI disease was 2.46 (95 % CI 1.17-5.18; heterogeneity I2 15 %) with a random-effects model. CONCLUSIONS The results of this systematic review indicate that AI diseases in childhood are associated with a moderately increased risk of leukemia. The association for individual AI diseases needs further investigation.
Collapse
Affiliation(s)
- Julia Ventelä
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Anni Alanko
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anssi Auvinen
- Faculty of Social Sciences, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Olli Lohi
- Tampere Center for Child, Adolescent, Maternal Health Research and Tays Cancer Center, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Atte Nikkilä
- Tampere Center for Child, Adolescent, Maternal Health Research and Tays Cancer Center, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
20
|
Krishna Swaroop A, Krishnan Namboori PK, Esakkimuthukumar M, Praveen TK, Nagarjuna P, Patnaik SK, Selvaraj J. Leveraging decagonal in-silico strategies for uncovering IL-6 inhibitors with precision. Comput Biol Med 2023; 163:107231. [PMID: 37421735 DOI: 10.1016/j.compbiomed.2023.107231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/27/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
Interleukin-6 upregulation leads to various acute phase reactions such as local inflammation and systemic inflammation in many diseases like cancer, multiple sclerosis, rheumatoid arthritis, anemia, and Alzheimer's disease stimulating JAK/STAT3, Ras/MAPK, PI3K-PKB/Akt pathogenic pathways. Since no small molecules are available in the market against IL-6 till now, we have designed a class of small bioactive 1,3 - indanedione (IDC) molecules for inhibiting IL-6 using a decagonal approach computational studies. The IL-6 mutations were mapped in the IL-6 protein (PDB ID: 1ALU) from thorough pharmacogenomic and proteomics studies. The protein-drug interaction networking analysis for 2637 FFDA-approved drugs with IL-6 protein using Cytoscape software showed that 14 drugs have prominent interactions with IL-6. Molecular docking studies showed that the designed compound IDC-24 (-11.8 kcal/mol) and methotrexate (-5.20) bound most strongly to the 1ALU south asian population mutated protein. MMGBSA results indicated that IDC-24 (-41.78 kcal/mol) and methotrexate (-36.81 kcal/mol) had the highest binding energy when compared to the standard molecules LMT-28 (-35.87 kcal/mol) and MDL-A (-26.18 kcal/mol). These results we substantiated by the molecular dynamic studies in which the compound IDC-24 and the methotrexate had the highest stability. Further, the MMPBSA computations produced energies of -28 kcal/mol and -14.69 kcal/mol for IDC-24 and LMT-28. KDeep absolute binding affinity computations revealed energies of -5.81 kcal/mol and -4.74 kcal/mol for IDC-24 and LMT-28 respectively. Finally, our decagonal approach established the compound IDC-24 from the designed 1,3-indanedione library and methotrexate from protein drug interaction networking as suitable HITs against IL-6.
Collapse
Affiliation(s)
- Akey Krishna Swaroop
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India
| | - P K Krishnan Namboori
- Amrita Molecular Modeling and Synthesis (AMMAS) Research Lab, Amrita Vishwavidyapeetham, Amrita Nagar, Ettimadai, Coimbatore, Tamilnadu, India
| | - M Esakkimuthukumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India
| | - T K Praveen
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India
| | - Palathoti Nagarjuna
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India
| | - Sunil Kumar Patnaik
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India
| | - Jubie Selvaraj
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamilnadu, India.
| |
Collapse
|
21
|
Casu A, Nunez Lopez YO, Yu G, Clifford C, Bilal A, Petrilli AM, Cornnell H, Carnero EA, Bhatheja A, Corbin KD, Iliuk A, Maahs DM, Pratley RE. The proteome and phosphoproteome of circulating extracellular vesicle-enriched preparations are associated with characteristic clinical features in type 1 diabetes. Front Endocrinol (Lausanne) 2023; 14:1219293. [PMID: 37576973 PMCID: PMC10417723 DOI: 10.3389/fendo.2023.1219293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/06/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction There are no validated clinical or laboratory biomarkers to identify and differentiate endotypes of type 1 diabetes (T1D) or the risk of progression to chronic complications. Extracellular vesicles (EVs) have been studied as biomarkers in several different disease states but have not been well studied in T1D. Methods As the initial step towards circulating biomarker identification in T1D, this pilot study aimed to provide an initial characterization of the proteomic and phosphoproteomic landscape of circulating EV-enriched preparations in participants with established T1D (N=10) and healthy normal volunteers (Controls) (N=7) (NCT03379792) carefully matched by age, race/ethnicity, sex, and BMI. EV-enriched preparations were obtained using EVtrap® technology. Proteins were identified and quantified by LC-MS analysis. Differential abundance and coexpression network (WGCNA), and pathway enrichment analyses were implemented. Results The detected proteins and phosphoproteins were enriched (75%) in exosomal proteins cataloged in the ExoCarta database. A total of 181 proteins and 8 phosphoproteins were differentially abundant in participants with T1D compared to controls, including some well-known EVproteins (i.e., CD63, RAB14, BSG, LAMP2, and EZR). Enrichment analyses of differentially abundant proteins and phosphoproteins of EV-enriched preparations identified associations with neutrophil, platelet, and immune response functions, as well as prion protein aggregation. Downregulated proteins were involved in MHC class II signaling and the regulation of monocyte differentiation. Potential key roles in T1D for C1q, plasminogen, IL6ST, CD40, HLA-DQB1, HLA-DRB1, CD74, NUCB1, and SAP, are highlighted. Remarkably, WGCNA uncovered two protein modules significantly associated with pancreas size, which may be implicated in the pathogenesis of T1D. Similarly, these modules showed significant enrichment for membrane compartments, processes associated with inflammation and the immune response, and regulation of viral processes, among others. Discussion This study demonstrates the potential of proteomic and phosphoproteomic signatures of EV-enriched preparations to provide insight into the pathobiology of T1D. The WGCNA analysis could be a powerful tool to discriminate signatures associated with different pathobiological components of the disease.
Collapse
Affiliation(s)
- Anna Casu
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | - Yury O. Nunez Lopez
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | - Gongxin Yu
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | - Christopher Clifford
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | - Anika Bilal
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | | | - Heather Cornnell
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | | | - Ananya Bhatheja
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | - Karen D. Corbin
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| | - Anton Iliuk
- Biomarker Discovery Department, Tymora Analytical Operations, West Lafayette, IN, United States
| | - David M. Maahs
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Richard E. Pratley
- AdventHealth, Translational Research Institute (TRI), Orlando, FL, United States
| |
Collapse
|
22
|
Zhao L, Li J, Mo G, Cao D, Li C, Huang G, Jiang L, Chen G, Yao H, Peng X. Recombinant protein EBI3 attenuates Clonorchis sinensis-induced liver fibrosis by inhibiting hepatic stellate cell activation in mice. Parasit Vectors 2023; 16:246. [PMID: 37480105 PMCID: PMC10360228 DOI: 10.1186/s13071-023-05863-5] [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: 03/01/2023] [Accepted: 07/03/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Chronic infection with Clonorchis sinensis can cause hepatobiliary fibrosis and even lead to hepatobiliary carcinoma. Epstein-Barr virus-induced gene 3 protein (EBI3) is a subunit of interleukin 35, which can regulate inflammatory response and the occurrence of fibrotic diseases. Previous studies have reported that the expression of EBI3 in the serum of patients with liver cirrhosis is reduced. The present study aims to investigate the biological effects of EBI3 on liver fibrosis caused by C. sinensis and the underlying molecular mechanisms. METHODS We first established a mouse model of liver fibrosis induced by C. sinensis infection and then measured the serum expression of EBI3 during the inflammatory and fibrotic phase. GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses were performed to investigate the potential role of EBI3 in liver fibrosis by regulating the extracellular matrix structural constituent and collagen catabolic process. Recombinant protein EBI3 (rEBI3) was added to hepatic stellate cells (HSCs) in vitro with C. sinensis antigen to explore its function. Finally, the therapeutic effect of rEBI3 was verified by intravenous injection into C. sinensis-infected mice. RESULTS The results showed that the serum expression of EBI3 increased in the inflammatory response phase but decreased in the fibrotic phase. The excretory-secretory products of C. sinensis (Cs.ESP) were able to stimulate HSC activation, while rEBI3 reduced the activation of HSCs induced by Cs.ESP. Also, the protein expression of gp130 and downstream protein expressions of JAK1, p-JAK1, STAT3 and p-STAT3 in HSCs were increased after rEBI3 incubation. Finally, intravenously injected rEBI3 inhibited hepatic epithelial-mesenchymal transition in C. sinensis-infected mice by inhibiting HSC activation and reducing liver injury. CONCLUSION This study confirms that rEBI3 can attenuate C. sinensis-induced liver fibrosis by inhibiting HSC activation and may be one of the potential treatments for liver fibrosis.
Collapse
Affiliation(s)
- Lei Zhao
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jia Li
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Gang Mo
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Deping Cao
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Chun Li
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Guoyang Huang
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Liping Jiang
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Gen Chen
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Hongbing Yao
- Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Xiaohong Peng
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China.
| |
Collapse
|
23
|
Graham R, Gazinska P, Zhang B, Khiabany A, Sinha S, Alaguthurai T, Flores-Borja F, Vicencio J, Beuron F, Roxanis I, Matkowski R, Liam-Or R, Tutt A, Ng T, Al-Jamal KT, Zhou Y, Irshad S. Serum-derived extracellular vesicles from breast cancer patients contribute to differential regulation of T-cell-mediated immune-escape mechanisms in breast cancer subtypes. Front Immunol 2023; 14:1204224. [PMID: 37441083 PMCID: PMC10335744 DOI: 10.3389/fimmu.2023.1204224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
Background Intracellular communication within the tumour is complex and extracellular vesicles (EVs) have been identified as major contributing factors for the cell-to-cell communication in the local and distant tumour environments. Here, we examine the differential effects of breast cancer (BC) subtype-specific patient serum and cell-line derived EVs in the regulation of T cell mediated immune responses. Methods Ultracentrifugation was used to isolate EVs from sera of 63 BC patients, 15 healthy volunteers and 4 human breast cancer cell lines. Longitudinal blood draws for EV isolation for patients on neoadjuvant chemotherapy was also performed. Characterization of EVs was performed by Nanoparticle Tracking Analysis (NTA), transmission electron microscopy (TEM) and immunoblotting. CD63 staining was performed on a tissue microarray of 218 BC patients. In-house bioinformatics algorithms were utilized for the computation of EV associated expression scores within The Cancer Genome Atlas (TCGA) and correlated with tumour infiltrating lymphocyte (TIL) scores. In vitro stimulation of PBMCs with EVs from serum and cell-line derived EVs was performed and changes in the immune phenotypes characterized by flow cytometry. Cytokine profiles were assessed using a 105-plex immunoassay or IL10 ELISA. Results Patients with triple negative breast cancers (TNBCs) exhibited the lowest number of EVs in the sera; whilst the highest was detected in ER+HER2+ cancers; reflected also in the higher level of CD63+ vesicles found within the ER+HER2+ local tumour microenvironment. Transcriptomic analysis of the TCGA data identified that samples assigned with lower EV scores had significantly higher abundance of CD4+ memory activated T cells, T follicular cells and CD8 T cells, plasma, and memory B cells; whilst samples with high EV scores were more enriched for anti-inflammatory M2 macrophages and mast cells. A negative correlation between EV expression scores and stromal TIL counts was also observed. In vitro experiments confirmed that circulating EVs within breast cancer subtypes have functionally differing immunomodulatory capabilities, with EVs from patients with the most aggressive breast cancer subtype (TNBCs) demonstrating the most immune-suppressive phenotype (decreased CD3+HLA-DR+ but increased CD3+PD-L1 T cells, increased CD4+CD127-CD25hi T regulatory cells with associated increase in IL10 cytokine production). In depth assessment of the cytokine modulation triggered by the serum/cell line derived exosomes confirmed differential inflammatory cytokine profiles across differing breast cancer subtypes. Studies using the MDA-231 TNBC breast cancer cell-line derived EVs provided further support that TNBC EVs induced the most immunosuppressive response within PBMCs. Discussion Our study supports further investigations into how tumour derived EVs are a mechanism that cancers can exploit to promote immune suppression; and breast cancer subtypes produce EVs with differing immunomodulatory capabilities. Understanding the intracellular/extracellular pathways implicated in alteration from active to suppressed immune state may provide a promising way forward for restoring immune competence in specific breast cancer patient populations.
Collapse
Affiliation(s)
- Rosalind Graham
- Breast Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Patrycja Gazinska
- Breast Cancer Now Research Unit, King's College London, Guy's Hospital, London, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
- Biobank Research Group, Lukasiewicz Research Network – PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Birong Zhang
- Systems Immunity University Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Atousa Khiabany
- Breast Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Shubhankar Sinha
- Breast Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Thanussuyah Alaguthurai
- Breast Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
- Breast Cancer Now Research Unit, King's College London, Guy's Hospital, London, United Kingdom
| | - Fabian Flores-Borja
- Richard Dimbleby Laboratory of Cancer Research School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Jose Vicencio
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, United Kingdom
| | - Fabienne Beuron
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Ioannis Roxanis
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Rafal Matkowski
- Breast Unit, Lower Silesian Oncology, Pulmunology and Hematology Center, Wroclaw, Poland
- Department of Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Revadee Liam-Or
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Andrew Tutt
- Breast Cancer Now Research Unit, King's College London, Guy's Hospital, London, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Tony Ng
- Breast Cancer Now Research Unit, King's College London, Guy's Hospital, London, United Kingdom
- Richard Dimbleby Laboratory of Cancer Research School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, United Kingdom
| | - Khuloud T. Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - You Zhou
- Systems Immunity University Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sheeba Irshad
- Breast Immunology Group, School of Cancer & Pharmaceutical Sciences, King’s College London, London, United Kingdom
- Breast Cancer Now Research Unit, King's College London, Guy's Hospital, London, United Kingdom
- Medical Oncology, Guy's & St Thomas' NHS Trust, London, United Kingdom
| |
Collapse
|
24
|
Sohrab SS, Raj R, Nagar A, Hawthorne S, Paiva-Santos AC, Kamal MA, El-Daly MM, Azhar EI, Sharma A. Chronic Inflammation's Transformation to Cancer: A Nanotherapeutic Paradigm. Molecules 2023; 28:molecules28114413. [PMID: 37298889 DOI: 10.3390/molecules28114413] [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: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The body's normal immune response against any invading pathogen that causes infection in the body results in inflammation. The sudden transformation in inflammation leads to the rise of inflammatory diseases such as chronic inflammatory bowel disease, autoimmune disorders, and colorectal cancer (different types of cancer develop at the site of chronic infection and inflammation). Inflammation results in two ways: short-term inflammation i.e., non-specific, involves the action of various immune cells; the other results in long-term reactions lasting for months or years. It is specific and causes angiogenesis, fibrosis, tissue destruction, and cancer progression at the site of inflammation. Cancer progression relies on the interaction between the host microenvironment and tumor cells along with the inflammatory responses, fibroblast, and vascular cells. The two pathways that have been identified connecting inflammation and cancer are the extrinsic and intrinsic pathways. Both have their own specific role in linking inflammation to cancer, involving various transcription factors such as Nuclear factor kappa B, Activator of transcription, Single transducer, and Hypoxia-inducible factor, which in turn regulates the inflammatory responses via Soluble mediators cytokines (such as Interleukin-6, Hematopoietin-1/Erythropoietin, and tumor necrosis factor), chemokines (such as Cyclooxygenase-2, C-X-C Motif chemokines ligand-8, and IL-8), inflammatory cells, cellular components (such as suppressor cells derived from myeloid, tumor-associated macrophage, and acidophils), and promotes tumorigenesis. The treatment of these chronic inflammatory diseases is challenging and needs early detection and diagnosis. Nanotechnology is a booming field nowadays for its rapid action and easy penetration inside the infected destined cells. Nanoparticles are widely classified into different categories based on their different factors and properties such as size, shape, cytotoxicity, and others. Nanoparticles emerged as excellent with highly progressive medical inventions to cure diseases such as cancer, inflammatory diseases, and others. Nanoparticles have shown higher binding capacity with the biomolecules in inflammation reduction and lowers the oxidative stress inside tissue/cells. In this review, we have overall discussed inflammatory pathways that link inflammation to cancer, major inflammatory diseases, and the potent action of nanoparticles in chronic inflammation-related diseases.
Collapse
Affiliation(s)
- Sayed Sartaj Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Riya Raj
- Department of Biochemistry, Bangalore University, Banglore 560056, India
| | - Amka Nagar
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida 201310, India
| | - Susan Hawthorne
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mohammad Amjad Kamal
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Enzymoics Inc., Hebersham, NSW 2770, Australia
- Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Mai M El-Daly
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ankur Sharma
- Strathclyde Institute of Pharmaceutical and Biomedical Sciences, University of Strathclyde, Glasgow G1 0RE, UK
| |
Collapse
|
25
|
Shin S, Awuah Boadi E, Shah S, Ezell M, Li P, Bandyopadhyay BC. Anti-inflammatory role of extracellular l-arginine through calcium sensing receptor in human renal proximal tubular epithelial (HK-2) cells. Int Immunopharmacol 2023; 117:109853. [PMID: 36827919 PMCID: PMC10124988 DOI: 10.1016/j.intimp.2023.109853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/24/2023]
Abstract
Renal tubular epithelial cells are capable of synthesizing interleukins (IL) in response to a variety of proinflammatory cytokines. Moreover, elevated urinary levels of IL have been shown in patients with various forms of nephritic diseases. However, the underlying intracellular signaling mechanism is unclear. Here we show the immunological signaling role of l-Arginine (l-Arg) through Ca2+-sensing receptor (CaSR) in human kidney 2 (HK-2) renal proximal tubular epithelial cells, using Ca2+ imaging and patch clamp techniques and its mechanistic link to the downstream cellular function. Both pharmacological and siRNA inhibitors support the activation CaSR by extracellular l-Arg to induced Ca2+ entry via a Transient receptor potential canonical (TRPC) channel in HK-2 cells mainly through the receptor operated Ca2+ entry (ROCE). Activation of CaSR by l-Arg led to the rise in p-p38/p38 expression suggesting [Ca2+]i as a regulator for p38-signaling pathways. Notably, l-Arg activated CaSR-induced Ca2+ signaling reduced the expressions of key fibrotic, inflammatory, and apoptotic genes, suggesting its nephroprotective role via Ca2+ signaling through CaSR in HK-2 cells. Since we found that the IL-6 expressions were inversely proportional to the increasing concentrations of l-Arg in HK-2 cells, we measured the release of IL-6, which steadily decreased as the concentrations of l-Arg were elevated. Taken together, extracellular l-Arg is a negative regulator for IL-6-induced inflammatory process, through the activation of CaSR and TRPC channel by ROCE pathway and can have a potential to alleviate inflammatory renal diseases.
Collapse
Affiliation(s)
- Samuel Shin
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Eugenia Awuah Boadi
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Saloni Shah
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Madison Ezell
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Peijun Li
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington, DC 20037, USA; Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064, USA.
| |
Collapse
|
26
|
Shkhyan R, Flynn C, Lamoure E, Sarkar A, Van Handel B, Li J, York J, Banks N, Van der Horst R, Liu NQ, Lee S, Bajaj P, Vadivel K, Harn HIC, Tassey J, Lozito T, Lieberman JR, Chuong CM, Hurtig MS, Evseenko D. Inhibition of a signaling modality within the gp130 receptor enhances tissue regeneration and mitigates osteoarthritis. Sci Transl Med 2023; 15:eabq2395. [PMID: 36947594 PMCID: PMC10792550 DOI: 10.1126/scitranslmed.abq2395] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 01/17/2023] [Indexed: 03/24/2023]
Abstract
Adult mammals are incapable of multitissue regeneration, and augmentation of this potential may shift current therapeutic paradigms. We found that a common co-receptor of interleukin 6 (IL-6) cytokines, glycoprotein 130 (gp130), serves as a major nexus integrating various context-specific signaling inputs to either promote regenerative outcomes or aggravate disease progression. Via genetic and pharmacological experiments in vitro and in vivo, we demonstrated that a signaling tyrosine 814 (Y814) within gp130 serves as a major cellular stress sensor. Mice with constitutively inactivated Y814 (F814) were resistant to surgically induced osteoarthritis as reflected by reduced loss of proteoglycans, reduced synovitis, and synovial fibrosis. The F814 mice also exhibited enhanced regenerative, not reparative, responses after wounding in the skin. In addition, pharmacological modulation of gp130 Y814 upstream of the SRC and MAPK circuit by a small molecule, R805, elicited a protective effect on tissues after injury. Topical administration of R805 on mouse skin wounds resulted in enhanced hair follicle neogenesis and dermal regeneration. Intra-articular administration of R805 to rats after medial meniscal tear and to canines after arthroscopic meniscal release markedly mitigated the appearance of osteoarthritis. Single-cell sequencing data demonstrated that genetic and pharmacological modulation of Y814 resulted in attenuation of inflammatory gene signature as visualized by the anti-inflammatory macrophage and nonpathological fibroblast subpopulations in the skin and joint tissue after injury. Together, our study characterized a molecular mechanism that, if manipulated, enhances the intrinsic regenerative capacity of tissues through suppression of a proinflammatory milieu and prevents pathological outcomes in injury and disease.
Collapse
Affiliation(s)
- Ruzanna Shkhyan
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Candace Flynn
- Ontario Veterinary College, Department of Clinical Studies, University of Guelph, ON N1G 2W1, Canada
| | - Emma Lamoure
- Ontario Veterinary College, Department of Clinical Studies, University of Guelph, ON N1G 2W1, Canada
| | - Arijita Sarkar
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Benjamin Van Handel
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Jinxiu Li
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Jesse York
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Nicholas Banks
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Robert Van der Horst
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Nancy Q. Liu
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Siyoung Lee
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Paul Bajaj
- UCLA Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | - Kanagasabai Vadivel
- UCLA Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | - Hans I.-Chen Harn
- Department of Pathology, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
- International Research Center of Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan 701401 Taiwan
| | - Jade Tassey
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Thomas Lozito
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Mark S. Hurtig
- Ontario Veterinary College, Department of Clinical Studies, University of Guelph, ON N1G 2W1, Canada
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
- Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA 90033, USA
| |
Collapse
|
27
|
Xu F, Wang S, Wang Y, Hu L, Zhu L. Inhibition of gp130 alleviates LPS-induced lung injury by attenuating apoptosis and inflammation through JAK1/STAT3 signaling pathway. Inflamm Res 2023; 72:493-507. [PMID: 36617342 DOI: 10.1007/s00011-022-01686-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/17/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Acute lung injury or acute respiratory distress syndrome (ALI/ARDS) is a life-threatening respiratory disease. Gp130 is a signal transduction receptor that participates in a variety of essential biological processes. The biological function of gp130 in ALI/ARDS is unclear. This study aims to investigate the roles and potential mechanisms of gp130 in lung injury induced by lipopolysaccharide (LPS). METHODS The ALI/ARDS mouse model was established using intratracheal LPS administration. Hematoxylin and eosin staining and bronchoalveolar lavage fluid analysis were used to evaluate the degree of lung injury. Cell apoptosis was assessed by TUNEL staining, flow cytometry, and western blot. Then the expression of gp130, IL-6, IL-10, TNF-α, and the JAK1/STAT3 signaling pathway-related proteins was assessed by RT-PCR, western blot, and immunohistochemistry. RESULTS The expression of gp130 increased after 24 h of LPS treatment. Inhibiting gp130 improved inflammatory infiltration and alveolar collapsed, decreased IL-6 and TNF-α levels, raised IL-10 levels, and decreased cell apoptosis in LPS-induced mice. Meanwhile, suppressing gp130 reduced the inflammatory response and cell apoptosis in LPS-induced Beas-2B cells. Furthermore, p-JAK1 and p-STAT3 expressions were elevated after LPS stimulation and decreased following gp130 inhibition, suggesting that gp130 may regulate the JAK1/STAT3 signaling pathway in LPS-induced mice and Beas-2B cells. CONCLUSION The findings suggest that gp130 regulates the inflammatory response and cell apoptosis through the JAK1/STAT3 signaling pathway, thereby mitigating LPS-induced lung injury. Gp130 may be a potential therapeutic target for ALI/ARDS.
Collapse
Affiliation(s)
- Fan Xu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, People's Republic of China
| | - Sijiao Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, People's Republic of China
| | - Yali Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, People's Republic of China
| | - Lijuan Hu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, People's Republic of China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, People's Republic of China.
- Department of Pulmonary and Critical Care Medicine, Huadong Hospital Affiliated to Fudan University, 221 Yan An Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
28
|
Jabeen K, Javed A, Waris A, Shahzad S. Restoration of IL-11 and IL-15 cytokine production post calcium modulators and ROS treatment can assist viral clearance both in vitro and in vivo. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:176-182. [PMID: 36742132 PMCID: PMC9869883 DOI: 10.22038/ijbms.2022.65983.14536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/31/2022] [Indexed: 02/07/2023]
Abstract
Objectives Hepatitis B virus (HBV) infection alters the cytokines production to establish persistent infection. A reversion of cytokines back to their normal state can be a promising therapeutic approach to establish an optimal host immune response. Materials and Methods We investigated the alteration in expression of IL-15 and IL-11 after HBV infection in vitro and in vivo in PBMCs of 63 individuals divided into various HBV-infected patient groups. The mRNA expression was evaluated post-anti-oxidant and calcium modulators treatment by Real-time qPCR. Results In vitro mRNA expression of both cytokines, post-infection was down-regulated considerably. Interestingly, in line with in vitro results, both cytokines' in vivo expression was intensively down-regulated in chronic HBV-infected individuals rather than healthy controls. Both cytokines' expression was up-regulated in cases of recovery compared with the inactive carriers and chronic HBV-infected individuals. IL-15 mRNA expression was significantly up-regulated in both cell lines post EGTA and Ru360 treatment while a significant increase was observed in the HepAD38 cell line with NAC and BAPTA treatment. IL-11 mRNA expression was significantly up-regulated in the HepG2 cell line after all modulator treatments, whereas in the HepAd38 cell line it was observed after BAPTA treatment. Our results thus indicate that viral infection tends to down-regulate the expression of cytokines and an in vivo up-regulation is an indication of recovery. Conclusion Treatment of anti-oxidants and calcium modulators has resulted in the successful restoration of these cytokines thus pointing towards the use of calcium modulators to boost natural antiviral cytokine production.
Collapse
Affiliation(s)
- Kehkshan Jabeen
- Genomics Research Lab, Department of Biological Sciences, International Islamic University Islamabad, Islamabad, Pakistan
| | - Aneela Javed
- Healthcare Biotechnology, Atta-ur Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, Pakistan,Corresponding author: Aneela Javed. Healthcare Biotechnology, Atta-ur Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, Pakistan.
| | - Asim Waris
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, Pakistan
| | - Shaheen Shahzad
- Genomics Research Lab, Department of Biological Sciences, International Islamic University Islamabad, Islamabad, Pakistan
| |
Collapse
|
29
|
Joly JA, Vallée A, Bourdin B, Bourbonnais S, Patey N, Gaboury L, Théorêt Y, Decaluwe H. Combined IFN-γ and JAK inhibition to treat hemophagocytic lymphohistiocytosis in mice. J Allergy Clin Immunol 2023; 151:247-259.e7. [PMID: 35973477 DOI: 10.1016/j.jaci.2022.07.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Familial hemophagocytic lymphohistiocytosis is a life-threatening hyperinflammatory disease caused by genetic defects in the granule-mediated cytotoxic pathway. Success of hematopoietic cell transplantation, the only cure, is correlated with the extent of disease control before transplantation. Unfortunately, disease refractoriness and toxicities to standard chemotherapy-based regimens are fatal in a fraction of patients. Novel targeted immunotherapies, such as IFN-γ blocking antibodies or ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor, are promising but only partially effective at controlling disease. OBJECTIVE We asked whether combinations of cytokine-targeted therapies, using antibodies or JAK inhibitor, work synergistically to counteract HLH. METHODS Genetically predisposed mice were infected and treated with distinct combinations of immunotherapies. Disease outcome was monitored and compared to monotherapies. RESULTS We showed that inhibiting IL-6 or IL-18 signaling in combination with IFN-γ blockade or ruxolitinib did not increase disease control compared to anti-IFN-γ antibodies or ruxolitinib monotherapies. In contrast, clinically relevant doses of ruxolitinib combined with low doses of anti-IFN-γ blocking antibodies corrected cytopenias, prevented overt neutrophilia, limited cytokinemia, and resolved HLH immunopathology and symptomatology. CONCLUSIONS Our findings demonstrate that IFN-γ blockade and ruxolitinib act synergistically to suppress HLH progression. This supports the use of combined cytokine-targeted therapies as a bridge to hematopoietic cell transplantation in severe familial hemophagocytic lymphohistiocytosis.
Collapse
Affiliation(s)
- Josée-Anne Joly
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Alexis Vallée
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Université de Montréal, Montréal, Québec, Canada; Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, Québec, Canada
| | - Benoîte Bourdin
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Sara Bourbonnais
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Natalie Patey
- Department of Pathology and Cellular Biology, Université de Montréal, Montréal, Québec, Canada
| | - Louis Gaboury
- Department of Pathology and Cellular Biology, Université de Montréal, Montréal, Québec, Canada; Histology and Molecular Pathology Research Unit, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, Québec, Canada
| | - Yves Théorêt
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada
| | - Hélène Decaluwe
- Cytokines and Adaptive Immunity Laboratory, Sainte-Justine University Hospital Research Center, Université de Montréal, Montréal, Québec, Canada; Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, Québec, Canada; Department of Pediatrics, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
30
|
Zysk W, Gleń J, Trzeciak M. Current Insight into the Role of IL-35 and Its Potential Involvement in the Pathogenesis and Therapy of Atopic Dermatitis. Int J Mol Sci 2022; 23:ijms232415709. [PMID: 36555351 PMCID: PMC9779445 DOI: 10.3390/ijms232415709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Interleukin 35 (IL-35), a new member of the IL-12 family of heterodimeric cytokines, could induce two different types of regulatory cells including regulatory T and B cells such as IL-35-induced regulatory T cells and IL-10-producing regulatory B cells (IL-10+Bregs), and IL-35-producing regulatory B cells (IL-35+Bregs). These cells appear to play an important role in modulating the immune system in numerous diseases. Several findings suggested that the expression of IL-35 is dysregulated in many autoimmune, inflammatory, and allergic diseases. Due to the functions of IL-35, it seems that this cytokine may act as an efficient therapeutic strategy for numerous conditions including atopic dermatitis (AD). We aimed to provide a comprehensive overview of the role of IL-35 in modulating the immune system. Additionally, we highlight IL-35 as a specific immunological target, discuss its possible involvement in the pathogenesis of AD, and hypothesize that IL-35 may become a novel target for the treatment of AD. However, further studies are required to evaluate this hypothesis.
Collapse
Affiliation(s)
- Weronika Zysk
- Dermatological Students Scientific Association, Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland
| | - Jolanta Gleń
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdańsk, Poland
- Correspondence: ; Tel.: +48-58-584-40-10
| |
Collapse
|
31
|
SRRM1 promotes the proliferation, migration, and invasion of hepatocellular carcinoma cells by regulating the JAK/STAT signaling pathway. Tissue Cell 2022; 79:101954. [DOI: 10.1016/j.tice.2022.101954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
|
32
|
Chidomere CI, Wahid M, Kemble S, Chadwick C, Thomas R, Hardy RS, McGettrick HM, Naylor AJ. Bench to Bedside: Modelling Inflammatory Arthritis. DISCOVERY IMMUNOLOGY 2022; 2:kyac010. [PMID: 38567064 PMCID: PMC10917191 DOI: 10.1093/discim/kyac010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 04/04/2024]
Abstract
Inflammatory arthritides such as rheumatoid arthritis are a major cause of disability. Pre-clinical murine models of inflammatory arthritis continue to be invaluable tools with which to identify and validate therapeutic targets and compounds. The models used are well-characterised and, whilst none truly recapitulates the human disease, they are crucial to researchers seeking to identify novel therapeutic targets and to test efficacy during preclinical trials of novel drug candidates. The arthritis parameters recorded during clinical trials and routine clinical patient care have been carefully standardised, allowing comparison between centres, trials, and treatments. Similar standardisation of scoring across in vivo models has not occurred, which makes interpretation of published results, and comparison between arthritis models, challenging. Here, we include a detailed and readily implementable arthritis scoring system, that increases the breadth of arthritis characteristics captured during experimental arthritis and supports responsive and adaptive monitoring of disease progression in murine models of inflammatory arthritis. In addition, we reference the wider ethical and experimental factors researchers should consider during the experimental design phase, with emphasis on the continued importance of replacement, reduction, and refinement of animal usage in arthritis research.
Collapse
Affiliation(s)
- Chiamaka I Chidomere
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Mussarat Wahid
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Samuel Kemble
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Caroline Chadwick
- Biomedical Services Unit, University of Birmingham, Birmingham, B15 2TT, UK
| | - Richard Thomas
- Biomedical Services Unit, University of Birmingham, Birmingham, B15 2TT, UK
| | - Rowan S Hardy
- Institute of Clinical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Helen M McGettrick
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Amy J Naylor
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
33
|
Mao L, Li YD, Chen RL, Li G, Zhou XX, Song F, Wu C, Hu Y, Hong YX, Dang X, Li GR, Wang Y. Heart-targeting exosomes from human cardiosphere-derived cells improve the therapeutic effect on cardiac hypertrophy. J Nanobiotechnology 2022; 20:435. [PMID: 36195937 PMCID: PMC9531502 DOI: 10.1186/s12951-022-01630-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/09/2022] [Indexed: 12/04/2022] Open
Abstract
Exosomes of human cardiosphere-derived cells (CDCs) are very promising for treating cardiovascular disorders. However, the current challenge is inconvenient delivery methods of exosomes for clinical application. The present study aims to explore the potential to enhance the therapeutic effect of exosome (EXO) from human CDCs to myocardial hypertrophy. A heart homing peptide (HHP) was displayed on the surface of exosomes derived from CDCs that were forced to express the HHP fused on the N-terminus of the lysosomal-associated membrane protein 2b (LAMP2b). The cardiomyocyte-targeting capability of exosomes were analyzed and their therapeutic effects were evaluated in a mouse model of myocardial hypertrophy induced by transverse aorta constriction (TAC). The molecular mechanisms of the therapeutic effects were dissected in angiotensin II-induced neonatal rat cardiomyocyte (NRCMs) hypertrophy model using a combination of biochemistry, immunohistochemistry and molecular biology techniques. We found that HHP-exosomes (HHP-EXO) accumulated more in mouse hearts after intravenous delivery and in cultured NRCMs than control exosomes (CON-EXO). Cardiac function of TAC mice was significantly improved with intravenous HHP-EXO administration. Left ventricular hypertrophy was reduced more by HHP-EXO than CON-EXO via inhibition of β-MHC, BNP, GP130, p-STAT3, p-ERK1/2, and p-AKT. Similar results were obtained in angiotensin II-induced hypertrophy of NRCMs, in which the beneficial effects of HHP-EXO were abolished by miRNA-148a inhibition. Our results indicate that HHP-EXO preferentially target the heart and improve the therapeutic effect of CDCs-exosomes on cardiac hypertrophy. The beneficial therapeutic effect is most likely attributed to miRNA-148a-mediated suppression of GP130, which in turn inhibits STAT3/ERK1/2/AKT signaling pathway, leading to improved cardiac function and remodeling.
Collapse
Affiliation(s)
- Liang Mao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Yun-Da Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Ruo-Lan Chen
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xiao-Xia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Fei Song
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yu Hu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xitong Dang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.,Nanjing Amaigh Pharma Limited, Nanjing, 210032, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
34
|
Antczak M, Cañete PF, Chen Z, Belle C, Yu D. Evolution of γ chain cytokines: Mechanisms, methods and applications. Comput Struct Biotechnol J 2022; 20:4746-4755. [PMID: 36147674 PMCID: PMC9465101 DOI: 10.1016/j.csbj.2022.08.050] [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: 06/14/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/12/2022] Open
Abstract
The common γ chain family of cytokines and their receptors play fundamental roles in the immune system. Evolutionary studies of γ chain cytokines have elegantly illustrated how the immune system adapts to ever-changing environmental conditions. Indeed, these studies have revealed the uniqueness of cytokine evolution, which exhibits strong positive selection pressure needed to adapt to rapidly evolving threats whilst still conserving their receptor binding capabilities. In this review, we summarise the evolutionary mechanisms that gave rise to the characteristically diverse family of γ chain cytokines. We also speculate on the benefits of studying cytokine evolution, which may provide alternative ways to design novel cytokine therapeutic strategies. Additionally, we discuss current evolutionary models that elucidate the emergence of distinct cytokines (IL-4 and IL-13) and cytokine receptors (IL-2Rα and IL-15Rα). Finally, we address and reflect on the difficulties associated with evolutionary studies of rapidly evolving genes and describe a variety of computational methods that have revealed numerous aspects of cytokine evolution.
Collapse
Affiliation(s)
- Magdalena Antczak
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Pablo F. Cañete
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Zhian Chen
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Clémence Belle
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
35
|
Li J, Li YR, Glessner JT, Yang J, March ME, Kao C, Vaccaro CN, Bradfield JP, Li J, Mentch FD, Qu H, Qi X, Chang X, Hou C, Abrams DJ, Qiu H, Wei Z, Connolly JJ, Wang F, Snyder J, Flatø B, Thompson SD, Langefeld CD, Lie BA, Munro JE, Wise C, Sleiman PMA, Hakonarson H. Identification of Novel Loci Shared by Juvenile Idiopathic Arthritis Subtypes Through Integrative Genetic Analysis. Arthritis Rheumatol 2022; 74:1420-1429. [PMID: 35347896 PMCID: PMC9542075 DOI: 10.1002/art.42129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 03/11/2022] [Accepted: 03/25/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Juvenile idiopathic arthritis (JIA) is the most common chronic immune-mediated joint disease among children and encompasses a heterogeneous group of immune-mediated joint disorders classified into 7 subtypes according to clinical presentation. However, phenotype overlap and biologic evidence suggest a shared mechanistic basis between subtypes. This study was undertaken to systematically investigate shared genetic underpinnings of JIA subtypes. METHODS We performed a heterogeneity-sensitive genome-wide association study encompassing a total of 1,245 JIA cases (classified into 7 subtypes) and 9,250 controls, followed by fine-mapping of candidate causal variants at each genome-wide significant locus, functional annotation, and pathway and network analysis. We further identified candidate drug targets and drug repurposing opportunities by in silico analyses. RESULTS In addition to the major histocompatibility complex locus, we identified 15 genome-wide significant loci shared between at least 2 JIA subtypes, including 10 novel loci. Functional annotation indicated that candidate genes at these loci were expressed in diverse immune cell types. CONCLUSION This study identified novel genetic loci shared by JIA subtypes. Our findings identified candidate mechanisms underlying JIA subtypes and candidate targets with drug repurposing opportunities for JIA treatment.
Collapse
Affiliation(s)
- Jin Li
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Yun R. Li
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, City of Hope Comprehensive Cancer Center, Duarte, California, and Translational Genomics Research InstitutePhoenixArizona
| | | | - Jie Yang
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | | | - Charlly Kao
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | | | | | - Junyi Li
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Frank D. Mentch
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Hui‐Qi Qu
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Xiaohui Qi
- Department of Cell Biology, the Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical Sciences, Tianjin Medical UniversityTianjinChina
| | - Xiao Chang
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Cuiping Hou
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Debra J. Abrams
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Haijun Qiu
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Zhi Wei
- New Jersey Institute of TechnologyNewark
| | | | - Fengxiang Wang
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - James Snyder
- The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Berit Flatø
- Oslo University HospitalRikshospitaletOsloNorway
| | - Susan D. Thompson
- University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical CenterCincinnatiOhio
| | - Carl D. Langefeld
- Wake Forest University School of MedicineWinston‐SalemNorth Carolina
| | | | | | | | | | - Hakon Hakonarson
- The Children's Hospital of Philadelphia and University of PennsylvaniaPhiladelphia
| |
Collapse
|
36
|
FLUOROFENIDONE ATTENUATES PULMONARY INFLAMMATION AND FIBROSIS BY INHIBITING THE IL-11/MEK/ERK SIGNALING PATHWAY. Shock 2022; 58:137-146. [PMID: 36166195 DOI: 10.1097/shk.0000000000001960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Idiopathic pulmonary fibrosis is defined as a specific form of chronic, progressive fibrosing interstitial pneumonia of unknown cause. Interleukin (IL)-11 plays an important role in the pathogenesis of idiopathic pulmonary fibrosis. In this study, we explore whether a potential antifibrotic agent fluorofenidone (FD) exerts its anti-inflammatory and antifibrotic effects through suppressing activation of the IL-11/MEK/ERK signaling pathway in vivo and in vitro. Male C57BL/6 J mice were intratracheally injected with bleomycin or saline. Fluorofenidone was administered throughout the course of the experiment. Lung tissue sections were stained with hemotoxylin and eosin, and Masson trichrome. Cytokines were measured using the enzyme-linked immunosorbent assay. The α-smooth muscle actin (α-SMA), fibronectin, and collagen I were measured using immunohistochemistry, and the phosphorylated extracellular signal-regulated kinase, phosphorylated mitogen-activated protein kinase, IL-11RA, and gp130 were measured using Western blot. The RAW264.7 cells and the normal human lung fibroblasts were treated with IL-11 and/or FD, IL-11RA-siRNA, or MEK inhibitor. The expressions of phosphorylated extracellular signal-regulated kinase, phosphorylated mitogen-activated protein kinase, IL-11RA, gp130, α-SMA, fibronectin, and collagen I were measured using Western blot and/or real-time polymerase chain reaction, and the cytokines were measured using enzyme-linked immunosorbent assay. Results showed that FD markedly reduced the expressions of IL-8, IL-18, IL-11, monocyte chemotactic protein-1, α-SMA, fibronectin, and collagen I in mice lung tissues. In addition, FD attenuated IL-11-induced expressions of α-SMA, fibronectin, and collagen I and inhibited IL-11RA, gp130, and phosphorylation of the ERK and MEK protein expression, as well as reduced the expressions of IL-8, IL-18, and monocyte chemotactic protein-1 in vitro. This study demonstrated that FD attenuated bleomycin-induced pulmonary inflammation and fibrosis in mice by inhibiting the IL-11/MEK/ERK signaling pathway.
Collapse
|
37
|
Liu NQ, Chen S, Geng D, Lei J, Zhang J, Li L, Lin Y, Ouyang Y, Shkhyan R, Van Handel B, Bian F, Mkaratigwa T, Chai Y, Evseenko D. Local Drug-Induced Modulation of gp130 Receptor Signaling Delays Disease Progression in a Pig Model of Temporo-Mandibular Joint Osteoarthritis. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.937819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Temporomandibular joint disorders (TMJs) are a multifaceted group of chronic disorders characterized by stiffness in the jaw, limited jaw mobility and pain when opening or closing the mouth. TMJs are relatively common, with incidence rates in the range of 5–12%, with nearly twice as many women as men being affected. One of the primary causes of TMJs is a degenerative disease of joints, such as osteoarthritis (OA), characterized by progressive loss of cartilage which causes stiffness, swelling, and pain. Currently, there are no disease-modifying agents on the market for OA. We have recently discovered a small molecule, R805 acting as a modulator of glycoprotein 130 (gp130) receptor for IL-6 family of cytokines. R805 enables regenerative outputs of endogenous joint stem and progenitor cells through immunomodulation in the joint microenvironment by reducing the levels of destructive cytokines and supporting chondrocyte survival and anabolism. Extensive testing has shown R805 to be safe at doses far above the therapeutic level. Here, we have conducted a pivotal efficacy study in our newly-established pig model of TMJ post-traumatic OA. IA injection of R805 has shown a highly significant reduction of articular cartilage degeneration, reduced synovitis and degenerative changes in subchondral bone in the mandibular condyle compared to the vehicle-treated group. These data will support additional pre-clinical development of R805 as a first-in-class injectable therapeutic for TMJ osteoarthritis.
Collapse
|
38
|
Ni Y, Low JT, Silke J, O’Reilly LA. Digesting the Role of JAK-STAT and Cytokine Signaling in Oral and Gastric Cancers. Front Immunol 2022; 13:835997. [PMID: 35844493 PMCID: PMC9277720 DOI: 10.3389/fimmu.2022.835997] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
When small proteins such as cytokines bind to their associated receptors on the plasma membrane, they can activate multiple internal signaling cascades allowing information from one cell to affect another. Frequently the signaling cascade leads to a change in gene expression that can affect cell functions such as proliferation, differentiation and homeostasis. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) and the tumor necrosis factor receptor (TNFR) are the pivotal mechanisms employed for such communication. When deregulated, the JAK-STAT and the TNF receptor signaling pathways can induce chronic inflammatory phenotypes by promoting more cytokine production. Furthermore, these signaling pathways can promote replication, survival and metastasis of cancer cells. This review will summarize the essentials of the JAK/STAT and TNF signaling pathways and their regulation and the molecular mechanisms that lead to the dysregulation of the JAK-STAT pathway. The consequences of dysregulation, as ascertained from founding work in haematopoietic malignancies to more recent research in solid oral-gastrointestinal cancers, will also be discussed. Finally, this review will highlight the development and future of therapeutic applications which modulate the JAK-STAT or the TNF signaling pathways in cancers.
Collapse
Affiliation(s)
- Yanhong Ni
- Central Laboratory, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jun T. Low
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - John Silke
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Lorraine A. O’Reilly
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
39
|
Amet R, Previtali V, Mihigo HB, Sheridan E, Brophy S, Hante NK, Santos-Martinez MJ, Hayden PJ, Browne PV, Rozas I, McElligott AM, Zisterer DM. A novel aryl-guanidinium derivative, VP79s, targets the signal transducer and activator of transcription 3 signaling pathway, downregulates myeloid cell leukaemia-1 and exhibits preclinical activity against multiple myeloma. Life Sci 2021; 290:120236. [PMID: 34953891 DOI: 10.1016/j.lfs.2021.120236] [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: 10/14/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
AIMS We have recently described a novel guanidinium-based compound, VP79s, which induces cytotoxicity in various cancer cell lines. Here, we aim to investigate the activity of VP79s and associated mechanisms of action in multiple myeloma (MM) cells in vitro and ex vivo. MAIN METHODS The effects of VP79s on cell viability and induction of apoptosis was examined in a panel of drug-sensitive and drug-resistant MM cell lines, as well as ex vivo patient samples and normal donor lymphocytes and platelets. Cell signaling pathways associated with the biological effects of VP79s were analysed by immunoblotting and flow cytometry. Gene expression changes were assessed by quantitative real-time PCR analysis. KEY FINDINGS VP79s was found to rapidly inhibit both constitutively active and IL-6-induced STAT3 signaling with concurrent downregulation of the IL-6 receptors, CD130 and CD126. VP79s induced a rapid and dose-dependent downregulation of anti-apoptotic Bcl-2 family member, myeloid cell leukaemia-1 (MCL-1). VP79s enhanced bortezomib induced cell death and was also found to overcome bone marrow stromal cell induced drug resistance. VP79s exhibited activity in ex vivo patient samples at concentrations which had no effect on peripheral blood mononuclear cells, lymphocytes and platelets isolated from healthy donors. SIGNIFICANCE As VP79s resulted in rapid inhibition of the key IL-6/STAT3 signaling pathway and downregulation of MCL-1 expression with subsequent selective anti-myeloma activity, VP79s may be a potential therapeutic agent with a novel mechanism of action in MM cells.
Collapse
Affiliation(s)
- Rebecca Amet
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland; John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Viola Previtali
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Helene B Mihigo
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Emily Sheridan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Sarah Brophy
- John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Nadhim Kamil Hante
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Maria Jose Santos-Martinez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Patrick J Hayden
- Department of Haematology, St. James's Hospital, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland
| | - Paul V Browne
- Department of Haematology, St. James's Hospital, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland
| | - Isabel Rozas
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Anthony M McElligott
- John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland.
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
40
|
Lee SY, Moon SJ, Moon YM, Seo HB, Ryu JG, Lee AR, Lee CR, Kim DS, Her YM, Choi JW, Kwok SK, Park SH, Cho ML. A novel cytokine consisting of the p40 and EBI3 subunits suppresses experimental autoimmune arthritis via reciprocal regulation of Th17 and Treg cells. Cell Mol Immunol 2021; 19:79-91. [PMID: 34782759 PMCID: PMC8752814 DOI: 10.1038/s41423-021-00798-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE The interleukin (IL)-12 cytokine family is closely related to the development of T helper cells, which are responsible for autoimmune disease enhancement or suppression. IL-12 family members are generally heterodimers and share three α-subunits (p35, p19, and p28) and two β-subunits (p40 and EBI3). However, a β-sheet p40 homodimer has been shown to exist and antagonize IL-12 and IL-23 signaling 1. Therefore, we assumed the existence of a p40-EBI3 heterodimer in nature and sought to investigate its role in immune regulation. METHODS The presence of the p40-EBI3 heterodimer was confirmed by ELISA, immunoprecipitation, and western blotting. A p40-EBI3 vector and p40-EBI3-Fc protein were synthesized to confirm the immunological role of this protein in mice with collagen-induced arthritis (CIA). The anti-inflammatory effects of p40-EBI3 were analyzed with regard to clinical, histological, and immune cell-regulating features in mice with CIA. RESULTS Clinical arthritis scores and the expression levels of proinflammatory cytokines (e.g., IL-17, IL-1β, IL-6, and TNF-α) were significantly attenuated in p40-EBI3-overexpressing and p40-EBI3-Fc-treated mice with CIA compared to vehicle-treated mice with CIA. Structural joint damage and vessel formation-related gene expression were also reduced by p40-EBI3 heterodimer treatment. In vitro, the p40-EBI3-Fc protein significantly suppressed the differentiation of Th17 cells and reciprocally induced CD4+CD25+Foxp3+ (regulatory T) cells. p40-EBI3 also inhibited osteoclast formation in a concentration-dependent manner. CONCLUSION In this study, p40-EBI3 ameliorated proinflammatory conditions both in vivo and in vitro. We propose that p40-EBI3 is a novel anti-inflammatory cytokine involved in suppressing the immune response through the expansion of Treg cells and suppression of Th17 cells and osteoclastogenesis.
Collapse
Affiliation(s)
- Seon-Yeong Lee
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Young-Mee Moon
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Hyeon-Beom Seo
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Geol Ryu
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - A Ram Lee
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Chae Rim Lee
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Da-Som Kim
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Yang-Mi Her
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Choi
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea. .,Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
41
|
Martínez-Pérez C, Kay C, Meehan J, Gray M, Dixon JM, Turnbull AK. The IL6-like Cytokine Family: Role and Biomarker Potential in Breast Cancer. J Pers Med 2021; 11:1073. [PMID: 34834425 PMCID: PMC8624266 DOI: 10.3390/jpm11111073] [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: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
IL6-like cytokines are a family of regulators with a complex, pleiotropic role in both the healthy organism, where they regulate immunity and homeostasis, and in different diseases, including cancer. Here we summarise how these cytokines exert their effect through the shared signal transducer IL6ST (gp130) and we review the extensive evidence on the role that different members of this family play in breast cancer. Additionally, we discuss how the different cytokines, their related receptors and downstream effectors, as well as specific polymorphisms in these molecules, can serve as predictive or prognostic biomarkers with the potential for clinical application in breast cancer. Lastly, we also discuss how our increasing understanding of this complex signalling axis presents promising opportunities for the development or repurposing of therapeutic strategies against cancer and, specifically, breast neoplasms.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Charlene Kay
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - James Meehan
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Mark Gray
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - J. Michael Dixon
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
| | - Arran K. Turnbull
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| |
Collapse
|
42
|
Christianson J, Oxford JT, Jorcyk CL. Emerging Perspectives on Leukemia Inhibitory Factor and its Receptor in Cancer. Front Oncol 2021; 11:693724. [PMID: 34395259 PMCID: PMC8358831 DOI: 10.3389/fonc.2021.693724] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/13/2021] [Indexed: 12/26/2022] Open
Abstract
Tumorigenesis and metastasis have deep connections to inflammation and inflammatory cytokines, but the mechanisms underlying these relationships are poorly understood. Leukemia Inhibitory Factor (LIF) and its receptor (LIFR), part of the interleukin-6 (IL-6) cytokine family, make up one such ill-defined piece of the puzzle connecting inflammation to cancer. Although other members of the IL-6 family have been shown to be involved in the metastasis of multiple types of cancer, the role of LIF and LIFR has been challenging to determine. Described by others in the past as enigmatic and paradoxical, LIF and LIFR are expressed in a diverse array of cells in the body, and the narrative surrounding them in cancer-related processes has been vague, and at times even contradictory. Despite this, recent insights into their functional roles in cancer have highlighted interesting patterns that may allude to a broader understanding of LIF and LIFR within tumor growth and metastasis. This review will discuss in depth the signaling pathways activated by LIF and LIFR specifically in the context of cancer-the purpose being to summarize recent literature concerning the downstream effects of LIF/LIFR signaling in a variety of cancer-related circumstances in an effort to begin teasing out the intricate web of contradictions that have made this pair so challenging to define.
Collapse
Affiliation(s)
- Joe Christianson
- Department of Biological Sciences, Boise State University, Boise, ID, United States
- Biomolecular Sciences Program, Boise State University, Boise, ID, United States
| | - Julia Thom Oxford
- Department of Biological Sciences, Boise State University, Boise, ID, United States
- Biomolecular Sciences Program, Boise State University, Boise, ID, United States
| | - Cheryl L. Jorcyk
- Department of Biological Sciences, Boise State University, Boise, ID, United States
- Biomolecular Sciences Program, Boise State University, Boise, ID, United States
| |
Collapse
|
43
|
Martínez-Pérez C, Leung J, Kay C, Meehan J, Gray M, Dixon JM, Turnbull AK. The Signal Transducer IL6ST (gp130) as a Predictive and Prognostic Biomarker in Breast Cancer. J Pers Med 2021; 11:618. [PMID: 34210062 PMCID: PMC8304290 DOI: 10.3390/jpm11070618] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/23/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
Novel biomarkers are needed to continue to improve breast cancer clinical management and outcome. IL6-like cytokines, whose pleiotropic functions include roles in many hallmarks of malignancy, rely on the signal transducer IL6ST (gp130) for all their signalling. To date, 10 separate independent studies based on the analysis of clinical breast cancer samples have identified IL6ST as a predictor. Consistent findings suggest that IL6ST is a positive prognostic factor and is associated with ER status. Interestingly, these studies include 4 multigene signatures (EndoPredict, EER4, IRSN-23 and 42GC) that incorporate IL6ST to predict risk of recurrence or outcome from endocrine or chemotherapy. Here we review the existing evidence on the promising predictive and prognostic value of IL6ST. We also discuss how this potential could be further translated into clinical practice beyond the EndoPredict tool, which is already available in the clinic. The most promising route to further exploit IL6ST's promising predicting power will likely be through additional hybrid multifactor signatures that allow for more robust stratification of ER+ breast tumours into discrete groups with distinct outcomes, thus enabling greater refinement of the treatment-selection process.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.L.); (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.M.); (M.G.)
| | - Jess Leung
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.L.); (C.K.); (J.M.D.); (A.K.T.)
| | - Charlene Kay
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.L.); (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.M.); (M.G.)
| | - James Meehan
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.M.); (M.G.)
| | - Mark Gray
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.M.); (M.G.)
| | - J Michael Dixon
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.L.); (C.K.); (J.M.D.); (A.K.T.)
| | - Arran K Turnbull
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.L.); (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (J.M.); (M.G.)
| |
Collapse
|
44
|
Davies R, Williams J, Sime K, Jin HS, Thompson C, Jordan L, Lang D, Halcox JP, Ellins E, Jones GW, Jones SA, Rose-John S, Williams A, Choy E. The role of interleukin-6 trans-signalling on cardiovascular dysfunction in inflammatory arthritis. Rheumatology (Oxford) 2021; 60:2852-2861. [PMID: 33313793 PMCID: PMC8213430 DOI: 10.1093/rheumatology/keaa725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/19/2020] [Indexed: 12/27/2022] Open
Abstract
Objectives Cardiovascular (CV) mortality in RA patients is 50% higher than in the general population. There is increasing recognition that systemic inflammation is a major driver of this. IL-6 is implicated in cardiovascular disease (CVD) in the general population but its role in CVD in RA is undefined. Of the two modes of IL-6 signalling, trans-signalling is pro-inflammatory whereas classical signalling is linked with inflammation resolution. This study examines the role of IL-6 trans-signalling in CVD in a mouse model and patients with RA. Methods Myography determined the effect of IL-6 trans-signalling blockade, using sgp130Fc, on aortic constriction in murine collagen-induced arthritis. Serum CCL2 and sVCAM-1 as soluble biomarkers of sIL-6R trans-signalling were investigated in a human cross-sectional study. An observational longitudinal study investigated the association between these biomarkers and progression of subclinical atherosclerosis in early RA by measuring carotid intima-media thickness (CIMT). Results sgp130Fc reduced arthritis severity, serum CCL2 and sVCAM-1 and restored vascular function in collagen-induced arthritis (CIA). In established RA, sVCAM-1 correlated with the 28-joint DAS (DAS28) and CV risk. In early RA, baseline DAS28 was associated with CIMT change at 6 months. CIMT ‘rapid progressors’ at 12 months had higher baseline sVCAM-1, haemoglobin A1c, cholesterol:high-density lipoprotein cholesterol ratio and LDL cholesterol. Conclusions IL-6 trans-signalling plays a pivotal role in vascular dysfunction in CIA. In early RA, sVCAM-1 was associated with progression of subclinical atherosclerosis. Inflammation from RA onset in CVD-susceptible individuals may accelerate atherosclerosis. IL-6 trans-signalling blockade may be beneficial to RA patients and perhaps for atherosclerosis in the general population.
Collapse
Affiliation(s)
- Ruth Davies
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Jessica Williams
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Katie Sime
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Hyun-Sun Jin
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | | | - Lauren Jordan
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Derek Lang
- Division of Medical Education, Cardiff University School of Medicine, Cardiff, UK
| | | | | | - Gareth W Jones
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, UK
| | - Simon A Jones
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrecht Universität, Kiel, Germany
| | - Anwen Williams
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| | - Ernest Choy
- CREATE Centre, Division of Infection and Immunity, , Cardiff, UK
| |
Collapse
|
45
|
Wrona E, Potemski P, Sclafani F, Borowiec M. Leukemia Inhibitory Factor: A Potential Biomarker and Therapeutic Target in Pancreatic Cancer. Arch Immunol Ther Exp (Warsz) 2021; 69:2. [PMID: 33630157 PMCID: PMC7907038 DOI: 10.1007/s00005-021-00605-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/12/2021] [Indexed: 01/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive, treatment-resistant cancer. Five-year survival rate is about 9%, one of the lowest among all solid tumors. Such a poor outcome is partly due to the limited knowledge of tumor biology, and the resulting lack of effective treatment options and robust predictive biomarkers. The leukemia inhibitory factor (LIF) has recently emerged as a potential biomarker and therapeutic target for PDAC. Accumulating evidence has suggested that LIF plays a role in supporting cancer evolution as a regulator of cell differentiation, renewal and survival. Interestingly, it can be detected in the serum of PDAC patients at higher concentrations than healthy individuals, this supporting its potential value as diagnostic biomarker. Furthermore, preliminary data indicate that testing for LIF serum concentration or tissue expression may help with treatment response monitoring and prognostication. Finally, studies in PDAC mouse models have also shown that LIF may be a valuable therapeutic target, and first-in-human clinical trial is currently ongoing. This article aims to review the available data on the role of LIF in PDAC promotion, and to discuss the evidence supporting its potential role as a biomarker and target of effective anti-cancer therapy in this setting.
Collapse
Affiliation(s)
- Ewa Wrona
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland.
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland.
| | - Piotr Potemski
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Francesco Sclafani
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Maciej Borowiec
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
46
|
Sobkowiak P, Narożna B, Wojsyk-Banaszak I, Bręborowicz A, Szczepankiewicz A. Expression of proteins associated with airway fibrosis differs between children with allergic asthma and allergic rhinitis. Int J Immunopathol Pharmacol 2021; 35:2058738421990493. [PMID: 33626956 PMCID: PMC7922620 DOI: 10.1177/2058738421990493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Allergic rhinitis (AR) and allergic asthma (AA) exhibit similar inflammatory
response in the airways. However, the remodelling is more extensive in the lower
airways, suggesting that the inflammation itself is not sufficient for allergic
phenotype. We aimed to analyse whether the expression of selected 27
inflammatory and fibrosis-related proteins may be altered in AR and AA in the
paediatric population and whether the expression pattern is either similar (due
to the inflammation) or disease-specific (due to the remodelling). We analysed
80 paediatric subjects: 39 with AA, 21 with AR and 20 healthy children. The
diagnosis of AR and AA was based on clinical manifestation, lung function,
positive skin prick tests and increased immunoglobulin E levels. Serum levels of
selected inflammatory proteins were measured with custom Magnetic Luminex Assay.
Statistical analysis was performed in Statistica v.13. CCL2/MCP1, GM-CSF, gp130
and periostin concentrations were significantly lower, whereas IL-5 levels were
higher in AA compared to the control group. CD-40L, CHI3L1/YKL-40, EGF, GM-CSF
and periostin levels were significantly decreased in patients with AR than in
the control group. Comparison of AA and AR patients revealed significant changes
in CHI3L1/YKL-40 (P = 0.021), IL-5
(P = 0.036), periostin (P = 0.013) and VEGFα
(P = 0.046). Significantly altered proteins were good
predictors to distinguish between AA and AR (P < 0.001, OR
46.00, accuracy 88.57%). Our results suggest that the expression of four
fibrotic proteins was significantly altered between AA and AR, suggesting
possible differences in airway remodelling between upper and lower airways.
Collapse
Affiliation(s)
- Paulina Sobkowiak
- Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Beata Narożna
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Irena Wojsyk-Banaszak
- Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Bręborowicz
- Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Aleksandra Szczepankiewicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
47
|
Murphy-Schafer AR, Paust S. Divergent Mast Cell Responses Modulate Antiviral Immunity During Influenza Virus Infection. Front Cell Infect Microbiol 2021; 11:580679. [PMID: 33680987 PMCID: PMC7935524 DOI: 10.3389/fcimb.2021.580679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus (IAV) is a respiratory pathogen that infects millions of people each year. Both seasonal and pandemic strains of IAV are capable of causing severe respiratory disease with a high risk of respiratory failure and opportunistic secondary infection. A strong inflammatory cytokine response is a hallmark of severe IAV infection. The widespread tissue damage and edema in the lung during severe influenza is largely attributed to an overexuberant production of inflammatory cytokines and cell killing by resident and infiltrating leukocytes. Mast cells (MCs) are a sentinel hematopoietic cell type situated at mucosal sites, including the lung. Poised to react immediately upon detecting infection, MCs produce a vast array of immune modulating molecules, including inflammatory cytokines, chemokines, and proteases. As such, MCs have been implicated as a source of the immunopathology observed in severe influenza. However, a growing body of evidence indicates that MCs play an essential role not only in inducing an inflammatory response but in suppressing inflammation as well. MC-derived immune suppressive cytokines are essential to the resolution of a number of viral infections and other immune insults. Absence of MCs prolongs infection, exacerbates tissue damage, and contributes to dissemination of the pathogen to other tissues. Production of cytokines such as IL-10 and IL-6 by MCs is essential for mitigating the inflammation and tissue damage caused by innate and adaptive immune cells alike. The two opposing functions of MCs-one pro-inflammatory and one anti-inflammatory-distinguish MCs as master regulators of immunity at the site of infection. Amongst the first cells to respond to infection or injury, MCs persist for the duration of the infection, modulating the recruitment, activation, and eventual suppression of other immune cells. In this review, we will discuss the immune modulatory roles of MCs over the course of viral infection and propose that the immune suppressive mediators produced by MCs are vital to minimizing immunopathology during influenza infection.
Collapse
Affiliation(s)
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
48
|
Bustos M, Dubrot J, Martinez-Anso E, Larequi E, Castaño D, Palazon A, Belza I, Sanmamed MF, Perez-Gracia JL, Ortiz de Solorzano C, Alfaro C, Melero I. Cardiotrophin-1 determines liver engraftment of syngenic colon carcinoma cells through an immune system-mediated mechanism. Oncoimmunology 2021; 1:1527-1536. [PMID: 23264899 PMCID: PMC3525608 DOI: 10.4161/onci.22504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiotrophin-1 (CT-1/CTF1) is a member of the interleukin-6 (IL-6) family of cytokines that stimulates STAT-3 phosphorylation in cells bearing the cognate receptor. We report that Ctf1−/− mice (hereby referred to as CT-1−/− mice) are resistant to the hepatic engraftment of MC38 colon carcinoma cells, while these cells engraft normally in the mouse subcutaneous tissue. Tumor intake in the liver could be enhanced by the systemic delivery of a recombinant adenovirus encoding CT-1, which also partly rescued the resistance of CT-1−/− mice to the hepatic engraftment of MC38 cells. Moreover, systemic treatment of wild-type (WT) mice with a novel antibody-neutralizing mouse CT-1 also reduced engraftment of this model. Conversely, experiments with Panc02 pancreatic cancer and B16-OVA melanoma cells in CT-1−/− mice revealed rates of hepatic engraftment similar to those observed in WT mice. The mechanism whereby CT-1 renders the liver permissive for MC38 metastasis involves T lymphocytes and natural killer (NK) cells, as shown by selective depletion experiments and in genetically deficient mice. However, no obvious changes in the number or cell killing capacity of liver lymphocytes in CT-1−/− animals could be substantiated. These findings demonstrate that the seed and soil concept to understand metastasis can be locally influenced by cytokines as well as by the cellular immune system.
Collapse
Affiliation(s)
- Matilde Bustos
- Gene Therapy and Hepatology Unit; Center for Applied Medical Research; University Clinic of Navarra; Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Prairie E, Côté F, Tsakpinoglou M, Mina M, Quiniou C, Leimert K, Olson D, Chemtob S. The determinant role of IL-6 in the establishment of inflammation leading to spontaneous preterm birth. Cytokine Growth Factor Rev 2021; 59:118-130. [PMID: 33551331 DOI: 10.1016/j.cytogfr.2020.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022]
Abstract
Preterm birth (PTB) and its consequences are a major public health concern as preterm delivery is the main cause of mortality and morbidity at birth. There are many causes of PTB, but inflammation is undeniably associated with the process of premature childbirth and fetal injury. At present, treatments clinically available mostly involve attempt to arrest contractions (tocolytics) but do not directly address upstream maternal inflammation on development of the fetus. One of the possible solutions may lie in the modulation of inflammatory mediators. Of the many pro-inflammatory cytokines involved in the induction of PTB, IL-6 stands out for its pleiotropic effects and its involvement in both acute and chronic inflammation. Here, we provide a detailed review of the effects of IL-6 on the timing of childbirth, its occurrence during PTB and its indissociable roles with associated fetal tissue damage.
Collapse
Affiliation(s)
- Elizabeth Prairie
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - France Côté
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Marika Tsakpinoglou
- Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Michael Mina
- Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada.
| | - Kelycia Leimert
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - David Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada.
| |
Collapse
|
50
|
Ritter K, Rousseau J, Hölscher C. The Role of gp130 Cytokines in Tuberculosis. Cells 2020; 9:E2695. [PMID: 33334075 PMCID: PMC7765486 DOI: 10.3390/cells9122695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/01/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Protective immune responses to Mycobacterium tuberculosis (Mtb) infection substantially depend on a delicate balance within cytokine networks. Thus, immunosuppressive therapy by cytokine blockers, as successfully used in the management of various chronic inflammatory diseases, is often connected with an increased risk for tuberculosis (TB) reactivation. Hence, identification of alternative therapeutics which allow the treatment of inflammatory diseases without compromising anti-mycobacterial immunity remains an important issue. On the other hand, in the context of novel therapeutic approaches for the management of TB, host-directed adjunct therapies, which combine administration of antibiotics with immunomodulatory drugs, play an increasingly important role, particularly to reduce the duration of treatment. In both respects, cytokines/cytokine receptors related to the common receptor subunit gp130 may serve as promising target candidates. Within the gp130 cytokine family, interleukin (IL)-6, IL-11 and IL-27 are most explored in the context of TB. This review summarizes the differential roles of these cytokines in protection and immunopathology during Mtb infection and discusses potential therapeutic implementations with respect to the aforementioned approaches.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
| | - Jasmin Rousseau
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
| | - Christoph Hölscher
- Infection Immunology, Research Centre Borstel, D-23845 Borstel, Germany; (K.R.); (J.R.)
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, D-23845 Borstel, Germany
| |
Collapse
|