1
|
Li X, Hu Q, Xu T. Janus kinase inhibitors and the risk of infections: a network meta-analysis across disease indications. Expert Opin Drug Saf 2025:1-13. [PMID: 40324885 DOI: 10.1080/14740338.2025.2502037] [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: 11/16/2024] [Accepted: 03/17/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION To compare the risks of serious infections, herpes zoster (HZ), and opportunistic infections associated with Janus kinase (JAK) inhibitors versus placebo, tumor necrosis factor-α inhibitors (TNFi), methotrexate (MTX), and among different JAK inhibitors. METHODS We searched PubMed, Embase, Cochrane Library, and Web of Science databases from their inception until 23 January 2024. Network meta-analysis estimated odds ratios for infections using restricted maximum likelihood models. RESULTS Eighty randomized controlled trials were included with 40,460 patients. Part of JAK inhibitors including tofacitinib (5 mg [2.01; 95%CI, 1.25-3.23], 10 mg [1.84; 95%CI, 1.06-3.17]), baricitinib (4 mg [1.57; 95%CI, 1.05-2.35]), and upadacitinib (15 mg [1.55; 95%CI, 1.06-2.27], 30 mg [1.94; 95%CI, 1.26-2.98]), exhibited a significantly different risk of serious infections compared to placebo. Similarly, tofacitinib (10 mg), baricitinib (4 mg), upadacitinib (15 mg, 30 mg), abrocitinib (200 mg), and peficitinib (100 mg) showed a significantly different risk of HZ infection compared to placebo. Most JAK inhibitors didn't raise opportunistic infections risks vs. TNFi and MTX, and risks among JAK inhibitors weren't statistically significant. CONCLUSION Attention should be paid to JAK inhibitor's types, dosages, and it is important to be aware of the risk of serious infections and HZ infections. Future long-term studies should be conducted. PROSPERO CRD42024523067.
Collapse
Affiliation(s)
- Xiaoqi Li
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qiaozhi Hu
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ting Xu
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Sahoo A, Paidesetty SK, Panda M. Target-specific high-throughput screening of anti-inflammatory phytosteroids for autoimmune diseases: A molecular docking-dynamics simulation approach. Steroids 2025; 217:109601. [PMID: 40120839 DOI: 10.1016/j.steroids.2025.109601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Without proper pathophysiology and recommended therapy, synthetic steroids are widely used as a first-line option for the management of autoimmune diseases. However, their prolonged use often leads to severe side effects such as osteoporosis, hypertension, cardiovascular, gastrointestinal complications, etc. To search for potential and safer therapeutic options, the present study aims to explore the potency and drug-ability profiles of anti-inflammatory phytosteroids (PSs). In a target-specific approach, we have selected three key molecular targets: glucocorticoid receptor/GR (PDB ID: 4P6W), cyclooxygenase-2/COX2 (PDB ID: 5F1A), and inducible nitric oxide synthase/iNOS (PDB ID: 4NOS) for a docking study of 167) selected PSs. The drug-chemistry profiles (physicochemical, toxicity, pharmacokinetic, drug-ability, etc.) of PSs were also assessed using various bioinformatics and chemoinformatics tools. The above assessment suggested that withaminilide B (PS46) is a lead candidate with higher drug-ability properties. Further, the drug stability and kinetic behaviour of the lead with the GR target 'GR-withaminilide B' in comparison with the control drug, 'GR-triamcinolone acetonide' docking complex, were studied through molecular dynamics (MD) simulation at 200 nanosecond with free energy calculation (MM/PBSA). Overall findings suggested that PSs exhibit distinct drug-ability profiles based on their functional attachments with a steroidal core moiety, where withaminilide B is a lead PSs among all to be used as alternative/ complementary candidates expected with limited adverse effects. Further experimentation is essential before mainstream application, but the study provided a platform to select drug-able candidates with a higher chance of experimental success and accelerate the drug discovery process within limited resources.
Collapse
Affiliation(s)
- Alaka Sahoo
- Department of Skin & VD, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar 751003 Odisha, India; Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar 751003 Odisha, India
| | - Sudhir Kumar Paidesetty
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar 751003 Odisha, India
| | - Maitreyee Panda
- Department of Skin & VD, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar 751003 Odisha, India.
| |
Collapse
|
3
|
Li L, Liu J, Lu J, Wu J, Zhang X, Ma T, Wu X, Zhu Q, Chen Z, Tai Z. Interventions in cytokine signaling: novel horizons for psoriasis treatment. Front Immunol 2025; 16:1573905. [PMID: 40303401 PMCID: PMC12037536 DOI: 10.3389/fimmu.2025.1573905] [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: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Intricate interactions between immune cells and cytokines define psoriasis, a chronic inflammatory skin condition that is immunological-mediated. Cytokines, including interleukins (ILs), interferons (IFNs), tumor necrosis factors (TNFs), chemokines, and transforming growth factor-β (TGF-β), are essential for controlling cellular activity and immunological responses, maintaining homeostasis and contributing to the pathogenesis of psoriasis. These molecules modulate the immune microenvironment by either promoting or suppressing inflammation, which significantly impacts therapeutic outcomes. Recent research indicates that treatment strategies targeting cytokines and chemokines have significant potential, offering new approaches for regulating the immune system, inhibiting the progression of psoriasis, and reducing adverse effects of traditional therapies. This review consolidates current knowledge on cytokine and chemokine signaling pathways in psoriasis and examines their significance in treatment. Specific attention is given to cytokines like IL-17, IL-23, and TNF-α, underscoring the necessity for innovative therapies to modulate these pathways and address inflammatory processes. This review emphasizes the principal part of cytokines in the -pathological process of psoriasis and explores the challenges and opportunities they present for therapeutic intervention. Furthermore, we examine recent advancements in targeted therapies, with a particular focus on monoclonal antibodies, in ongoing research and clinical trials.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Tianyou Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Topical Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Khademi Z, Mottaghi-Dastjerdi N, Morad H, Sahebkar A. The role of CRISPR-Cas9 and CRISPR interference technologies in the treatment of autoimmune diseases. Autoimmun Rev 2025; 24:103816. [PMID: 40221070 DOI: 10.1016/j.autrev.2025.103816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Autoimmune disorders can be described as inappropriate immune responses directed against self-antigens, which account for substantial healthcare concerns around the world. Immunosuppression or immune modulation are the main therapeutic modalities for autoimmune disorders. These modalities, however, impair the ability of the immune system to fight against infections, thereby predisposing to opportunistic diseases. This review explores existing therapies for autoimmune disorders, highlighting their limitations and challenges. Additionally, it describes the potential of CRISPR-Cas9 technology as a novel therapeutic approach to address these challenges.
Collapse
Affiliation(s)
- Zahra Khademi
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Mottaghi-Dastjerdi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Morad
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Temur BZ, Timucin AC, Atik AE, Kocagoz T, Can O. Peptide-Based Regulation of TNF-α-Mediated Cytotoxicity. Biomolecules 2025; 15:559. [PMID: 40338229 PMCID: PMC12024540 DOI: 10.3390/biom15040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a pro-inflammatory cytokine associated with TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), which play important roles in several inflammatory diseases. There is a growing interest in developing alternative molecules that can be used as TNF blockers. In this study, we focused on TNF-α-, TNFR1-, and TNFR2-mimicking peptides to inhibit TNF-α receptor binding in various ways. Six peptides (OB1, OB2, OB5, OB6, OB7, and OB8) were developed to bind TNFR1, TNFR2, and TNF-α. OB1 and OB2 bound to TNF-α with lower Kd values of 300 and 46.7 nM, respectively, compared to previously published sequences. These synthetic peptides directly and indirectly inhibited TNF-α in vitro without cytotoxicity to L929 cells, and OB1 significantly inhibited apoptosis in the presence of hTNF-α. Peptides developed in this study may prove to be useful for therapeutic inhibition of TNF-α.
Collapse
MESH Headings
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
- Peptides/pharmacology
- Peptides/chemistry
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/chemistry
- Apoptosis/drug effects
- Animals
- Mice
- Humans
- Cell Line
- Protein Binding
Collapse
Affiliation(s)
- Betul Zehra Temur
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey; (B.Z.T.); (T.K.)
| | - Ahmet Can Timucin
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 İstanbul, Turkey;
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 İstanbul, Turkey
| | - Ahmet Emin Atik
- Turgut Ilaclari A.S., 41400 Kocaeli, Turkey;
- Department of Natural Sciences, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey
| | - Tanil Kocagoz
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey; (B.Z.T.); (T.K.)
- Departmen of Medical Microbiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey
| | - Ozge Can
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey
| |
Collapse
|
6
|
Jenkins BJ, Jenkins YR, Ponce-Garcia FM, Moscrop C, Perry IA, Hitchings MD, Uribe AH, Bernuzzi F, Eastham S, Cronin JG, Berisha A, Howell A, Davies J, Blagih J, Veale DJ, Davies LC, Niphakis M, Finlay DK, Sinclair LV, Cravatt BF, Hogan AE, Nathan JA, Fearon U, Sumpton D, Vande Voorde J, Dias do Vale G, McDonald JG, Jones GW, Pearson JA, Vincent EE, Jones N. ABHD11 inhibition drives sterol metabolism to modulate T cell effector function and alleviate autoimmunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.643996. [PMID: 40166327 PMCID: PMC11957007 DOI: 10.1101/2025.03.19.643996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chronic inflammation in autoimmunity is driven by T cell hyperactivation. This unregulated response to self is fuelled by heightened metabolic programmes, which offers a promising new direction to uncover novel treatment strategies. α/β-hydrolase domain-containing protein 11 (ABHD11) is a mitochondrial hydrolase that maintains the catalytic function of α-ketoglutarate dehydrogenase (α-KGDH), and its expression in CD4+ T cells has been linked to remission status in rheumatoid arthritis (RA). However, the importance of ABHD11 in regulating T cell metabolism and function - and thus, the downstream implication for autoimmunity - is yet to be explored. Here, we show that pharmacological inhibition of ABHD11 dampens cytokine production by human and mouse T cells. Mechanistically, the anti-inflammatory effects of ABHD11 inhibition are attributed to increased 24,25-epoxycholesterol (24,25-EC) biosynthesis and subsequent liver X receptor (LXR) activation, which arise from a compromised TCA cycle. The impaired cytokine profile established by ABHD11 inhibition is extended to two patient cohorts of autoimmunity. Importantly, using a murine model of accelerated type 1 diabetes (T1D), we show that targeting ABHD11 suppresses cytokine production in antigen-specific T cells and delays the onset of diabetes in vivo. Collectively, our work provides pre-clinical evidence that ABHD11 is an encouraging drug target in T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Benjamin J. Jenkins
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| | - Yasmin R. Jenkins
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| | | | - Chloe Moscrop
- Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - Iain A. Perry
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| | - Matthew D. Hitchings
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| | - Alejandro H. Uribe
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Federico Bernuzzi
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Simon Eastham
- Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - James G. Cronin
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| | - Ardena Berisha
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Alexandra Howell
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, CF14 4XN, UK
| | - Joanne Davies
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, CF14 4XN, UK
| | - Julianna Blagih
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- University of Montreal, Maisonneuve-Rosemont Hospital Research Centre, Montreal, 5414 Assomption Blvd, H1T 2M4, Canada
| | - Douglas J. Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent’s University Hospital, Dublin, Ireland
| | - Luke C. Davies
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| | - Micah Niphakis
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - David K. Finlay
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearce Street, Dublin, Ireland
| | - Linda V. Sinclair
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Benjamin F. Cravatt
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Andrew E. Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - James A. Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearce Street, Dublin, Ireland
| | - David Sumpton
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Johan Vande Voorde
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, UK
| | - Goncalo Dias do Vale
- Center for Human Nutrition, Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jeffrey G. McDonald
- Center for Human Nutrition, Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Gareth W. Jones
- Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| | - James A. Pearson
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, CF14 4XN, UK
| | - Emma E. Vincent
- School of Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
- Integrative Epidemiology Unit, School of Population Health Science, University of Bristol, Bristol, BS8 2BN, UK
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, UK
| |
Collapse
|
7
|
Zhang L, Guan C, Wang S, Pfeiffer N, Grus FH. Anti-TNFα and Anti-IL-1β Monoclonal Antibodies Preserve BV-2 Microglial Homeostasis Under Hypoxia by Mitigating Inflammatory Reactivity and ATF4/MAPK-Mediated Apoptosis. Antioxidants (Basel) 2025; 14:363. [PMID: 40338234 PMCID: PMC11939723 DOI: 10.3390/antiox14030363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 05/09/2025] Open
Abstract
The disruption of microglial homeostasis and cytokine release are critical for neuroinflammation post-injury and strongly implicated in retinal neurodegenerative diseases like glaucoma. This study examines microglial responses to chemical hypoxia induced by cobalt chloride (CoCl2) in BV-2 murine microglial cells, focusing on signaling pathways and proteomic alterations. We assessed the protective effects of monoclonal antibodies against TNFα and IL-1β. CoCl2 exposure led to decreased cell viability, reduced mitochondrial membrane potential, increased lactate dehydrogenase release, elevated reactive oxygen species generation, and activation of inflammatory pathways, including nitric oxide synthase (iNOS), STAT1, and NF-κB/NLRP3. These responses were significantly mitigated by treatment with anti-TNFα and anti-IL-1β, suggesting their dual role in reducing microglial damage and inhibiting inflammatory reactivity. Additionally, these treatments reduced apoptosis by modulating ATF4 and the p38 MAPK/caspase-3 pathways. Label-free quantitative mass spectrometry-based proteomics and Gene Ontology revealed that CoCl2 exposure led to the upregulation of proteins primarily involved in endoplasmic reticulum and catabolic processes, while downregulated proteins are associated with biosynthesis. Anti-TNFα and anti-IL-1β treatments partially restored the proteomic profile toward normalcy, with network analysis identifying heat shock protein family A member 8 (HSPA8) as a central mediator in recovery. These findings offer insights into the pathogenesis of hypoxic microglial impairment and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Linglin Zhang
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.Z.); (C.G.); (N.P.)
| | - Chaoqiang Guan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.Z.); (C.G.); (N.P.)
| | - Sudena Wang
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.Z.); (C.G.); (N.P.)
| | - Franz H. Grus
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.Z.); (C.G.); (N.P.)
| |
Collapse
|
8
|
Dobbs ER. Screening, immunotherapy and the future of type 1 diabetes care in children and young people. Nurs Child Young People 2025:e1548. [PMID: 40091459 DOI: 10.7748/ncyp.2025.e1548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 03/19/2025]
Abstract
Over the past decade there has been a drive towards prevention of type 1 diabetes (T1D), which has led to the development of screening programmes to identify individuals with early-stage disease. In the same period, clinical trials have been taking place on the use of immunotherapy in preventing T1D progression. These developments in screening and immunotherapy require care pathways that provide monitoring, information and support to children and young people with early-stage type 1 diabetes and their families. This article provides an overview of the developments in screening and immunotherapy and considers the implications for children and young people and their families and for the healthcare professionals involved in their care.
Collapse
Affiliation(s)
- Emily Rose Dobbs
- Whittington Hospital, Whittington Health NHS Trust, London, England
| |
Collapse
|
9
|
Jones MR, Jones J, Pandu P, Liu C, Carey CD, Falo LD, Albers KM. Neurturin GF Enhances the Acute Cytokine Response of Inflamed Skin. J Invest Dermatol 2025; 145:583-592. [PMID: 39122143 DOI: 10.1016/j.jid.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Epidermal keratinocytes, immune cells, and sensory nerves all contribute to immune balance and skin homeostasis. Keratinocyte's release of GFs, neuromodulators, and immune activators is particularly important because each can evoke local (skin) and systemic (ie, immune and neural) responses that can initiate and exacerbate skin pathophysiology. From studies of skin and neural GFs, we hypothesized that neurturin (Nrtn), a member of the GDNF family that is expressed in the skin, has particular importance in this process. In this study, we examine how elevation of Nrtn in skin keratinocytes impacts early cytokine expression in response to complete Freund's adjuvant-mediated inflammation. Nrtn-overexpressing mice and wild-type mice injected with Nrtn exhibit an enhanced level of TNFα and IL-1β cytokines in the skin, a response previously shown to support healing. In vitro assays suggest that one source of the Nrtn-induced TNFα increase is keratinocytes, which are shown to express Nrtn and mRNAs encoding the Nrtn receptors GFRα2, Ret, ITGB1, and NCAM. These findings support the contribution of keratinocyte-derived Nrtn as an autocrine/paracrine factor that acts as a first-line defense molecule that regulates the initial cytokine response to inflammatory challenge.
Collapse
Affiliation(s)
- Marsha Ritter Jones
- Department of Anesthesia & Perioperative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James Jones
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Prathyusha Pandu
- Rutgers University School of Medicine, East Brunswick, New Jersey, USA
| | - Chunyan Liu
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Cara D Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
10
|
Hilewitz D, Yacobovitz S, Reiss-Huss S, Matar M, Weintraub Y, Shouval DS, Pavlovsky L, Friedland R. Paradoxical psoriasiform skin eruption in paediatric patients with inflammatory bowel disease treated with tumour necrosis factor-α inhibitors. Clin Exp Dermatol 2025; 50:558-563. [PMID: 39400046 DOI: 10.1093/ced/llae432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Tumour necrosis factor α (TNF-α) inhibitor (TNFi)-induced psoriasiform eruptions are a well-known phenomenon among adults. However, data are limited regarding this reaction in children. OBJECTIVES To describe in paediatric patients with inflammatory bowel disease (IBD) the clinical characteristics of TNFi-induced psoriasiform eruptions and the outcomes of various therapeutic options. METHODS We reviewed the medical charts of paediatric patients (aged < 18 years) with IBD who developed TNFi-induced psoriasiform eruptions during 2006-2022. RESULTS Among 454 patients with IBD treated with TNFis, 58 (12.8%) were diagnosed with TNFi-induced psoriasiform eruptions, of whom 51 were included in the study. The female to male ratio was 1 : 1.3. The median age at skin eruption was 14.11 [interquartile range (IQR) 12.11-16.05] years. The median elapsed time to eruption appearance was 15.00 months (IQR 7.00-24.00) after initiation of the treatment. All 51 patients were treated with topical steroids and 17 (33%) needed systemic treatment (phototherapy, methotrexate or acitretin). Sixteen of 51 patients (31%) needed to stop TNFi treatment because of an intractable eruption. Female patients, patients with inflammatory alopecia and patients who were treated with methotrexate or phototherapy were more prone to stop TNFis. CONCLUSIONS TNFi-induced psoriasiform eruptions are common in paediatric patients with IBD. The eruption may appear months or even years after treatment initiation. Almost one-third of the described patients had to replace their treatment because of a recalcitrant cutaneous eruption. This indicates that a multidisciplinary approach is required.
Collapse
Affiliation(s)
- Daniel Hilewitz
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Yacobovitz
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shiran Reiss-Huss
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Pediatric Dermatology Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Manar Matar
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Yael Weintraub
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Dror S Shouval
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Lev Pavlovsky
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Division of Dermatology, Rabin Medical Center, Petah Tikva, Israel
| | - Rivka Friedland
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Pediatric Dermatology Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| |
Collapse
|
11
|
Bertin D, Aghzadi J, Balandraud N, Roman C, Serrero M, Desplat-Jégo S. Detection of antibodies to infliximab in routine care: a 4-year French retrospective study. Clin Exp Immunol 2025; 219:uxae122. [PMID: 39714327 PMCID: PMC11747995 DOI: 10.1093/cei/uxae122] [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: 10/26/2024] [Revised: 11/25/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024] Open
Abstract
Despite its wide use to treat various inflammatory diseases, infliximab becomes ineffective in some patients due to inadequate drug levels and production of anti-drug antibodies (ADA). The aim of this study was to compare the prevalence and ADA levels in a large cohort of patients. ADA and infliximab (IFX) through levels measured by enzyme-linked immunosorbent assay were collected from 505 patients within a period of 4 years. The results indicate that (i) 13.5% of patients produce ADA, (ii) male patients were more likely to produce ADA at levels above 10 000 ng/ml than female patients, (iii) ADA levels were lower when associated with immunosuppressant drugs, (iv) there was an inverse relationship between ADA presence and IFX detection, and (v) no correlation was observed between ADA levels and number of injections or brand of IFX administered. This study improves our understanding of the factors promoting IFX immunogenicity and highlights the need to develop personalized treatment strategies.
Collapse
Affiliation(s)
- Daniel Bertin
- Immunology Laboratory, Biogenopôle, Hôpital de la Timone, APHM, Marseille, France
- CNRS, INP, Institute of Neurophysiopathology, Aix-Marseille University, Marseille, France
| | - Jehanne Aghzadi
- CNRS, INP, Institute of Neurophysiopathology, Aix-Marseille University, Marseille, France
| | | | - Céline Roman
- Multidisciplinary Pediatrics Department, Hôpital de la Timone, APHM, Marseille, France
| | - Mélanie Serrero
- Gastroenterology Department, Hôpital Nord, APHM, Marseille, France
| | - Sophie Desplat-Jégo
- Immunology Laboratory, Biogenopôle, Hôpital de la Timone, APHM, Marseille, France
- CNRS, INP, Institute of Neurophysiopathology, Aix-Marseille University, Marseille, France
| |
Collapse
|
12
|
Liu R, Zhao E, Wang Y, Zuo H, Li L, Xia Q, He H. Silk-engineered bioactive nanoparticles for targeted alleviation of acute inflammatory disease via macrophage reprogramming. J Nanobiotechnology 2025; 23:11. [PMID: 39794773 PMCID: PMC11724578 DOI: 10.1186/s12951-024-03055-6] [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: 09/04/2024] [Accepted: 11/30/2024] [Indexed: 01/13/2025] Open
Abstract
Significant progress has been made in the development of potential therapies for diseases associated with inflammation and oxidative stress. Nevertheless, the availability of effective clinical treatments remains limited. Herein, we introduce a novel silk-based bioactive material, TPSF, developed by sequentially conjugating Tempol and phenylboronic acid pinacol ester to silk fibroin. This innovative reactive oxygen species (ROS) scavenging material not only effectively eliminates free radicals and hydrogen peroxide but also readily self-assembles into nanoparticle forms (TPSN). In vitro experiments have demonstrated that TPSN exhibits significant anti-inflammatory activities and cytoprotective effects against ROS-mediated damage. Consistently, in murine models of acute lung and kidney injury, TPSN outperforms the small-molecule antioxidant NAC, exhibiting superior therapeutic efficacy. Mechanistically, TPSN has the capability to reprogram M1-like macrophages toward an M2-like state. Importantly, biocompatibility assays confirm that TPSN has good safety profiles. Consequently, TPSN, characterized by its favorable protective effects and excellent biocompatibility, exhibits considerable promise as a therapeutic intervention for inflammation-related diseases. This innovative strategy, which incorporates multifunctional antioxidant components into the silk fibroin matrix, effectively addresses oxidative stress and acute inflammation. Furthermore, it highlights the potential of modified silk fibroin materials in the management and mitigation of inflammation-led tissue damage.
Collapse
Affiliation(s)
- Renfeng Liu
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Erkang Zhao
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Yejing Wang
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China.
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing Key Laboratory of Soft-Matter Material Manufacturing, Southwest University, Chongqing, 400715, China.
| | - Hua Zuo
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Lanlan Li
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Huawei He
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, Biological Science Research Center, Southwest University, Chongqing, 400715, China.
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Chongqing Key Laboratory of Soft-Matter Material Manufacturing, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
13
|
Akram F, Ali AM, Akhtar MT, Fatima T, Shabbir I, Ul Haq I. The journey of antibody-drug conjugates for revolutionizing cancer therapy: A review. Bioorg Med Chem 2025; 117:118010. [PMID: 39586174 DOI: 10.1016/j.bmc.2024.118010] [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: 09/07/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a powerful class of targeted cancer therapies that harness the specificity of monoclonal antibodies to deliver cytotoxic payloads directly to tumor cells, minimizing off-target effects. This review explores the advancements in ADC technologies, focusing on advancing next-generation ADCs with novel payloads, conjugation strategies, and enhanced pharmacokinetic profiles. In particular, we highlight innovative payloads, including microtubule inhibitors, spliceosome modulators, and RNA polymerase inhibitors, that offer new mechanisms of cytotoxicity beyond traditional apoptosis induction. Additionally, the introduction of sophisticated conjugation techniques, such as site-specific conjugation using engineered cysteines, enzymatic methods, and integration of non-natural amino acids, has greatly improved the homogeneity, efficacy, and safety of ADCs. Furthermore, the review delves into the mechanistic insights into ADC action, detailing the intracellular pathways that facilitate drug release and cell death, and discussing the significance of bioconjugation methods in optimizing drug-antibody ratios (DARs). The establishment of comprehensive databases like ADCdb, which catalog vital pharmacological and biological data for ADCs, is also explored as a critical resource for advancing ADC research and clinical application. Finally, the clinical landscape of ADCs is examined, with a focus on the evolution of FDA-approved ADCs, such as Gemtuzumab Ozogamicin and Trastuzumab Emtansine, as well as emerging candidates in ongoing trials. As ADCs continue to evolve, their potential to revolutionize cancer therapy remains immense, offering new hope for more effective and personalized treatment options. ADCs also offer a significant advancement in targeted cancer therapy by merging the specificity of monoclonal antibodies with cytotoxic potency of chemotherapeutic agents. Hence, this dual mechanism intensifies tumor selectivity while minimizing systemic toxicity, paving the way for more effective and safer cancer treatments.
Collapse
Affiliation(s)
- Fatima Akram
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan; Department of Biology, Saint Louis University, St. Louis, MO, USA.
| | - Amna Murrawat Ali
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Muhammad Tayyab Akhtar
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Taseer Fatima
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Ifrah Shabbir
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Ikram Ul Haq
- Dr. Ikram-ul-Haq Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan; Pakistan Academy of Sciences, Islamabad, Pakistan
| |
Collapse
|
14
|
Mallick S, Duttaroy AK, Bose B. A Snapshot of Cytokine Dynamics: A Fine Balance Between Health and Disease. J Cell Biochem 2025; 126:e30680. [PMID: 39668456 DOI: 10.1002/jcb.30680] [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: 06/24/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024]
Abstract
Health and disease are intricately intertwined and often determined by the delicate balance of biological processes. Cytokines, a family of small signalling molecules, are pivotal in maintaining this balance, ensuring the body's immune system functions optimally. In a healthy condition, cytokines act as potent mediators of immune responses. They orchestrate the activities of immune cells, coordinating their proliferation, differentiation, and migration. This intricate role of cytokine signalling enables the body to effectively combat infections, repair damaged tissues, and regulate inflammation. However, the delicate equilibrium of cytokine production is susceptible to disruption. Excessive or abnormal cytokine levels can lead to a cascade of pathological conditions, including autoimmune diseases, chronic inflammation, infections, allergies, and even cancer. Interestingly, from the bunch of cytokines, few cytokines play an essential role in maintaining the balance between normal physiological status and diseases. In this review, we have appraised key cytokines' potential role and feedback loops in augmenting the imbalances in the body's biological functions, presenting a critical link between inflammation and disease pathology. Moreover, we have also highlighted the significance of cytokines and their molecular interplay, particularly in the recent viral pandemic COVID-19 disease. Hence, understandings regarding the interplay between viral infection and cytokine responses are essential and fascinating for developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Sumit Mallick
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, India
| |
Collapse
|
15
|
Zaroon, Aslam S, Hafsa, Mustafa U, Fatima S, Bashir H. Interleukin in Immune-Mediated Diseases: An Updated Review. Mol Biotechnol 2024:10.1007/s12033-024-01347-8. [PMID: 39715931 DOI: 10.1007/s12033-024-01347-8] [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: 08/20/2024] [Accepted: 11/29/2024] [Indexed: 12/25/2024]
Abstract
The immune system comprises various regulators and effectors that elicit immune responses against various attacks on the body. The pathogenesis of autoimmune diseases is derived from the deregulated expression of cytokines, the major regulators of the immune system. Among cytokines, interleukins have a major influence on immune-mediated diseases. These interleukins initiate the immune response against healthy and normal cells of the body, resulting in immune-mediated disease. The major interleukins in this respect are IL-1, IL-3, IL-4, IL-6, IL-10 and IL-12 which cause immune responses such as excessive inflammation, loss of immune tolerance, altered T-cell differentiation, immune suppression dysfunction, and inflammatory cell recruitment. Systemic Lupus Erythematosus (SLE) is an autoimmune illness characterized by dysregulation of interleukins. These immune responses are the signs of diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis, type I diabetes, and multiple sclerosis.
Collapse
Affiliation(s)
- Zaroon
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Shakira Aslam
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hafsa
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Usama Mustafa
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Sana Fatima
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hamid Bashir
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
16
|
Lenzen S, Jörns A. Therapy concepts in type 1 diabetes mellitus treatment: disease modifying versus curative approaches. J Mol Med (Berl) 2024; 102:1451-1455. [PMID: 39420138 PMCID: PMC11579207 DOI: 10.1007/s00109-024-02494-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: 07/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
For many autoimmune diseases, including type 1 diabetes mellitus (T1DM), efforts have been made to modify the disease process through pharmacotherapy. The ultimate goal must be to develop therapies with curative potential by achieving an organ without signs of parenchymal cell destruction and without signs of immune cell infiltration. In the case of the pancreas, this means regenerated and well-preserved beta cells in the islets without activated infiltrating immune cells. Recent research has opened up the prospect of successful antibody combination therapy for autoimmune diabetes with curative potential. This goal cannot be achieved with monotherapies. The requirements for the implementation of such a therapy with curative potential for the benefit of patients with T1DM and LADA (latent autoimmune diabetes in adults) are considered.
Collapse
Affiliation(s)
- Sigurd Lenzen
- Institute of Experimental Diabetes Research, Hannover Medical School, 30625, Hannover, Germany.
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Gregersen L, Jessen PD, Lund HW, Overgaard SH, Hikmat Z, Ellingsen T, Kjeldsen J, Pedersen AK, Petersen SR, Jawhara M, Nexøe AB, Bygum A, Hvas CL, Dahlerup JF, Bergenheim FO, Glerup H, Henriksen RH, Guldmann T, Hvid L, Brodersen J, Munk HL, Pedersen N, Saboori S, Nielsen OH, Heitmann BL, Haldorsson TI, Christensen R, Andersen V. Impact of gluten intake on clinical outcomes in patients with chronic inflammatory diseases initiating biologics: Secondary analysis of the prospective multicentre BELIEVE cohort study. Scand J Immunol 2024; 100:e13409. [PMID: 39358910 DOI: 10.1111/sji.13409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024]
Abstract
Chronic inflammatory diseases (CIDs) pose a growing healthcare challenge, with a substantial proportion of patients showing inadequate response to biological treatment. There is renewed interest in dietary changes to optimize treatment regimens, with a growing body of evidence suggesting beneficial effects with adherence to a gluten-free diet. This study compared the likelihood of achieving clinical response to biological treatment after 14-16 weeks in patients with CID with high versus low-to-medium gluten intake. Secondary outcomes of interest included changes in disease activity, health-related quality of life and C-reactive protein. The study was a multicentre prospective cohort of 193 participants with a CID diagnosis (i.e. Crohn's Disease, Ulcerative Colitis, Rheumatoid Arthritis, Axial Spondyloarthritis, Psoriatic Arthritis or Psoriasis) who initiated biological treatment between 2017 and 2020. Participants were stratified based on their habitual gluten intake: the upper 33.3% (high gluten intake) and the remaining 66.6% (low-to-medium gluten intake). The proportion of patients achieving clinical response to biological treatment after 14-16 weeks was compared using logistic regression models. The median gluten intake differed significantly between groups (12.5 g/day vs. 5.9 g/day, standardized mean difference = 1.399). In total, 108 (56%) achieved clinical response to treatment, with no difference between 35 (55%) in the high gluten group and 73 (57%) in the medium-to-low gluten group (OR = 0.96 [0.51-1.79], p = 0.897). No differences were found with secondary outcomes. In conclusion, this study found no association between gluten intake and response to biological treatment in patients with CID.
Collapse
Affiliation(s)
- Laura Gregersen
- Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Pernille Dyhre Jessen
- Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Helene Wiencke Lund
- Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Silja Hvid Overgaard
- Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Zainab Hikmat
- Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Torkell Ellingsen
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Jens Kjeldsen
- Research Unit of Gastroenterology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Andreas Kristian Pedersen
- Department of Clinical Research, University Hospital of Southern Denmark, Aabenraa, Denmark
- Open Patient Data Exploration Network (OPEN), Odense, Denmark
| | - Sofie Ronja Petersen
- Department of Clinical Research, University Hospital of Southern Denmark, Aabenraa, Denmark
| | - Mohamad Jawhara
- Section of Upper Benign and Hernia Surgery (G3), Department of Surgery, Odense University Hospital, Svendborg, Denmark
| | - Anders Bathum Nexøe
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Anette Bygum
- Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Christian Lodberg Hvas
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Frederik Dahlerup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
- The Abdominal Center, Medical Section, Bispebjerg University Hospital, Copenhagen, Denmark
| | | | - Henning Glerup
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Rikke Holm Henriksen
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Tanja Guldmann
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Lone Hvid
- Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| | - Jacob Brodersen
- The Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Internal Medicine-Gastroenterology, Esbjerg & Grindsted Hospital, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Heidi Lausten Munk
- Department of Rheumatology, Odense University Hospital, Odense, Denmark
- Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Natalia Pedersen
- Department of Gastroenterology, Slagelse Regional Hospital, Slagelse, Denmark
| | - Sanaz Saboori
- Department of Gastroenterology, Slagelse Regional Hospital, Slagelse, Denmark
| | | | - Berit Lillenthal Heitmann
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Thorhallur Ingi Haldorsson
- Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Faculty of Food Science and Nutrition, University of Iceland, Reykjavik, Iceland
| | - Robin Christensen
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Vibeke Andersen
- Open Patient Data Exploration Network (OPEN), Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
18
|
Khelghati F, Rahmanian M, Eghbal E, Seghatoleslami ZS, Goudarzi M, Keramatinia A, Ong CWM, Goletti D, D'Ambrosio L, Centis R, Nasiri MJ, Migliori GB. Risk of tuberculosis disease in patients receiving TNF-α antagonist therapy: A meta-analysis of randomized controlled trials. New Microbes New Infect 2024; 62:101533. [PMID: 39639969 PMCID: PMC11617757 DOI: 10.1016/j.nmni.2024.101533] [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: 09/14/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Tuberculosis (TB) risk associated with tumor necrosis factor-alpha (TNF-α) antagonist therapy in patients with autoimmune diseases is a significant concern. This study aims to evaluate the risk of TB disease associated with TNF-α antagonist therapy. Methods An extensive search of PubMed/MEDLINE, EMBASE, and the Cochrane CENTRAL databases was conducted to identify randomized controlled trials (RCTs) assessing TB disease risk in patients receiving TNF-α antagonist therapy available until November 1, 2024. The pooled statistic used was the weighted odds ratio (OR) and a corresponding 95 % confidence interval (CI). Statistical analysis was performed using Comprehensive Meta-Analysis software, version 3.0 (Biostat Inc., Englewood, NJ, USA). Results Fifty-six RCTs, totaling 22,212 adult patients, met the specified eligibility criteria. Pooled analysis revealed an increased risk of TB disease associated with TNF-α antagonist therapy (OR 1.52, 95 % CI 1.03-2.26, p = 0.03). Subgroup analyses indicated a higher risk in patients with rheumatoid arthritis (RA) (OR 2.25, 95 % CI 1.13-4.45, p = 0.02), while no significant associations were found in patients with ankylosing spondylitis (AS) or psoriasis (Ps). Analyses by specific TNF-α antagonist drugs did not yield significant associations with risk of TB disease. Conclusion Our study highlights an increased risk of TB disease associated with TNF-α antagonist therapy, particularly in patients with RA. However, the absence of significant associations in AS or Ps patients suggests disease-specific variations in risk of TB disease. Further research is needed to elucidate the long-term safety profile of TNF-α antagonist drugs and their associations with risk of TB disease in different patient populations.
Collapse
Affiliation(s)
- Fatemeh Khelghati
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmanian
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elaheh Eghbal
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aliasghar Keramatinia
- Department of Community Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Catherine WM. Ong
- Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research National Institute for Infectious Diseases L. Spallanzani-IRCCS, Roma, Italy
| | | | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| |
Collapse
|
19
|
Sastre DE, Bournazos S, Du J, Boder EJ, Edgar JE, Azzam T, Sultana N, Huliciak M, Flowers M, Yoza L, Xu T, Chernova TA, Ravetch JV, Sundberg EJ. Potent efficacy of an IgG-specific endoglycosidase against IgG-mediated pathologies. Cell 2024; 187:6994-7007.e12. [PMID: 39437779 PMCID: PMC11606778 DOI: 10.1016/j.cell.2024.09.038] [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: 04/09/2024] [Revised: 08/09/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Endo-β-N-acetylglucosaminidases (ENGases) that specifically hydrolyze the Asn297-linked glycan on immunoglobulin G (IgG) antibodies, the major molecular determinant of fragment crystallizable (Fc) γ receptor (FcγR) binding, are exceedingly rare. All previously characterized IgG-specific ENGases are multi-domain proteins secreted as an immune evasion strategy by Streptococcus pyogenes strains. Here, using in silico analysis and mass spectrometry techniques, we identified a family of single-domain ENGases secreted by pathogenic corynebacterial species that exhibit strict specificity for IgG antibodies. By X-ray crystallographic and surface plasmon resonance analyses, we found that the most catalytically efficient IgG-specific ENGase family member recognizes both protein and glycan components of IgG. Employing in vivo models, we demonstrated the remarkable efficacy of this IgG-specific ENGase in mitigating numerous pathologies that rely on FcγR-mediated effector functions, including T and B lymphocyte depletion, autoimmune hemolytic anemia, and antibody-dependent enhancement of dengue disease, revealing its potential for treating and/or preventing a wide range of IgG-mediated diseases in humans.
Collapse
Affiliation(s)
- Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jonathan Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - E Josephine Boder
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Julia E Edgar
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Tala Azzam
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nazneen Sultana
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maros Huliciak
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria Flowers
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lea Yoza
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ting Xu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tatiana A Chernova
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
20
|
McGahon J, Woods S, D'Elia R, Roberts CW. Non-ionic surfactant vesicles exert anti-inflammatory effects through inhibition of NFκB. J Inflamm (Lond) 2024; 21:49. [PMID: 39593021 PMCID: PMC11590361 DOI: 10.1186/s12950-024-00419-5] [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: 02/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Inflammation can be an unwanted consequence or cause of debilitating diseases of infectious and non-infectious aetiologies. Current anti-inflammatory medications have several deficiencies including lack of specificity and undesirable side effects. Herein, the potential of non-ionic surfactant vesicles (NISV) comprised of monopalmityol glycerol, dicetyl phosphate and cholesterol) as an anti-inflammatory drug and their mode of action is investigated. NISV were able to inhibit LPS-induced IL-6 from BMD macrophages. The individual components of NISV, monopalmityol glycerol, dicetyl phosphate and cholesterol did not affect LPS induced IL-6 levels, proving that formulation of NISV is essential for their anti-inflammatory effects. Transcriptomic analyses showed NISV mediated down-regulation of transcripts for inflammatory mediators in LPS stimulated macrophages. Notably, NISV downregulate NF-κB transcripts in LPS stimulated macrophages. Measurement of inflammatory mediators by cytometric bead array validated a number of transcriptomic findings as NISV were found to inhibit LPS induced IL-6, IL-12, and multiple chemokines. Further investigation demonstrated that NISV inhibited Poly(I:C) or Pam3csk4 induced inflammatory mediators. This indicates that the effects of NISV are distal to both MyD88 and TRIF signalling. Overall, the data generated highlights the potential of NISV as an anti-inflammatory therapeutic.
Collapse
Affiliation(s)
- Jonathan McGahon
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
- School of Health and Life Sciences, University of West Scotland, Stephenson Place, Glasgow, Lanarkshire, G72 0LH, UK
| | - Riccardo D'Elia
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK
- Chemical Biological and Radiological Division, Dstl, Porton Down, Salisbury, SP4 0JQ, UK
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK.
| |
Collapse
|
21
|
Javaid N, Ahmad B, Patra MC, Choi S. Decoy peptides that inhibit TNF signaling by disrupting the TNF homotrimeric oligomer. FEBS J 2024; 291:4372-4391. [PMID: 39003565 DOI: 10.1111/febs.17220] [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: 03/26/2023] [Revised: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor necrosis factor (TNF) is a pro-inflammatory cytokine and its functional homotrimeric form interacts with the TNF receptor (TNFR) to activate downstream apoptotic, necroptotic, and inflammatory signaling pathways. Excessive activation of these pathways leads to various inflammatory diseases, which makes TNF a promising therapeutic target. Here, 12-mer peptides were selected from the interface of TNF-TNFR based upon their relative binding energies and were named 'TNF-inhibiting decoys' (TIDs). These decoy peptides inhibited TNF-mediated secretion of cytokines and cell death, as well as activation of downstream signaling effectors. Effective TIDs inhibited TNF signaling by disrupting the formation of TNF's functional homotrimeric form. Among derivatives of TIDs, TID3c showed slightly better efficacy in cell-based assays by disrupting TNF trimer formation. Moreover, TID3c oligomerized TNF to a high molecular weight configuration. In silico modeling and simulations revealed that TID3c and its parent peptide, TID3, form a stable complex with TNF through hydrogen bonds and electrostatic interactions, which makes them the promising lead to develop peptide-based anti-TNF therapeutics.
Collapse
Affiliation(s)
- Nasir Javaid
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Suwon, Korea
| | - Bilal Ahmad
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Suwon, Korea
| | | | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- S&K Therapeutics, Suwon, Korea
| |
Collapse
|
22
|
Pang L, Zhao Y, Xu Y, Gao C, Wang C, Yu X, Wang F, He K. Mechanisms Underlying the Therapeutic Effects of JianPiYiFei II Granules in Treating COPD Based on GEO Datasets, Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulations. BIOLOGY 2024; 13:711. [PMID: 39336138 PMCID: PMC11428342 DOI: 10.3390/biology13090711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND JianPiYiFei (JPYF) II granules are a Chinese medicine for the treatment of chronic obstructive pulmonary disease (COPD). However, the main components and underlying mechanisms of JPYF II granules are not well understood. This study aimed to elucidate the potential mechanism of JPYF II granules in the treatment of COPD using network pharmacology, molecular docking, and molecular dynamics simulation techniques. METHODS The active compounds and corresponding protein targets of the JPYF II granules were found using the TCMSP, ETCM, and Uniport databases, and a compound-target network was constructed using Cytoscape3.9.1. The COPD targets were searched for in GEO datasets and the OMIM and GeneCards databases. The intersection between the effective compound-related targets and disease-related targets was obtained, PPI networks were constructed, and GO and KEGG enrichment analyses were performed. Then, molecular docking analysis verified the results obtained using network pharmacology. Finally, the protein-compound complexes obtained from the molecular docking analysis were simulated using molecular dynamics (MD) simulations. RESULTS The network pharmacological results showed that quercetin, kaempferol, and stigmasterol are the main active compounds in JPYF II granules, and AKT1, IL-6, and TNF are key target proteins. The PI3K/AKT signaling pathway is a potential pathway through which the JPYF II granules affect COPD. The results of the molecular docking analysis suggested that quercetin, kaempferol, and stigmasterol have a good binding affinity with AKT1, IL-6, and TNF. The MD simulation results showed that TNF has a good binding affinity with the compounds. CONCLUSIONS This study identified the effective compounds, targets, and related underlying molecular mechanisms of JPYF II granules in the treatment of COPD through network pharmacology, molecular docking, and MD simulation techniques, which provides a reference for subsequent research on the treatment of COPD.
Collapse
Affiliation(s)
- Liyuan Pang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yongjuan Zhao
- Department of Pulmonary and Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Yang Xu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Chencheng Gao
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Chao Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xiao Yu
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
23
|
Konry T, Sulllivan M, Rozzo A, Ward A, Rao P, Soler-Ferran D, Greenberg S. Single Cell Droplet-Based Efficacy and Transcriptomic Analysis of a Novel Anti-KLRG1 Antibody for Elimination of Autoreactive T Cells. RESEARCH SQUARE 2024:rs.3.rs-4745216. [PMID: 39281869 PMCID: PMC11398585 DOI: 10.21203/rs.3.rs-4745216/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Progress in developing improvements in the treatment of autoimmune disease has been gradual, due to challenges presented by the nature of these conditions. Namely, the need to suppress a patient's immune response while maintaining the essential activity of the immune system in controlling disease. Targeted treatments to eliminate the autoreactive immune cells driving disease symptoms present a promising new option for major improvements in treatment efficacy and side effect management. Monoclonal antibody therapies can be applied to target autoreactive immune cells if the cells possess unique surface marker expression patterns. Killer cell lectin like receptor G1 (KLRG1) expression on autoreactive T cells presents an optimal target for this type of cell depleting antibody therapy. In this study, we apply a variety of in vitro screening methods to determine the efficacy of a novel anti-KLRG1 antibody at mediating specific natural killer (NK) cell mediated antibody-dependent cellular cytotoxicity (ADCC). The methods include single-cell droplet microfluidic techniques, allowing timelapse imaging and sorting based on cellular interactions. Included in this study is the development of a novel method of sorting cells using a droplet-sorting platform and a fluorescent calcium dye to separate cells based on CD16 recognition of cell-bound antibody. We applied this novel sorting method to visualize transcriptomic variation between NK cells that are or are not activated by binding the anti-KLRG1 antibody using RNA sequencing. The data in this study reveals a reliable and target-specific cytotoxicity of the cell depleting anti-KLRG1 antibody, and supports our droplet-sorting calcium assay as a novel method of sorting cells based on receptor activation.
Collapse
|
24
|
Larsen MGR, Overgaard SH, Petersen SR, Møllegaard KM, Munk HL, Nexøe AB, Glerup H, Guldmann T, Pedersen N, Saboori S, Dahlerup JF, Hvas CL, Andersen KW, Jawhara M, Haagen Nielsen O, Bergenheim FO, Brodersen JB, Bygum A, Ellingsen T, Kjeldsen J, Christensen R, Andersen V. Effects of smoking on clinical treatment outcomes amongst patients with chronic inflammatory diseases initiating biologics: secondary analyses of the prospective BELIEVE cohort study. Scand J Immunol 2024; 100:e13395. [PMID: 38973149 DOI: 10.1111/sji.13395] [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: 04/09/2024] [Revised: 06/05/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
The prevalence and disease burden of chronic inflammatory diseases (CIDs) are predicted to rise. Patients are commonly treated with biological agents, but the individual treatment responses vary, warranting further research into optimizing treatment strategies. This study aimed to compare the clinical treatment responses in patients with CIDs initiating biologic therapy based on smoking status, a notorious risk factor in CIDs. In this multicentre cohort study including 233 patients with a diagnosis of Crohn's disease, ulcerative colitis, rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis or psoriasis initiating biologic therapy, we compared treatment response rates after 14 to 16 weeks and secondary outcomes between smokers and non-smokers. We evaluated the contrast between groups using logistic regression models: (i) a "crude" model, only adjusted for the CID type, and (ii) an adjusted model (including sex and age). Among the 205 patients eligible for this study, 53 (26%) were smokers. The treatment response rate among smokers (n = 23 [43%]) was lower compared to the non-smoking CID population (n = 92 [61%]), corresponding to a "crude" OR of 0.51 (95% CI: [0.26;1.01]) while adjusting for sex and age resulted in consistent findings: 0.51 [0.26;1.02]. The contrast was apparently most prominent among the 38 RA patients, with significantly lower treatment response rates for smokers in both the "crude" and adjusted models (adjusted OR 0.13, [0.02;0.81]). Despite a significant risk of residual confounding, patients with CIDs (rheumatoid arthritis in particular) should be informed that smoking probably lowers the odds of responding sufficiently to biological therapy. Registration: Clinical.Trials.gov NCT03173144.
Collapse
Affiliation(s)
- Maja Graves Rosenkilde Larsen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Silja Hvid Overgaard
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Sofie Ronja Petersen
- Department of Clinical Research, University Hospital of Southern Denmark, Odense, Denmark
| | - Karen Mai Møllegaard
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Heidi Lausten Munk
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
- Center for Rheumatology and Spine Diseases, Copenhagen, Denmark
| | - Anders Bathum Nexøe
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Cancer and Inflammation Research, Odense University Hospital, Odense, Denmark
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Henning Glerup
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Tanja Guldmann
- University Research Clinic for Innovative Patient Pathways, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Natalia Pedersen
- Department of Gastroenterology, Slagelse Hospital, Slagelse, Denmark
| | - Sanaz Saboori
- Department of Gastroenterology, Slagelse Hospital, Slagelse, Denmark
| | - Jens Frederik Dahlerup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Christian Lodberg Hvas
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Karina Winther Andersen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Mohamad Jawhara
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Esbjerg and Grindsted Hospital, University of Southern Denmark, Esbjerg, Denmark
| | - Fredrik Olof Bergenheim
- Department of Gastroenterology, Esbjerg and Grindsted Hospital, University of Southern Denmark, Esbjerg, Denmark
| | - Jacob Broder Brodersen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Department of Surgery, University Hospital of Southern Denmark, Aabenraa, Denmark
| | - Anette Bygum
- Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Torkell Ellingsen
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Jens Kjeldsen
- Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
- Research Unit of Medical Gastroenterology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Robin Christensen
- Section for Biostatistics and Evidence-Based Research, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Research Unit of Rheumatology, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, Odense, Denmark
| | - Vibeke Andersen
- Department of Internal Medicine, Molecular Diagnostics and Clinical Research Unit, University Hospital of Southern Denmark, Aabenraa, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
25
|
Galla MS, Sharma N, Mishra P, Shankaraiah N. Recent insights of PROTAC developments in inflammation-mediated and autoimmune targets: a critical review. RSC Med Chem 2024; 15:2585-2600. [PMID: 39149114 PMCID: PMC11324044 DOI: 10.1039/d4md00142g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/08/2024] [Indexed: 08/17/2024] Open
Abstract
According to the mounting evidence in the literature, pro-inflammatory mediators/targets activate multiple signalling pathways to trigger illnesses that are ultimately responsible for acute pain, chronic inflammatory diseases, and several auto-immune disorders. Conventional drugs have been ruled out since proteolysis-targeting chimeras (PROTACs) are poised to overcome the limitations of traditional therapies. These heterobifunctional molecules help to degrade the targeted proteins of interest through ubiquitination. This review encompasses current and future aspects of PROTACs in inflammation-mediated and autoimmune targets. Different key points are highlighted and discussed, such as why PROTACs are preferred in this disease area, drawbacks and lessons learnt from the past, the role of linkers in establishing crucial degradation, in vitro findings, pharmacokinetics, in silico parameters, limitations of PROTACs in clinical settings, and future outcomes.
Collapse
Affiliation(s)
- Mary Sravani Galla
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Nitika Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Priyanka Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| |
Collapse
|
26
|
Arras P, Zimmermann J, Lipinski B, Valldorf B, Evers A, Elter D, Krah S, Doerner A, Guarnera E, Siegmund V, Kolmar H, Pekar L, Zielonka S. Bovine ultralong CDR-H3 derived knob paratopes elicit potent TNF-α neutralization and enable the generation of novel adalimumab-based antibody architectures with augmented features. Biol Chem 2024; 405:461-470. [PMID: 38373142 DOI: 10.1515/hsz-2023-0370] [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: 12/14/2023] [Accepted: 01/29/2024] [Indexed: 02/21/2024]
Abstract
In this work we have generated cattle-derived chimeric ultralong CDR-H3 antibodies targeting tumor necrosis factor α (TNF-α) via immunization and yeast surface display. We identified one particular ultralong CDR-H3 paratope that potently neutralized TNF-α. Interestingly, grafting of the knob architecture onto a peripheral loop of the CH3 domain of the Fc part of an IgG1 resulted in the generation of a TNF-α neutralizing Fc (Fcknob) that did not show any potency loss compared with the parental chimeric IgG format. Eventually, grafting this knob onto the CH3 region of adalimumab enabled the engineering of a novel TNF-α targeting antibody architecture displaying augmented TNF-α inhibition.
Collapse
Affiliation(s)
- Paul Arras
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
- Biomolecular Immunotherapy, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
- Targeted mRNA Delivery, Merck KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Jasmin Zimmermann
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Britta Lipinski
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
- Biomolecular Immunotherapy, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Bernhard Valldorf
- Targeted mRNA Delivery, Merck KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Andreas Evers
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Desislava Elter
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Simon Krah
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Achim Doerner
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Enrico Guarnera
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Vanessa Siegmund
- Early Protein Supply & Characterization, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Harald Kolmar
- Applied Biochemistry, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| | - Lukas Pekar
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
| | - Stefan Zielonka
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Frankfurter Straße 250, D-64293 Darmstadt, Germany
- Biomolecular Immunotherapy, Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Peter-Grünberg-Strasse 4, D-64287 Darmstadt, Germany
| |
Collapse
|
27
|
Bas TG, Duarte V. Biosimilars in the Era of Artificial Intelligence-International Regulations and the Use in Oncological Treatments. Pharmaceuticals (Basel) 2024; 17:925. [PMID: 39065775 PMCID: PMC11279612 DOI: 10.3390/ph17070925] [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: 05/16/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
This research is based on three fundamental aspects of successful biosimilar development in the challenging biopharmaceutical market. First, biosimilar regulations in eight selected countries: Japan, South Korea, the United States, Canada, Brazil, Argentina, Australia, and South Africa, represent the four continents. The regulatory aspects of the countries studied are analyzed, highlighting the challenges facing biosimilars, including their complex approval processes and the need for standardized regulatory guidelines. There is an inconsistency depending on whether the biosimilar is used in a developed or developing country. In the countries observed, biosimilars are considered excellent alternatives to patent-protected biological products for the treatment of chronic diseases. In the second aspect addressed, various analytical AI modeling methods (such as machine learning tools, reinforcement learning, supervised, unsupervised, and deep learning tools) were analyzed to observe patterns that lead to the prevalence of biosimilars used in cancer to model the behaviors of the most prominent active compounds with spectroscopy. Finally, an analysis of the use of active compounds of biosimilars used in cancer and approved by the FDA and EMA was proposed.
Collapse
Affiliation(s)
- Tomas Gabriel Bas
- Escuela de Ciencias Empresariales, Universidad Católica del Norte, Coquimbo 1781421, Chile;
| | | |
Collapse
|
28
|
Ki MR, Youn S, Kim DH, Pack SP. Natural Compounds for Preventing Age-Related Diseases and Cancers. Int J Mol Sci 2024; 25:7530. [PMID: 39062777 PMCID: PMC11276798 DOI: 10.3390/ijms25147530] [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: 05/04/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Aging is a multifaceted process influenced by hereditary factors, lifestyle, and environmental elements. As time progresses, the human body experiences degenerative changes in major functions. The external and internal signs of aging manifest in various ways, including skin dryness, wrinkles, musculoskeletal disorders, cardiovascular diseases, diabetes, neurodegenerative disorders, and cancer. Additionally, cancer, like aging, is a complex disease that arises from the accumulation of various genetic and epigenetic alterations. Circadian clock dysregulation has recently been identified as an important risk factor for aging and cancer development. Natural compounds and herbal medicines have gained significant attention for their potential in preventing age-related diseases and inhibiting cancer progression. These compounds demonstrate antioxidant, anti-inflammatory, anti-proliferative, pro-apoptotic, anti-metastatic, and anti-angiogenic effects as well as circadian clock regulation. This review explores age-related diseases, cancers, and the potential of specific natural compounds in targeting the key features of these conditions.
Collapse
Affiliation(s)
- Mi-Ran Ki
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea; (M.-R.K.); (S.Y.); (D.H.K.)
- Institute of Industrial Technology, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea
| | - Sol Youn
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea; (M.-R.K.); (S.Y.); (D.H.K.)
| | - Dong Hyun Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea; (M.-R.K.); (S.Y.); (D.H.K.)
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-Ro 2511, Sejong 30019, Republic of Korea; (M.-R.K.); (S.Y.); (D.H.K.)
| |
Collapse
|
29
|
Biji CA, Balde A, Nazeer RA. Anti-inflammatory peptide therapeutics and the role of sulphur containing amino acids (cysteine and methionine) in inflammation suppression: A review. Inflamm Res 2024; 73:1203-1221. [PMID: 38769154 DOI: 10.1007/s00011-024-01893-6] [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: 02/01/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Inflammation serves as our body's immune response to combat infections, pathogens, viruses, and external stimuli. Inflammation can be classified into two types: acute inflammation and chronic inflammation. Non-steroidal anti-inflammatory medications (NSAIDs) are used to treat both acute and chronic inflammatory disorders. However, these treatments have various side effects such as reduced healing efficiency, peptic ulcers, gastrointestinal toxicities, etc. METHOD: This review assesses the potential of anti-inflammatory peptides (AIPs) derived from various natural sources, such as algae, fungi, plants, animals, and marine organisms. Focusing on peptides rich in cysteines and methionine, sulphur-containing amino acids known for their role in suppression of inflammation. RESULT Due to their varied biological activity, ability to penetrate cells, and low cytotoxicity, bioactive peptides have garnered interest as possible therapeutic agents. The utilisation of AIPs has shown great potential in the treatment of disorders associated with inflammation. AIPs can be obtained from diverse natural sources such as algae, fungi, plants, and animals. Cysteine and methionine are sulphur-containing amino acids that aid in the elimination of free radicals, hence assisting in the treatment of inflammatory diseases. CONCLUSION This review specifically examines several sources of AIPs including peptides that contain numerous cysteines and methionine. In addition, the biological characteristics of these amino acids and advancements in peptide delivery are also discussed.
Collapse
Affiliation(s)
- Catherin Ann Biji
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamilnadu, India.
| |
Collapse
|
30
|
Muzammil K, Rayyani S, Abbas Sahib A, Gholizadeh O, Naji Sameer H, Jwad Kazem T, Badran Mohammed H, Ghafouri Kalajahi H, Zainul R, Yasamineh S. Recent Advances in Crimean-Congo Hemorrhagic Fever Virus Detection, Treatment, and Vaccination: Overview of Current Status and Challenges. Biol Proced Online 2024; 26:20. [PMID: 38926669 PMCID: PMC11201903 DOI: 10.1186/s12575-024-00244-3] [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: 02/29/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus, and zoonosis, and affects large regions of Asia, Southwestern and Southeastern Europe, and Africa. CCHFV can produce symptoms, including no specific clinical symptoms, mild to severe clinical symptoms, or deadly infections. Virus isolation attempts, antigen-capture enzyme-linked immunosorbent assay (ELISA), and reverse transcription polymerase chain reaction (RT-PCR) are all possible diagnostic tests for CCHFV. Furthermore, an efficient, quick, and cheap technology, including biosensors, must be designed and developed to detect CCHFV. The goal of this article is to offer an overview of modern laboratory tests available as well as other innovative detection methods such as biosensors for CCHFV, as well as the benefits and limits of the assays. Furthermore, confirmed cases of CCHF are managed with symptomatic assistance and general supportive care. This study examined the various treatment modalities, as well as their respective limitations and developments, including immunotherapy and antivirals. Recent biotechnology advancements and the availability of suitable animal models have accelerated the development of CCHF vaccines by a substantial margin. We examined a range of potential vaccines for CCHF in this research, comprising nucleic acid, viral particles, inactivated, and multi-epitope vaccines, as well as the present obstacles and developments in this field. Thus, the purpose of this review is to present a comprehensive summary of the endeavors dedicated to advancing various diagnostic, therapeutic, and preventive strategies for CCHF infection in anticipation of forthcoming hazards.
Collapse
Affiliation(s)
- Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Khamis Mushait Campus, Abha, 62561, Saudi Arabia
| | - Saba Rayyani
- Medical Faculty, University of Georgi, Tbilisi, Georgia
| | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Tareq Jwad Kazem
- Scientific Affairs Department, Al-Mustaqbal University, Hillah, Babylon, 51001, Iraq
| | - Haneen Badran Mohammed
- Optics techniques department, health and medical techniques college, Al-Noor University, Mosul, Iraq
| | | | - Rahadian Zainul
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Padang, Padang, Indonesia.
| | - Saman Yasamineh
- Center for Advanced Material Processing, Artificial Intelligence, and Biophysics Informatics (CAMPBIOTICS), Universitas Negeri Padang, Padang, Indonesia.
| |
Collapse
|
31
|
Al-Sofi RF, Bergmann MS, Nielsen CH, Andersen V, Skov L, Loft N. The Association between Genetics and Response to Treatment with Biologics in Patients with Psoriasis, Psoriatic Arthritis, Rheumatoid Arthritis, and Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis. Int J Mol Sci 2024; 25:5793. [PMID: 38891983 PMCID: PMC11171831 DOI: 10.3390/ijms25115793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Genetic biomarkers could potentially lower the risk of treatment failure in chronic inflammatory diseases (CID) like psoriasis, psoriatic arthritis (PsA), rheumatoid arthritis (RA), and inflammatory bowel disease (IBD). We performed a systematic review and meta-analysis assessing the association between single nucleotide polymorphisms (SNPs) and response to biologics. Odds ratio (OR) with 95% confidence interval (CI) meta-analyses were performed. In total, 185 studies examining 62,774 individuals were included. For the diseases combined, the minor allele of MYD88 (rs7744) was associated with good response to TNFi (OR: 1.24 [1.02-1.51], 6 studies, 3158 patients with psoriasis or RA) and the minor alleles of NLRP3 (rs4612666) (OR: 0.71 [0.58-0.87], 5 studies, 3819 patients with RA or IBD), TNF-308 (rs1800629) (OR: 0.71 [0.55-0.92], 25 studies, 4341 patients with psoriasis, RA, or IBD), FCGR3A (rs396991) (OR: 0.77 [0.65-0.93], 18 studies, 2562 patients with psoriasis, PsA, RA, or IBD), and TNF-238 (rs361525) (OR: 0.57 [0.34-0.96]), 7 studies, 818 patients with psoriasis, RA, or IBD) were associated with poor response to TNFi together or infliximab alone. Genetic variants in TNFα, NLRP3, MYD88, and FcRγ genes are associated with response to TNFi across several inflammatory diseases. Most other genetic variants associated with response were observed in a few studies, and further validation is needed.
Collapse
Affiliation(s)
- Rownaq Fares Al-Sofi
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Mie Siewertsen Bergmann
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Claus Henrik Nielsen
- Center for Rheumatology and Spine Diseases, Institute for Inflammation Research, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark
| | - Vibeke Andersen
- Institute of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark
- Molecular Diagnostics and Clinical Research Unit, Department of Internal Medicine, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Lone Skov
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Nikolai Loft
- Department of Dermatology and Allergy, Copenhagen University Hospital—Herlev and Gentofte, 1165 Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| |
Collapse
|
32
|
Chernikov IV, Bachkova IK, Sen’kova AV, Meschaninova MI, Savin IA, Vlassov VV, Zenkova MA, Chernolovskaya EL. Cholesterol-Modified Anti-Il6 siRNA Reduces the Severity of Acute Lung Injury in Mice. Cells 2024; 13:767. [PMID: 38727303 PMCID: PMC11083178 DOI: 10.3390/cells13090767] [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: 04/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Small interfering RNA (siRNA) holds significant therapeutic potential by silencing target genes through RNA interference. Current clinical applications of siRNA have been primarily limited to liver diseases, while achievements in delivery methods are expanding their applications to various organs, including the lungs. Cholesterol-conjugated siRNA emerges as a promising delivery approach due to its low toxicity and high efficiency. This study focuses on developing a cholesterol-conjugated anti-Il6 siRNA and the evaluation of its potency for the potential treatment of inflammatory diseases using the example of acute lung injury (ALI). The biological activities of different Il6-targeted siRNAs containing chemical modifications were evaluated in J774 cells in vitro. The lead cholesterol-conjugated anti-Il6 siRNA after intranasal instillation demonstrated dose-dependent therapeutic effects in a mouse model of ALI induced by lipopolysaccharide (LPS). The treatment significantly reduced Il6 mRNA levels, inflammatory cell infiltration, and the severity of lung inflammation. IL6 silencing by cholesterol-conjugated siRNA proves to be a promising strategy for treating inflammatory diseases, with potential applications beyond the lungs.
Collapse
Affiliation(s)
- Ivan V. Chernikov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Irina K. Bachkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova Str., 1, 630090 Novosibirsk, Russia
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Mariya I. Meschaninova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Innokenty A. Savin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Valentin V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Elena L. Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| |
Collapse
|
33
|
Boosani CS, Burela L. The Exacerbating Effects of the Tumor Necrosis Factor in Cardiovascular Stenosis: Intimal Hyperplasia. Cancers (Basel) 2024; 16:1435. [PMID: 38611112 PMCID: PMC11010976 DOI: 10.3390/cancers16071435] [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: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
TNF-α functions as a master regulator of inflammation, and it plays a prominent role in several immunological diseases. By promoting important cellular mechanisms, such as cell proliferation, migration, and phenotype switch, TNF-α induces its exacerbating effects, which are the underlying cause of many proliferative diseases such as cancer and cardiovascular disease. TNF-α primarily alters the immune component of the disease, which subsequently affects normal functioning of the cells. Monoclonal antibodies and synthetic drugs that can target TNF-α and impair its effects have been developed and are currently used in the treatment of a few select human diseases. Vascular restenosis is a proliferative disorder that is initiated by immunological mechanisms. In this review, the role of TNF-α in exacerbating restenosis resulting from neointimal hyperplasia, as well as molecular mechanisms and cellular processes affected or induced by TNF-α, are discussed. As TNF-α-targeting drugs are currently not approved for the treatment of restenosis, the summation of the topics discussed here is anticipated to provide information that can emphasize on the use of TNF-α-targeting drug candidates to prevent vascular restenosis.
Collapse
Affiliation(s)
- Chandra Shekhar Boosani
- Somatic Cell and Genome Editing Center, Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
- MU HealthCare, University of Missouri, Columbia, MO 65211, USA
- Technology and Platform Development, Soma Life Science Solutions, Winston-Salem, NC 27103, USA
| | | |
Collapse
|
34
|
Ning R, Zhao J, Chen L, Mu H, Chen Z, Yang K, Xu X, Litifu D, Zuo J, He S, Jiang M, Zhao W. Macrolide sesquiterpene pyridine alkaloids from Celastrus monospermus and evaluation of their immunosuppressive and anti-osteoclastogenesis activities. Bioorg Chem 2024; 145:107246. [PMID: 38428283 DOI: 10.1016/j.bioorg.2024.107246] [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: 01/15/2024] [Revised: 02/09/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Phytochemical investigation of the stems of Celastrus monospermus Roxb enabled isolation and identification of fifteen new macrolide sesquiterpene pyridine alkaloids (1-15) along with five known analogues. Their structures were elucidated by comprehensive spectroscopic analysis (NMR, HRESIMS, IR, UV), chemical hydrolysis, and single crystal X-ray diffraction analysis. Bioassay of the abundant isolates revealed that seven compounds inhibited the proliferation of B lymphocytes with IC50 values ranging between 1.4 and 19.9 μM. Among them, celasmondine C (3) could significantly promote the apoptosis of activated B lymphocyte, especially late-stage apoptosis. Besides, compounds 3, 16, and 20 exhibited potent suppression of osteoclast formation at a concentration of 1.0 μM. This investigation enriched the chemical diversity of macrolide sesquiterpene pyridine alkaloids, and supported evidence for the development of new immunosuppressive and anti-osteoclastogenesis agents.
Collapse
Affiliation(s)
- Ruonan Ning
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, People's Republic of China; Natural Product Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China
| | - Jie Zhao
- Natural Product Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China
| | - Li Chen
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Hongyan Mu
- Natural Product Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China
| | - Zhongxian Chen
- Natural Product Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China
| | - Kai Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, People's Republic of China
| | - Xing Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, People's Republic of China
| | - Dilinaer Litifu
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Jianping Zuo
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Shijun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China.
| | - Min Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, People's Republic of China.
| | - Weimin Zhao
- Natural Product Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, People's Republic of China.
| |
Collapse
|
35
|
Yurkina DM, Romanova EA, Tvorogova AV, Naydenysheva ZK, Feoktistov AV, Yashin DV, Sashchenko LP. The 12-Membered TNFR1 Peptide, as Well as the 16-Membered and 6-Membered TNF Peptides, Regulate TNFR1-Dependent Cytotoxic Activity of TNF. Int J Mol Sci 2024; 25:3900. [PMID: 38612709 PMCID: PMC11011327 DOI: 10.3390/ijms25073900] [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] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Understanding the exact mechanisms of the activation of proinflammatory immune response receptors is very important for the targeted regulation of their functioning. In this work, we were able to identify the sites of the molecules in the proinflammatory cytokine TNF (tumor necrosis factor) and its TNFR1 (tumor necrosis factor receptor 1), which are necessary for the two-stage cytotoxic signal transduction required for tumor cell killing. A 12-membered TNFR1 peptide was identified and synthesized, interacting with the ligands of this receptor protein's TNF and Tag7 and blocking their binding to the receptor. Two TNF cytokine peptides interacting with different sites of TNFR1 receptors were identified and synthesized. It has been demonstrated that the long 16-membered TNF peptide interferes with the binding of TNFR1 ligands to this receptor, and the short 6-membered peptide interacts with the receptor site necessary for the transmission of a cytotoxic signal into the cell after the ligands' interaction with the binding site. This study may help in the development of therapeutic approaches to regulate the activity of the cytokine TNF.
Collapse
Affiliation(s)
- Daria M. Yurkina
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Elena A. Romanova
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Anna V. Tvorogova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia;
| | - Zlata K. Naydenysheva
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Alexey V. Feoktistov
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
- Engelhardt Institute of Molecular Biology (RAS), Moscow 119334, Russia
| | - Denis V. Yashin
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Lidia P. Sashchenko
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| |
Collapse
|
36
|
Sadeghian I, Akbarpour M, Chafjiri FMA, Chafjiri PMA, Heidari R, Morowvat MH, Sadeghian R, Raee MJ, Negahdaripour M. Potential of oligonucleotide- and protein/peptide-based therapeutics in the management of toxicant/stressor-induced diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1275-1310. [PMID: 37688622 DOI: 10.1007/s00210-023-02683-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/21/2023] [Indexed: 09/11/2023]
Abstract
Exposure to toxicants/stressors has been linked to the development of many human diseases. They could affect various cellular components, such as DNA, proteins, lipids, and non-coding RNAs (ncRNA), thereby triggering various cellular pathways, particularly oxidative stress, inflammatory responses, and apoptosis, which can contribute to pathophysiological states. Accordingly, modulation of these pathways has been the focus of numerous investigations for managing related diseases. The involvement of various ncRNAs, such as small interfering RNA (siRNA), microRNAs (miRNA), and long non-coding RNAs (lncRNA), as well as various proteins and peptides in mediating these pathways, provides many target sites for pharmaceutical intervention. In this regard, various oligonucleotide- and protein/peptide-based therapies have been developed to treat toxicity-induced diseases, which have shown promising results in vitro and in vivo. This comprehensive review provides information about various aspects of toxicity-related diseases including their causing factors, main underlying mechanisms and intermediates, and their roles in pathophysiological states. Particularly, it highlights the principles and mechanisms of oligonucleotide- and protein/peptide-based therapies in the treatment of toxicity-related diseases. Furthermore, various issues of oligonucleotides and proteins/peptides for clinical usage and potential solutions are discussed.
Collapse
Affiliation(s)
- Issa Sadeghian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Biotechnology Incubator, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Akbarpour
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Morowvat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Javad Raee
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
37
|
Tong X, Shen Q. Identification of immune-related regulatory networks and diagnostic biomarkers in thyroid eye disease. Int Ophthalmol 2024; 44:38. [PMID: 38332455 DOI: 10.1007/s10792-024-03017-9] [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: 10/12/2023] [Accepted: 01/09/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Thyroid eye disease (TED) is an orbit-associated autoimmune inflammatory disorder intricately linked to immune dysregulation. Complete pathogenesis of TED remains elusive. This work aimed to mine pathogenesis of TED from immunological perspective and identify diagnostic genes. METHODS Gene expression microarray data for TED patients were downloaded from Gene Expression Omnibus, immune-related genes (IRGs) were from ImmPort database, and TED-related transcription factors (TFs) were from Cirtrome Cancer database. Differential analysis, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed. Regulatory networks of TFs and IRGs were constructed with Cytoscape. Diagnostic biomarkers in TED were identified through LASSO. Immune cell infiltration analysis was performed using CIBERSORT. RESULTS Twenty-three immune-related DEmRNAs were revealed and were primarily enriched in humoral immune response, positive regulation of inflammatory response, IL-17, and TNF pathways. Co-expression regulatory network included four TFs and 16 immune-related DEmRNAs. Seven diagnostic genes were identified, with Area Under the Curve (AUC) of 0.993 for training set and AUC value of 0.836 for validation set. TED patients exhibited elevated infiltration levels by macrophages M2, mast cells, and CD8 T cells among 22 immune cell types, whereas macrophages M2 and mast cells resting were significantly lower than normal group. CONCLUSIONS The seven feature genes had high diagnostic value for TED patients. Our work explored regulatory network and diagnostic biomarkers, laying theoretical basis for TED diagnosis and treatment.
Collapse
Affiliation(s)
- Xiangmei Tong
- The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310002, China
- Department of General Surgery, The First People's Hospital of Tonglu County, Tonglu, 311500, China
| | - Qianyun Shen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, 310002, China.
| |
Collapse
|
38
|
Giavatto C, Mourani J, Fitzpatrick C, Hardin B, Skrtic A, Evans A, Sredzinski E, Trieu S, Setter AI, Kobiska L, Lopez-Medina AI. Biosimilar perceptions among autoimmune prescribers and pharmacists in health system specialty pharmacy. J Manag Care Spec Pharm 2024; 30:175-182. [PMID: 38308629 PMCID: PMC10839464 DOI: 10.18553/jmcp.2024.30.2.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2024]
Abstract
BACKGROUND By the end of 2023, 10 self-administered biosimilars indicated for autoimmune conditions are expected to launch in the United States, resulting in alternative treatment options for patients and a potential for cost savings. However, studies about perception and knowledge of self-administered biosimilars among health system prescribers and health system specialty pharmacists are limited. OBJECTIVE To assesses knowledge and perceptions of biosimilars among autoimmune prescribers and health system specialty pharmacists across the United States. METHODS An anonymous, cross-sectional self-administered online questionnaire was conducted among prescribers and health system specialty pharmacists practicing in the specialties of rheumatology, dermatology, and gastroenterology across the United States. The survey was available from January 2023 to February 2023. RESULTS 31 prescribers and 44 pharmacists completed the questionnaire. Only 16.0% of prescribers and 13.4% of pharmacists reported being "very prepared" to have conversations with patients about biosimilar options. 43% of prescribers indicated they would prescribe a biosimilar to biologic naive patients. However, 13.3% of prescribers would be willing to prescribe a biosimilar to patients successfully established on biologic therapy. Among pharmacists, 68.1% were comfortable recommending a biosimilar substitution to a biologic naive patient, but only 18.1% would recommend a biosimilar substitution to an existing patient successfully established on a biologic therapy. Less than half of prescribers (48.0%) and pharmacists (42.0%) understood regulations of interchangeability and substitution. CONCLUSIONS Our study highlights several knowledge gaps and hesitancies that exist among health system specialty prescribers and pharmacists regarding biosimilar products. Education efforts are needed to overcome the lack of biosimilar adoption, which will increase affordability of therapy for patients and health care savings.
Collapse
|
39
|
Asenjo-Lobos C, González L, Bulnes JF, Roque M, Muñoz Venturelli P, Rodríguez GM. Cardiovascular events risk in patients with systemic autoimmune diseases: a prognostic systematic review and meta-analysis. Clin Res Cardiol 2024; 113:246-259. [PMID: 37650912 DOI: 10.1007/s00392-023-02291-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Chronic inflammation is considered a risk factor for the development of atherosclerosis and cardiovascular (CV) events. We seek to assess the risk of CV events in patients with Systemic autoimmune diseases (SAD), such as Systemic Lupus Erythematosus (SLE), Rheumatoid Arthritis (RA), Psoriasis (Ps) and Ankylosing Spondylitis (AS), compared with the general population. METHODS AND RESULTS A systematic search of MEDLINE from inception up to May 2021 was performed. Observational studies including individuals with and without autoimmune diseases (SLE, RA, Ps, AS), which reported a measure of association and variability for the effect of SAD on CV events, were included. The random effects meta-analysis was performed using the Hartung-Knapp-Sidik-Jonkman approach to obtain the pooled estimates. Cardiovascular Events including CV mortality, non-fatal myocardial infarction (MI), non-fatal stroke and coronary revascularization were the main outcomes evaluated. Fifty-four studies were selected, with a total of 24,107,072 participants. The presence of SAD was associated with an increased risk of CV mortality (HR 1.49 [95% CI 1.10-2.03]), non-fatal MI (HR 1.42 [95% CI 1.23-1.62]), and non-fatal stroke (HR 1.47 [95% CI 1.28-1.70]). RA, SLE, and Ps (particularly with arthritis) were significantly associated with a higher risk of MI and stroke. SAD was also associated with an increased risk of Major Adverse Cardiovascular Events (MACE) (HR 1.45 [95% CI 1.16-1.83]). CONCLUSION Patients with SAD present an increased risk of CV morbidity and mortality, which should be considered when establishing therapeutic strategies. These findings support the role of systemic inflammation in the development of atherosclerosis-driven disease.
Collapse
Affiliation(s)
- Claudia Asenjo-Lobos
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad de Desarrollo, Santiago, Chile
| | - Leticia González
- Centro de Imágenes Biomédicas, Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Francisco Bulnes
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marta Roque
- Iberoamerican Cochrane Centre, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Paula Muñoz Venturelli
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad de Desarrollo, Santiago, Chile
- Faculty of Medicine, The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
40
|
Yoon C, Ham YS, Gil WJ, Yang CS. Exploring the potential of Toxoplasma gondii in drug development and as a delivery system. Exp Mol Med 2024; 56:289-300. [PMID: 38297164 PMCID: PMC10907749 DOI: 10.1038/s12276-024-01165-7] [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: 09/13/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 02/02/2024] Open
Abstract
Immune-mediated inflammatory diseases are various groups of conditions that result in immune system disorders and increased cancer risk. Despite the identification of causative cytokines and pathways, current clinical treatment for immune-mediated inflammatory diseases is limited. In addition, immune-mediated inflammatory disease treatment can increase the risk of cancer. Several previous studies have demonstrated that Toxoplasma gondii manipulates the immune response by inhibiting or stimulating cytokines, suggesting the potential for controlling and maintaining a balanced immune system. Additionally, T. gondii also has the unique characteristic of being a so-called "Trojan horse" bacterium that can be used as a drug delivery system to treat regions that have been resistant to previous drug delivery therapies. In this study, we reviewed the potential of T. gondii in drug development and as a delivery system through current research on inflammation-regulating mechanisms in immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chanjin Yoon
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
- Institute of Natural Science & Technology, Hanyang University, Ansan, 15588, South Korea
| | - Yu Seong Ham
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
| | - Woo Jin Gil
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, Hanyang University, Ansan, 15588, South Korea.
- Center for Bionano Intelligence Education and Research, Ansan, 15588, South Korea.
- Department of Medicinal and Life Science, Hanyang University, Ansan, 15588, South Korea.
| |
Collapse
|
41
|
Li L, Lu J, Liu J, Wu J, Zhang X, Meng Y, Wu X, Tai Z, Zhu Q, Chen Z. Immune cells in the epithelial immune microenvironment of psoriasis: emerging therapeutic targets. Front Immunol 2024; 14:1340677. [PMID: 38239345 PMCID: PMC10794746 DOI: 10.3389/fimmu.2023.1340677] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory disease characterized by erroneous metabolism of keratinocytes. The development of psoriasis is closely related to abnormal activation and disorders of the immune system. Dysregulated skin protective mechanisms can activate inflammatory pathways within the epithelial immune microenvironment (EIME), leading to the development of autoimmune-related and inflammatory skin diseases. In this review, we initially emphasized the pathogenesis of psoriasis, paying particular attention to the interactions between the abnormal activation of immune cells and the production of cytokines in psoriasis. Subsequently, we delved into the significance of the interactions between EIME and immune cells in the emergence of psoriasis. A thorough understanding of these immune processes is crucial to the development of targeted therapies for psoriasis. Finally, we discussed the potential novel targeted therapies aimed at modulating the EIME in psoriasis. This comprehensive examination sheds light on the intricate underlying immune mechanisms and provides insights into potential therapeutic avenues of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Yu Meng
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| |
Collapse
|
42
|
Luz IS, Takaya R, Ribeiro DG, Castro MS, Fontes W. Proteomics: Unraveling the Cross Talk Between Innate Immunity and Disease Pathophysiology, Diagnostics, and Treatment Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:221-242. [PMID: 38409424 DOI: 10.1007/978-3-031-50624-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Inflammation is crucial in diseases, and proteins play a key role in the interplay between innate immunity and pathology. This review explores how proteomics helps understanding this relationship, focusing on diagnosis and treatment. We explore the dynamic innate response and the significance of proteomic techniques in deciphering the complex network of proteins involved in prevalent diseases, including infections, cancer, autoimmune and neurodegenerative disorders. Proteomics identifies key proteins in host-pathogen interactions, shedding light on infection mechanisms and inflammation. These discoveries hold promise for diagnostic tools, therapies, and vaccines. In cancer research, proteomics reveals innate signatures associated with tumor development, immune evasion, and therapeutic response. Additionally, proteomic analysis has unveiled autoantigens and dysregulation of the innate immune system in autoimmunity, offering opportunities for early diagnosis, disease monitoring, and new therapeutic targets. Moreover, proteomic analysis has identified altered protein expression patterns in neurodegenerative diseases like Alzheimer's and Parkinson's, providing insights into potential therapeutic strategies. Proteomics of the innate immune system provides a comprehensive understanding of disease mechanisms, identifies biomarkers, and enables effective interventions in various diseases. Despite still in its early stages, this approach holds great promise to revolutionize innate immunity research and significantly improve patient outcomes across a wide range of diseases.
Collapse
Affiliation(s)
- Isabelle Souza Luz
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Raquel Takaya
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Daiane Gonzaga Ribeiro
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Mariana S Castro
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, University of Brasilia, Brasília, Federal District, Brazil.
| |
Collapse
|
43
|
Clarke K, Ainslie-Garcia M, Ferko N, Shastri K. Modelling the opportunity for cost-savings or patient access with biosimilar adalimumab and tocilizumab: a European perspective. J Med Econ 2024; 27:952-962. [PMID: 39015093 DOI: 10.1080/13696998.2024.2379212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 07/09/2024] [Indexed: 07/18/2024]
Abstract
OBJECTIVES Biosimilars improve patient access by providing cost-effective treatment options. This study assessed the potential for savings and expanded patient access with increased use of two biosimilar disease modifying anti-rheumatic drugs (DMARDs): (a) approved adalimumab biosimilars and (b) the first tocilizumab biosimilar, representing an established biosimilar field and a recent biosimilar entrant in France, Germany, Italy, Spain, and the United Kingdom (UK). METHODS Separate ex-ante analyses were conducted for each country, parameterized using country-specific list prices, unit volumes annually, and market shares for each therapy. Discounting scenarios of 10%, 20%, and 30% were tested for tocilizumab. Outputs included direct cost-savings associated with drug acquisition or the incremental number of patients that could be treated if savings were redirected. Two biosimilar conversion scenarios were tested. RESULTS Savings associated with a 100% conversion to adalimumab biosimilar ranged from €10.5 to €187 million (UK and Germany, respectively), or an additional 1,096 to 19,454 patients that could be treated using the cost-savings. Introduction of a tocilizumab biosimilar provided savings up to €29.3 million in the most conservative scenario. Exclusive use of tocilizumab biosimilars (at a 30% discount) could increase savings to €28.8 to €113 million or expand access to an additional 43% of existing tocilizumab users across countries. CONCLUSION This study demonstrates the benefits that can be realized through increased biosimilar adoption, not only in an untapped tocilizumab market, but also through incremental increases in well-established markets such as adalimumab. As healthcare budgets continue to face downwards pressure globally, strategies to increase biosimilar market share could prove useful to help manage financial constraints.
Collapse
Affiliation(s)
| | | | | | - Kunal Shastri
- Fresenius Kabi SwissBioSim GmbH, Eysins, Switzerland
| |
Collapse
|
44
|
Hyldahl E, Gotfredsen K, Lynge Pedersen AM, Storgård Jensen S. Survival and Success of Dental Implants in Patients with Autoimmune Diseases: a Systematic Review. J Oral Maxillofac Res 2024; 15:e1. [PMID: 38812949 PMCID: PMC11131373 DOI: 10.5037/jomr.2024.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/31/2024] [Indexed: 05/31/2024]
Abstract
Objectives The purpose of this systematic review is to disclose the impact of autoimmune diseases and their medical treatment on dental implant survival and success. Material and Methods A literature search was conducted using MEDLINE (PubMed), The Cochrane Library and Embase up to December 6th, 2021. Any clinical study on patients with an autoimmune disease in whom implant therapy was performed was eligible. The quality of included studies was assessed using the Newcastle-Ottawa Scale. For each autoimmune disease group, data synthesis was divided into three groups: 1) overall results of the autoimmune disease, 2) overall results of corresponding control groups and 3) overall results of the autoimmune disease with a concomitant autoimmune disease (a subgroup of group 1). Descriptive statistics were used. Results Of 4,865 identified articles, 67 could be included and mainly comprising case reports and retrospective studies with an overall low quality. Implant survival rate was 50 to 100% on patient and implant level after a weighted mean follow-up of 17.7 to 68.1 months. Implant success was sporadically reported. Data on immunosuppressive medication were too heterogeneously reported to allow detailed analysis. Conclusions Overall, a high implant survival rate was reported in patients with autoimmune diseases. However, the identified studies were characterized by a low quality. No conclusions could be made regarding implant success and the effect of immunosuppressants due to heterogeneous reporting.
Collapse
Affiliation(s)
- Emil Hyldahl
- Oral Surgery, Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, CopenhagenDenmark.
| | - Klaus Gotfredsen
- Oral Rehabilitation, Section for Oral Health, Society and Technology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, CopenhagenDenmark.
| | - Anne Marie Lynge Pedersen
- Oral Pathology and Medicine, Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, CopenhagenDenmark.
| | - Simon Storgård Jensen
- Oral Surgery, Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, CopenhagenDenmark.
| |
Collapse
|
45
|
Bayati P, Taherian M, Mojtabavi N. Immunomodulatory effects of the induced pluripotent stem cells through expressing IGF-related factors and IL-10 in vitro. Int J Immunopathol Pharmacol 2024; 38:3946320241276899. [PMID: 39162714 PMCID: PMC11337182 DOI: 10.1177/03946320241276899] [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: 02/06/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Induced Pluripotent Stem Cells (IPSCs) represent an innovative strategy for addressing challenging diseases, including various rheumatologic conditions. Aside from their regenerative capacities, some studies have shown the potential of these cells in the modulation of inflammatory responses. The underlying mechanisms by which they exert their effects have yet to be fully comprehended. Therefore, we aimed to explore the gene expression linked to the IGF pathway as well as IL-10 and TGF-β, which are known to exert immunomodulatory effects. METHODS A C57/Bl6 pregnant mouse was used for obtaining mouse embryonic fibroblasts (MEFs), then the IPSCs were induced using lentiviral vectors expressing the pluripotency genes (OCT4, SOX2, KLF1, and c-MYC). Cells were cultured for 72 h in DMEM high glucose plus leukemia inhibitory factor; Evaluating the gene expression was conducted using specific primers for Igf1, Igf2, Igfbp3, Igfbp4, Irs1, Il-10, and Tgf-β genes, as well as SYBR green qPCR master mix. The data were analyzed using the 2-ΔΔCT method and were compared by employing the t test; the results were plotted using GraphPad PRISM software. MEFs were utilized as controls. RESULTS Gene expression analyses revealed that Igf-1, Igf-bp3, Igf-bp4, and Il-10 were significantly overexpressed (p ≤ .01), while Igf-2 and Tgf-b genes were significantly downregulated in the lysates from IPSCs in comparison with the control MEFs. The Irs1 gene expression was not altered significantly. CONCLUSION IPSCs are potentially capable of modulating inflammatory responses through the expression of various anti-inflammatory mediators from the IGF signaling, as well as IL-10. This discovery uncovers a previously unknown dimension of IPSCs' therapeutic effects, potentially leading to more advanced in vivo research and subsequent clinical trials.
Collapse
Affiliation(s)
- Paria Bayati
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Kraus S, Lee E. A human initial lymphatic chip reveals distinct mechanisms of primary lymphatic valve dysfunction in acute and chronic inflammation. LAB ON A CHIP 2023; 23:5180-5194. [PMID: 37981867 PMCID: PMC10908576 DOI: 10.1039/d3lc00486d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Interstitial fluid uptake and retention by lymphatic vessels (LVs) play a role in maintaining interstitial fluid homeostasis. While it is well-established that intraluminal lymphatic valves in the collecting LVs prevent fluid backflow (secondary lymphatic valves), a separate valve system in the initial LVs that only permits interstitial fluid influx into the LVs, preventing fluid leakage back to the interstitium (primary lymphatic valves), remains incompletely understood. Although lymphatic dysfunction is commonly observed in inflammation and autoimmune diseases, how the primary lymphatic valves are affected by acute and chronic inflammation has scarcely been explored and even less so using in vitro lymphatic models. Here, we developed a human initial lymphatic vessel chip where interstitial fluid pressure and luminal fluid pressure are controlled to examine primary lymph valve function. In normal conditions, lymphatic drainage (fluid uptake) and permeability (fluid leakage) in engineered LVs were maintained high and low, respectively, which was consistent with our understanding of healthy primary lymph valves. Next, we examined the effects of acute and chronic inflammation. Under the acute inflammation condition with a TNF-α treatment (2 hours), degradation of fibrillin and impeded lymphatic drainage were observed, which were reversed by treatment with anti-inflammatory dexamethasone. Surprisingly, the chronic inflammation condition (repeated TNF-α treatments during 48 hours) deposited fibrillin to compensate for the fibrillin loss, showing no change in lymphatic drainage. Instead, the chronic inflammation condition led to cell death and disruption of lymphatic endothelial cell-cell junctions, increasing lymphatic permeability and fluid leakage. Our human lymphatic model shows two distinct mechanisms by which primary lymphatic valve dysfunction occurs in acute and chronic inflammation.
Collapse
Affiliation(s)
- Samantha Kraus
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
47
|
Aliabadi A, Khanniri E, Mahboubi-Rabbani M, Bayanati M. Dual COX-2/15-LOX inhibitors: A new avenue in the prevention of cancer. Eur J Med Chem 2023; 261:115866. [PMID: 37862815 DOI: 10.1016/j.ejmech.2023.115866] [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: 08/27/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
Dual cyclooxygenase 2/15-lipoxygenase inhibitors constitute a valuable alternative to classical non-steroidal anti-inflammatory drugs (NSAIDs) and selective COX-2 (cyclooxygenase-2) inhibitors for the treatment of inflammatory diseases, as well as preventing the cancer. Indeed, these latter present diverse side effects, which are reduced or absent in dual-acting agents. In this review, COX-2 and 15-LOX (15-lipoxygenase) pathways are first described in order to highlight the therapeutic interest of designing such compounds. Various structural families of dual inhibitors are illustrated. This study discloses various structural families of dual 15-LOX/COX-2 inhibitors, thus pave the way to design potentially-active anticancer agents with balanced dual inhibition of these enzymes.
Collapse
Affiliation(s)
- Ali Aliabadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elham Khanniri
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahboubi-Rabbani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maryam Bayanati
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Gouda NA, Alshammari SO, Abourehab MAS, Alshammari QA, Elkamhawy A. Therapeutic potential of natural products in inflammation: underlying molecular mechanisms, clinical outcomes, technological advances, and future perspectives. Inflammopharmacology 2023; 31:2857-2883. [PMID: 37950803 DOI: 10.1007/s10787-023-01366-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/06/2023] [Indexed: 11/13/2023]
Abstract
Chronic inflammation is a common underlying factor in many major diseases, including heart disease, diabetes, cancer, and autoimmune disorders, and is responsible for up to 60% of all deaths worldwide. Metformin, statins, and corticosteroids, and NSAIDs (non-steroidal anti-inflammatory drugs) are often given as anti-inflammatory pharmaceuticals, however, often have even more debilitating side effects than the illness itself. The natural product-based therapy of inflammation-related diseases has no adverse effects and good beneficial results compared to substitute conventional anti-inflammatory medications. In this review article, we provide a concise overview of present pharmacological treatments, the pathophysiology of inflammation, and the signaling pathways that underlie it. In addition, we focus on the most promising natural products identified as potential anti-inflammatory therapeutic agents. Moreover, preclinical studies and clinical trials evaluating the efficacy of natural products as anti-inflammatory therapeutic agents and their pragmatic applications with promising outcomes are reviewed. In addition, the safety, side effects and technical barriers of natural products are discussed. Furthermore, we also summarized the latest technological advances in the discovery and scientific development of natural products-based medicine.
Collapse
Affiliation(s)
- Noha A Gouda
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea.
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
49
|
Fang Z, Sun H, Wang Y, Sun Z, Yin M. Discovery of WD-890: A novel allosteric TYK2 inhibitor for the treatment of multiple autoimmune diseases. Biomed Pharmacother 2023; 167:115611. [PMID: 37778274 DOI: 10.1016/j.biopha.2023.115611] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023] Open
Abstract
Tyrosine kinase 2 (TYK2) as a member of Janus kinase (JAK) family, mainly mediates the signaling of type I interferons (IFN), interleukin-12 (IL-12) and interleukin-23 (IL-23), which has become an attractive target for treatment of immune and inflammatory diseases. However, the development of selective TYK2 inhibitors is challenging due to the high homology of the catalytic kinase domain among the JAK family members. Here, we report a novel and potent allosteric inhibitor, WD-890, which binds to the pseudokinase domain of TYK2 with high selectivity and inhibits its function. We accomplished a series of preclinical studies to demonstrate the therapeutic efficacy of WD-890 in four animal models: systemic lupus erythematosus (SLE), psoriasis, psoriatic arthritis (PsA), and inflammatory bowel disease (IBD). The pharmacokinetic and toxicology results further indicate that WD-890 has favorable absorption, distribution, metabolism, and excretion (ADME) properties and tolerable toxicity. In conclusion, our study shows that WD-890 could be a promising oral TYK2 inhibitor for future treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Zhiqin Fang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongyin Sun
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Yutong Wang
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhenliang Sun
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, China.
| | - Mingzhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China; Translational Medicine Research Center (TMRC), School of Medicine Chongqing University, Shapingba, Chongqing, China.
| |
Collapse
|
50
|
Kao FC, Hsu YC, Tu YK, Chen TS, Wang HH, Lin J(CF. Long-Term Use of Immunosuppressive Agents Increased the Risk of Fractures in Patients with Autoimmune Diseases: An 18-Year Population-Based Cohort Study. Biomedicines 2023; 11:2764. [PMID: 37893136 PMCID: PMC10604306 DOI: 10.3390/biomedicines11102764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The risk of fractures is higher in patients with autoimmune diseases, but it is not clear whether the use of immunosuppressive agents can further increase this risk. To investigate this issue, a retrospective study was conducted using data from Taiwan's National Health Insurance Research Database. Patients diagnosed with autoimmune diseases between 2000 and 2014, including psoriatic arthritis, rheumatoid arthritis, ankylosing spondylitis, and systemic lupus erythematosus, were included in the study. A control group of patients without autoimmune diseases was selected from the same database during the same period. Patients with autoimmune diseases were divided into two sub-cohorts based on their use of immunosuppressive agents. This study found the risk of fractures was 1.14 times higher in patients with autoimmune diseases than in those without. Moreover, we found that patients in the immunosuppressant sub-cohort had a higher risk of fractures compared to those in the non-immunosuppressant sub-cohort. The adjusted sub-distribution hazard ratio for shoulder fractures was 1.27 (95% CI = 1.01-1.58), for spine fractures was 1.43 (95% CI = 1.26-1.62), for wrist fractures was 0.95 (95% CI = 0.75-1.22), and for hip fractures was 1.67 (95% CI = 1.38-2.03). In conclusion, the long-term use of immunosuppressive agents in patients with autoimmune diseases may increase the risk of fractures.
Collapse
Affiliation(s)
- Feng-Chen Kao
- Department of Orthopedics, E-Da Hospital, Kaohsiung City 824, Taiwan; (F.-C.K.); (Y.-K.T.)
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 824, Taiwan; (Y.-C.H.); (H.-H.W.)
- Department of Orthopedics, E-Da Dachang Hospital, Kaohsiung City 807, Taiwan
| | - Yao-Chun Hsu
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 824, Taiwan; (Y.-C.H.); (H.-H.W.)
- Division of Gastroenterology, E-Da Hospital, Kaohsiung City 824, Taiwan
| | - Yuan-Kun Tu
- Department of Orthopedics, E-Da Hospital, Kaohsiung City 824, Taiwan; (F.-C.K.); (Y.-K.T.)
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 824, Taiwan; (Y.-C.H.); (H.-H.W.)
| | - Tzu-Shan Chen
- Department of Medical Research, E-Da Hospital, Kaohsiung City 824, Taiwan;
- Department of Medical Imaging and Radiological Sciences, College of Medicine, I-Shou University, Kaohsiung City 824, Taiwan
| | - Hsi-Hao Wang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 824, Taiwan; (Y.-C.H.); (H.-H.W.)
- Division of Nephrology, Department of Internal Medicine, E-DA Hospital, Kaohsiung City 824, Taiwan
- Department of Medical Quality, E-DA Hospital, Kaohsiung City 824, Taiwan
| | - Jeff (Chien-Fu) Lin
- Department of Statistics, National Taipei University, Taipei City 23741, Taiwan
- Department of Orthopedic Surgery, Wan Fang Hospital, Taipei City 116, Taiwan
| |
Collapse
|