1
|
Raju V, Reddy R, Javan AC, Hajihossainlou B, Weissleder R, Guiseppi-Elie A, Kurabayashi K, Jones SA, Faghih RT. Tracking inflammation status for improving patient prognosis: A review of current methods, unmet clinical needs and opportunities. Biotechnol Adv 2025; 82:108592. [PMID: 40324661 PMCID: PMC12173025 DOI: 10.1016/j.biotechadv.2025.108592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/07/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Inflammation is the body's response to infection, trauma or injury and is activated in a coordinated fashion to ensure the restoration of tissue homeostasis and healthy physiology. This process requires communication between stromal cells resident to the tissue compartment and infiltrating immune cells which is dysregulated in disease. Clinical innovations in patient diagnosis and stratification include measures of inflammatory activation that support the assessment of patient prognosis and response to therapy. We propose that (i) the recent advances in fast, dynamic monitoring of inflammatory markers (e.g., cytokines) and (ii) data-dependent theoretical and computational modeling of inflammatory marker dynamics will enable the quantification of the inflammatory response, identification of optimal, disease-specific biomarkers and the design of personalized interventions to improve patient outcomes - multidisciplinary efforts in which biomedical engineers may potentially contribute. To illustrate these ideas, we describe the actions of cytokines, acute phase proteins and hormones in the inflammatory response and discuss their role in local wounds, COVID-19, cancer, autoimmune diseases, neurodegenerative diseases and aging, with a central focus on cardiac surgery. We also discuss the challenges and opportunities involved in tracking and modulating inflammation in clinical settings.
Collapse
Affiliation(s)
- Vidya Raju
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA
| | - Revanth Reddy
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA
| | | | - Behnam Hajihossainlou
- Department of Infectious Diseases, Harlem Medical Center, and Columbia University, New York, 10032, NY, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Department of Systems Biology, Harvard Medical School, and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Anthony Guiseppi-Elie
- Department of Biomedical Engineering, Center for Bioelectronics, Biosensors and Biochips (C3B), and Department of Electrical and Computer Engineering, Texas A & M University, College Station, 77843, TX, USA; Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute, Houston, 77030, TX, USA; ABTECH Scientific, Inc., Biotechnology Research Park, Richmond, 23219, Virginia, USA
| | - Katsuo Kurabayashi
- Department of Mechanical and Aerospace Engineering, New York University, New York 11201, NY, USA
| | - Simon A Jones
- Division of Infection and Immunity, and School of Medicine, Cardiff University, UK; Systems Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Rose T Faghih
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, 11201, NY, USA.
| |
Collapse
|
2
|
Gheorghe-Milea A, Stănoiu-Pînzariu O, Georgescu CE. Unravelling the link between chronic inflammation and primary hyperparathyroidism: a systematic review. Front Immunol 2025; 16:1563967. [PMID: 40529359 PMCID: PMC12171116 DOI: 10.3389/fimmu.2025.1563967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/14/2025] [Indexed: 06/20/2025] Open
Abstract
Introduction Primary hyperparathyroidism (PHPT) is a multisystemic endocrine disorder characterized by an incompletely understood pathogenesis, a complex clinical picture and various complications. Chronic inflammation represents a state that can affect the normal function of cells and cause tissue damage, therefore increasing the risk of certain diseases, including cancer, metabolic, cardiovascular or neurodegenerative disorders. Aim Reviewing existing data on markers of inflammation in patients with PHPT, with potential implications in understanding the pathogenesis of PHPT, stratifying the risk for complications and providing new diagnostic biomarkers and a personalized therapeutic approach, especially in patients who cannot be operated on. Methods A systematic review was conducted by searching in four electronic databases (PubMed, Embase, Web of Science and Scopus) and summarizing data from studies that evaluated inflammatory markers in patients with PHPT. Results The review included a total of 28 articles, encompassing data from 1572 patients diagnosed with PHPT. Various markers associated with chronic inflammation, including High sensitivity C-Reactive Protein (CRP), Tumor Necrosis Factor-α, Interleukin (IL)-6, and fibrinogen, were found to be elevated in PHPT patients. White blood count (WBC) values were similar in patients and controls in most studies, while for some markers derived from the full blood count significant differences were found between these groups. Correlations between PTH levels and several biomarkers, including IL-6, CRP and WBC, were also identified. Data on the impact of parathyroidectomy on inflammation parameters were conflicting. Conclusion The findings from this systematic review suggest an association between chronic inflammation and primary hyperparathyroidism, underscoring the potential role of inflammation as a mediator of PHPT-related complications. Targeting inflammatory pathways may offer novel therapeutic strategies for mitigating systemic effects of PHPT and improving patient outcomes.
Collapse
Affiliation(s)
- Ana Gheorghe-Milea
- Department of Endocrinology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Endocrinology Clinic, Cluj County Emergency Clinical Hospital, Cluj-Napoca, Romania
| | - Oana Stănoiu-Pînzariu
- Department of Endocrinology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Endocrinology Clinic, Cluj County Emergency Clinical Hospital, Cluj-Napoca, Romania
| | - Carmen Emanuela Georgescu
- Department of Endocrinology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Endocrinology Clinic, Cluj County Emergency Clinical Hospital, Cluj-Napoca, Romania
| |
Collapse
|
3
|
Rajpoot A, Sharma V. Protective effects of Allium sativum essential oil against lead nitrate-induced cardiotoxicity: Modulation of lipid metabolism, nitric oxide dynamics, inflammatory mediators, and histological profiles in Swiss albino mice. Toxicol Rep 2025; 14:101950. [PMID: 40026478 PMCID: PMC11869988 DOI: 10.1016/j.toxrep.2025.101950] [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] [Indexed: 03/05/2025] Open
Abstract
Background Lead (Pb²⁺) is a toxic metal known to induce oxidative stress and inflammation, contributing to cardiovascular diseases such as hypertension and atherosclerosis. Natural compounds like Allium sativum essential oil (ASEO) offer potential therapeutic benefits against lead-induced damage, but their cardioprotective effects remain underexplored. This study investigates the efficacy of ASEO in mitigating cardiovascular toxicity induced by lead nitrate in male Swiss albino mice. Methods Thirty-six male mice were divided into six groups: Control, Lead Nitrate (50 mg/kg), Lead Nitrate + Low-dose ASEO (50 mg/kg), Lead Nitrate + High-dose ASEO (80 mg/kg), Lead Nitrate + Silymarin (25 mg/kg), and Lead Nitrate + Olive Oil. After 12 days of lead exposure, treatments were administered for 30 days. Key cardiovascular parameters such as lipid profiles (total cholesterol, LDL, HDL), nitric oxide (NO), and inflammatory markers (TNF-α, IL-6, IFN-γ, IL-10, NF-κB) were evaluated alongside histological analysis of cardiac tissue. Results Lead nitrate exposure significantly increased total cholesterol (88.27 µg/mL) and LDL (93.78 µg/mL) while reducing HDL (17.51 µg/mL) compared to controls (P < 0.001). High-dose ASEO lowered total cholesterol (66.07 µg/mL) and LDL (49.62 µg/mL) while increased HDL (27.2 µg/mL) (P < 0.001). NO levels, reduced by lead exposure, were significantly restored by high-dose ASEO (P < 0.001). Inflammatory markers, including TNF-α, NF-kB, and IL-6, were elevated in the lead group but decreased significantly following ASEO treatment (P < 0.001). Histological analysis showed that ASEO markedly preserved myocardial architecture, reducing degeneration and inflammation. Conclusion High-dose ASEO demonstrated significant cardioprotective effects against lead-induced toxicity by improving lipid profiles, enhancing NO levels, and modulating inflammatory markers. Further studies are warranted to validate these results.
Collapse
Affiliation(s)
| | - Veena Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| |
Collapse
|
4
|
Westhoff TA, Sipka AS, Overton TR, Klaessig S, Mann S. Evaluation of circulating cytokine concentrations and ex vivo indicators of the inflammatory response in transition dairy cows fed pre- and postpartum diets differing in metabolizable protein supply. J Dairy Sci 2025; 108:6427-6438. [PMID: 40221035 DOI: 10.3168/jds.2024-26074] [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: 11/26/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025]
Abstract
The nutrient deficit during the transition period might alter activity of the nutrient-sensing mechanistic target of rapamycin, thereby influencing immune phenotype and the inflammatory balance of transition cows. We investigated changes in circulating markers of inflammation during the transition period. Additionally, we assessed changes in ex vivo indicators of the whole blood leukocyte cytokine response to LPS stimulation and leukocyte phagocytosis and oxidative burst. The second objective was to determine whether increasing the MP supply in the prepartum, the postpartum, or both diets would affect the measured parameters. Multiparous Holstein cows (n = 96) were assigned to 1 of 4 treatment groups at 28 d before expected calving following a randomized block design. Prepartum diets were formulated to contain either a control (85 g MP/kg DM) or high (113 g MP/kg DM) level of estimated MP. Postpartum diets were formulated to contain either a control (104 g MP/kg DM) or high (131 g MP/kg DM) level of estimated MP. To control the potential confounding effect of Met and Lys supply, diets were formulated to supply an equal amount at 1.24 and 3.84 g/Mcal of ME in both prepartum diets and 1.15 and 3.16 g/Mcal of ME in both postpartum diets, respectively. The combination of a pre- and a postpartum diet resulted in treatment groups: control-control (CC), control-high (CH), high-control (HC), and high-high (HH). Serum concentrations of tumor necrosis factor (TNF), IL-10, and IFN-γ were determined at -6, 3, 10, and 21 d relative to calving using a multiplex assay. Complete blood cell count, whole blood cytokine response to LPS stimulation, and PMN and peripheral blood mononuclear cell (PBMC) phagocytosis and oxidative burst were determined at -28, -10, 7, and 21 d relative to calving. Serum concentrations of TNF were below the lower limit of detection (≤12 pg/mL) in 282 (75.4%) samples. Serum concentrations of IL-10 and IFN-γ were greatest at -6 d relative to calving. Serum concentrations of IFN-γ did not differ by treatment, but IL-10 was greater in CH compared with HH and HC at 10 and 21 DIM, respectively. Compared with CC, white blood cell counts were 13.9% higher in HC, granulocyte counts were 17.6% and 14.7% higher in CH and HC, respectively, and monocyte counts were 27.4% higher at 7 DIM in HC. Lymphocyte counts were 12.7% and 13.9% higher in HC compared with CC and CH, respectively. Phagocytic ability and oxidative burst of PMN and PBMC did not differ by treatment. Whole blood LPS-induced IL-10 and TNF concentrations increased to a greater extent at 7 DIM and 7 and 21 DIM compared with -10 d relative to calving, respectively, and similarly in all treatments. In summary, whereas serum IL-10 and IFN-γ concentrations were greatest during late gestation, whole blood LPS-induced cytokines and phagocytosis increased to a greater extent during early lactation, suggesting a robust inflammatory response. However, increasing the MP supply during the transition period did not meaningfully influence indicators of the inflammatory response.
Collapse
Affiliation(s)
- Trent A Westhoff
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Anja S Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Thomas R Overton
- Department of Animal Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853
| | - Suzanne Klaessig
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Sabine Mann
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
5
|
Gaillard SL, Broadwater G, Berchuck A, Andrews WS, Havrilesky LJ, Davidson BA, Previs RA, Starr MD, Yi JS, Nixon AB, Neff JL, Conejo-Garcia JR, Secord AA. A pilot study investigating the effect of pembrolizumab on the tumoral immunoprofile of newly diagnosed mullerian cancers. Gynecol Oncol Rep 2025; 59:101770. [PMID: 40521348 PMCID: PMC12166832 DOI: 10.1016/j.gore.2025.101770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025] Open
Abstract
Objective This pilot window of opportunity study was conducted to assess feasibility, toxicity, and changes in immune parameters in response to one dose of the PD-1 inhibitor, pembrolizumab, in patients newly diagnosed with mullerian epithelial cancers. Methods Eligible patients received pembrolizumab 200 mg IV once ≥ 7 days prior to further standard therapy, including adjuvant chemotherapy. Tissue and blood were collected before and ≥ 7 days after pembrolizumab administration. Primary endpoints included change in tumor infiltrating lymphocytes (TIL), feasibility, and toxicity based on frequency and severity of adverse events. Exploratory objectives included tumor assessment of immunohistochemical PD-L1 staining using a quantitative modified proportion score and qualitative assessment of immune presence at the stromal interface. Measurement of cytokine levels and digital spatial profiling were performed from plasma and tissue samples, respectively, before and after pembrolizumab. Results Fifteen patients enrolled and received pembrolizumab. TIL levels changed in 4 of 11 paired sets, with 3 decreasing and 1 increasing post-treatment. PD-L1 modified proportion score increased in 7 cases, decreased in 2, and remained unchanged in 2. The stromal interface switched from negative to positive in 3 cases. Collectively, 8 of 10 assessable tumor pairs demonstrated either an increase in PD-L1 modified proportion score or the stromal interface switched from negative to positive. Circulating CXCL10 and TNFα levels increased after pembrolizumab in patients with response, but decreased in the one patient with progression on adjuvant chemotherapy. Digital spatial profiling showed increased IDO1 protein expression in immune and tumor compartments after treatment. Conclusion A single dose of pembrolizumab increased PD-L1 modified proportion score and/or stromal interface immune cells suggesting potential for local tumor immunologic recruitment. Additionally, increases in systemic inflammation, measured by cytokine production and differential IDO1 expression, reflect an interferon response. These hypothesis-generating data need to be confirmed and validated in larger subsets.
Collapse
Affiliation(s)
- Stéphanie L. Gaillard
- Departments of Oncology and Obstetrics and Gynecology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States
- Division of Gynecologic Oncology, Duke Cancer Institute, United States
- Department of Medicine, Duke Cancer Institute, United States
| | - Gloria Broadwater
- Duke Cancer Institute Biostatistics, Duke Cancer Institute, United States
| | - Andrew Berchuck
- Division of Gynecologic Oncology, Duke Cancer Institute, United States
| | | | | | | | - Rebecca A. Previs
- Division of Gynecologic Oncology, Duke Cancer Institute, United States
| | - Mark D. Starr
- Department of Surgery, Duke Cancer Institute, United States
| | - John S. Yi
- Department of Surgery, Duke Cancer Institute, United States
| | - Andrew B. Nixon
- Department of Medicine, Duke Cancer Institute, United States
| | | | | | | |
Collapse
|
6
|
Miles MA, Jayawardena S, Liong S, Liong F, Trollope GS, O'Leary JJ, Brooks DA, Selemidis S. TLR7 deficiency enhances inflammation in the URT but reduces LRT immunity following influenza A infection. Sci Rep 2025; 15:18918. [PMID: 40442368 PMCID: PMC12123008 DOI: 10.1038/s41598-025-04154-6] [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: 02/18/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025] Open
Abstract
Immune responses in the upper respiratory tract (URT) following influenza A virus (IAV) infection can influence disease severity, and subsequently inflammation and lung tissue damage in the lower respiratory tract (LRT). This study investigated the role of toll-like receptor 7 (TLR7), a key pattern recognition receptor that senses viral RNA and triggers antiviral and proinflammatory signaling to activate immune responses, in specifically shaping URT and LRT immune responses to IAV infection. Wild type C57Bl/6 and TLR7 knockout (TLR7 KO) mice were infected with the H3N2 IAV strain Hk-X31, and key immune responses in the nasal tissue (URT) and lower airways and lung tissue (LRT) measured after acute infection. We found reduced body weight loss, and increased type II/III interferons and proinflammatory cytokines in the URT of TLR7 KO mice; while LRT inflammation was reduced. TLR7 was essential for activating immune responses in the LRT but played a more selective role in the URT, primarily influencing monocytes, pDCs and B cells. Our data suggest that TLR7 plays a critical role in the transition of inflammation from the URT to the LRT during IAV infection, making it a promising therapeutic target to modulate disease severity.
Collapse
Affiliation(s)
- Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Sahan Jayawardena
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Stella Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Felicia Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Gemma S Trollope
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - John J O'Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin, Ireland
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
7
|
Sancho-Saldaña A, Gil-Sánchez A, Quirant-Sánchez B, Boigues M, Canudes M, Peralta S, Solana MJ, González-Mingot C, Quibus L, Martínez-Cáceres E, Torres P, Hervás JV, Moreno-Magallon J, Brieva L. Profile of Cytokines Associated with SARS-CoV2 Seropositivity in Multiple Sclerosis Patients and Its Persistence over Six Months. J Clin Med 2025; 14:3736. [PMID: 40507498 PMCID: PMC12155705 DOI: 10.3390/jcm14113736] [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: 03/26/2025] [Revised: 05/08/2025] [Accepted: 05/18/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Patients with multiple sclerosis (pwMS) receiving disease-modifying therapies (DMTs) may exhibit altered immune responses to infections such as SARS-CoV-2. This study aimed to characterize the cytokine profiles associated with prior SARS-CoV-2 infection and to identify immune markers related to the persistence of the humoral response in pwMS. Methods: A total of 90 pwMS were recruited before the introduction of COVID-19 vaccination in Spain; 46 were seropositive-defined by the presence of IgG, IgM, or IgA antibodies against SARS-CoV-2-and 44 were seronegative. We compared baseline cytokine levels between groups and followed seropositive individuals for six months to assess IgG antibody persistence. Results: Seropositive patients showed significantly lower baseline levels of IL-10, IL-23, and IFN-α compared to seronegative individuals. Notably, elevated IL-18 at baseline was associated with persistent IgG seropositivity at six months. Conclusions: These findings suggest a distinct cytokine profile in SARS-CoV-2-exposed pwMS and highlight IL-18 as a potential marker of sustained humoral response. This study provides insight into host-virus immune dynamics in MS patients and may help guide future strategies for infection monitoring and immune evaluation in this population.
Collapse
Affiliation(s)
- Agustín Sancho-Saldaña
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Anna Gil-Sánchez
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Bibiana Quirant-Sánchez
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
- Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Cerdanyola del Valles, Spain
| | - Marc Boigues
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
| | - Marc Canudes
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Silvia Peralta
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - María José Solana
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Cristina González-Mingot
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Department of Medicine, University of Lleida (UdL), 25198 Lleida, Spain
| | - Laura Quibus
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Eva Martínez-Cáceres
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (B.Q.-S.); (M.B.); (E.M.-C.)
- Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Cerdanyola del Valles, Spain
| | - Pascual Torres
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, Lleida Biomedical Research Institute (IRBLleida), University of Lleida (UdL), 25198 Lleida, Spain
| | - José Vicente Hervás
- Servicio de Neurología, Hospital de Sant Joan Despí Moisés Broggi, 08970 Barcelona, Spain;
| | - Judith Moreno-Magallon
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
| | - Luis Brieva
- Servicio de Neurología, Hospital Universitario Arnau de Vilanova, 25198 Lleida, Spain; (A.S.-S.); (S.P.); (M.J.S.); (C.G.-M.)
- Servicio de Neuroinmunología, Institut de Recerca Biomèdica de Lleida-IRBLleida, 25198 Lleida, Spain; (A.G.-S.); (M.C.); (L.Q.); (P.T.); (J.M.-M.)
- Department of Medicine, University of Lleida (UdL), 25198 Lleida, Spain
| |
Collapse
|
8
|
Lai WY, Chuang CW, Huang YC, Huang CJ. Therapeutic Potential of Plant-Derived Small Extracellular Vesicles in Sepsis: A Network Meta-analysis. Pharmacol Res 2025:107795. [PMID: 40414583 DOI: 10.1016/j.phrs.2025.107795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Sepsis is a life-threatening condition characterized by systemic inflammation and multi-organ dysfunction. Plant-derived small extracellular vesicles (sEVs) have emerged as promising therapeutic agents due to their antioxidant, anti-inflammatory, and immunomodulatory properties. This study conducted a network meta-analysis to identify the most effective plant-derived sEVs for reducing sepsis-induced inflammation and oxidative stress. The analysis included 13 studies evaluating 10 plant-derived sEVs in sepsis-mimicking conditions, with primary outcomes focused on cytokine levels and reactive oxygen species (ROS) production in vitro and in vivo. Secondary outcomes included nuclear factor erythroid 2-related factor 2 (Nrf2) expression and cell viability. The study protocol was registered with PROSPERO (CRD420251011005). Ginger-derived sEVs were identified as the most effective, significantly reducing pro-inflammatory cytokines (interleukin-6 and tumor necrosis factor-α), increasing the anti-inflammatory cytokine (interleukin-10), and suppressing ROS production. They also enhanced Nrf2 expression and improved cell viability, highlighting their role in antioxidant defense and cytoprotection. In conclusion, ginger-derived sEVs are the most effective plant-derived sEVs for mitigating sepsis-induced inflammation and oxidation in both in vitro and in vivo sepsis-mimicking models.
Collapse
Affiliation(s)
- Wen-Yi Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wei Chuang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Jen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Schreiber CS, Ramil LN, Bieligk J, Meineke R, Käufer C, Richter F. Intravenous SARS-CoV-2 Spike protein induces neuroinflammation and alpha-synuclein accumulation in brain regions relevant to Parkinson's disease. Brain Behav Immun 2025:S0889-1591(25)00197-7. [PMID: 40404020 DOI: 10.1016/j.bbi.2025.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/28/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) frequently presents with neurological symptoms in human patients and leads to long-lasting brain pathology in a hamster model. There is no overt SARS-CoV-2 virus replication in central neurons. Whether viral proteins are sufficient to cause this pathology requires further investigations. The SARS-CoV-2 Spike-protein S1-subunit (S1-protein) has recently gained interest for causing neuroinflammation and accelerating aggregation of alpha-synuclein (aSyn) in vitro. Here, we show the impact of S1-protein in a broad spectrum of brain regions after injection via three different application routes in C57/BL6 mice. METHODS S1-protein was administered either intranasally, intravenously or intracerebrally. We quantified aSyn immunoreactivity and phosphorylated aSyn (pS129), microglia and astrocyte reactivity, ACE2/Neuropilin-1 receptor expression, and parvalbumin-positive interneurons in limbic system, basal ganglia, and cortical regions 14 days post-application. Plasma cytokine profiles were assessed 6 days post-injection. RESULTS While intracerebral injection resulted in decreased aSyn immunoreactivity with increased pS129 in males, intravenous injection led to increased levels of aSyn immunoreactivity and microglia cell density, predominantly in brain regions associated with Parkinson's disease pathology. Intranasal application of S1-protein induced microgliosis in some brain regions but resulted in sex-dependent alterations of aSyn levels, with increases in females and decreases in males. All routes showed sex-dependent alterations in astrocytic reactivity, receptor expression, and parvalbumin-positive interneurons. CONCLUSION Our results demonstrate that S1-protein itself leads to neuroinflammation, altered aSyn homeostasis, and disruption of inhibitory circuits in a route- and sex-dependent manner. These findings indicate the possibility of S1-protein being a crucial agent for both neuroinflammatory processes and altered protein regulation mechanisms. S1-protein trapped in tissue reservoirs could therefore explain symptoms occurring or persisting beyond viral clearance (Post COVID-19 condition).
Collapse
Affiliation(s)
- Cara Sophie Schreiber
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover, (ZSN), Germany
| | - Lucas Navarro Ramil
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Juliette Bieligk
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Robert Meineke
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover, (ZSN), Germany.
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover, (ZSN), Germany.
| |
Collapse
|
10
|
Cuyàs B, Cantó E, Sanchez-Ardid E, Miró E, Alvarado-Tapias E, Román E, Poca M, Navarro F, Ferrero-Gregori A, Escorsell MÀ, Vidal S, Soriano G. In Vitro Immune Response of Mononuclear Cells to Multidrug-Resistant Escherichia coli. Microorganisms 2025; 13:1164. [PMID: 40431335 PMCID: PMC12114291 DOI: 10.3390/microorganisms13051164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/16/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025] Open
Abstract
Infections caused by multidrug-resistant organisms (MDRO) are linked to poor outcomes, particularly in patients with cirrhosis. The underlying mechanisms are not fully understood and may involve a different immune response against MDRO. This study aimed to compare the in vitro immune response between multidrug-resistant (MDR) Escherichia coli and antibiotic-susceptible E. coli strains. Surface protein extract and DNA extract were obtained from MDR E. coli (n = 6) and antibiotic-susceptible E. coli (n = 6) strains isolated from infected patients with cirrhosis. The extracts were used to stimulate in vitro peripheral blood mononuclear cells from healthy donors. After 48 h, cytokine levels (IFN-γ, IL-1β, IL-10, IL-12p70, MCP-1, IL-8, IL-6, MIP-1α, and MIP-1β) were measured. We observed no significant differences in cytokine production between MDR and susceptible strains. However, we identified notable interindividual variability in cytokine production for most of the cytokines studied. Only IFN-γ and IL-6 in surface extract and MCP-1 in DNA extract showed similar levels across all donors. We conclude that the cytokine profiles induced by MDR E. coli in vitro were similar to those in susceptible strains. These findings suggest that the poor prognosis associated with MDR E. coli infections is not due to a differential immune response but rather to other factors.
Collapse
Affiliation(s)
- Berta Cuyàs
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08023 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elisabet Cantó
- Inflammatory Diseases Department, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Elisabet Sanchez-Ardid
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elisenda Miró
- Department of Microbiology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain
- Department of Genetics and Microbiology, Institut de Recerca Sant Pau (IR Sant Pau), Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Edilmar Alvarado-Tapias
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08023 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva Román
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08023 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria Poca
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08023 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ferran Navarro
- Department of Microbiology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain
- Department of Genetics and Microbiology, Institut de Recerca Sant Pau (IR Sant Pau), Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Andreu Ferrero-Gregori
- Inflammatory Diseases Department, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Maria Àngels Escorsell
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08023 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Silvia Vidal
- Inflammatory Diseases Department, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - German Soriano
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, C/Mas Casanovas 90, 08041 Barcelona, Spain; (B.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08023 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
11
|
Jackson DM, Castanon-Cervantes O. Impaired Responses to In Vitro Lipopolysaccharide-Induced Stimulation After Long-Term, Rotating Shift Work. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:791. [PMID: 40427905 PMCID: PMC12110847 DOI: 10.3390/ijerph22050791] [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] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025]
Abstract
Shift work is a common labor practice affecting nearly 30% of the U.S. workforce. Long-term, rotating-shift work is particularly harmful to health. Persistent sleep deprivation in shift workers, among other factors, facilitates the development of a state of subclinical but chronic systemic inflammation with a high incidence and prevalence of infections and inflammation-related pathologies, suggesting an underlying disruption of immune responses. However, despite this state of chronic immune activation, cell-mediated inflammatory responses in rotating-shift workers are poorly understood. Here, we used lipopolysaccharide (LPS) to stimulate peripheral blood mononuclear cells (PBMCs) isolated from rotating-shift workers and healthy day-shift workers and investigate their immune responses. The results showed that PBMCs from rotating-shift workers had a dampened inflammatory response. Specifically, the secretion of LPS-induced TNF-α in culture supernatants was significantly reduced compared to the response found in PBMCs from day-shift workers. However, anti-inflammatory responses, reflected by the secretion of LPS-induced IL-10, were indistinguishable between PBMCs from day-shift and rotating-shift workers. In addition, the correlation between the plasma concentration of lipopolysaccharide-binding protein (LBP, a marker of systemic inflammation) and LPS-induced responses was disrupted only in rotating-shift workers, suggesting that in this group, an impaired mechanism that weakens the relationship between pro- and anti-inflammatory signaling may underlie the hypo-responsiveness of PBMCs. Our results suggest that persistent subclinical systemic inflammation in rotating-shift workers disrupts cell-mediated immunity, increasing the risk of infection and other inflammation-related pathologies in this population.
Collapse
Affiliation(s)
| | - Oscar Castanon-Cervantes
- Department of Neurobiology and Neuroscience Institute, Morehouse School of Medicine, 720 Westview DR SW, Atlanta, GA 30310, USA
| |
Collapse
|
12
|
Chen J, Xin B, Tan W, Chen Z, Zhang L, Zhu X. Genetic liability to critically ill COVID-19 increased risk of HER2-positive breast cancer through the immune pathway: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42372. [PMID: 40355230 PMCID: PMC12074039 DOI: 10.1097/md.0000000000042372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/18/2025] [Indexed: 05/14/2025] Open
Abstract
The clinical management of patients with both coronavirus disease 2019 (COVID-19) and breast cancer remains a complex and unresolved issue that despite extensive discussion has not reached a consensus. In contemporary literature, the association between COVID-19 and HER2-positive breast cancer has received minimal attention. Genetic instruments for severe acute respiratory syndrome coronavirus 2 infection (N = 2,597,856), hospitalized COVID-19 (N = 2,095,324), and critically ill COVID-19 (N = 1,086,211) were obtained from the COVID-19 Host Genetics Initiative genome-wide association study (GWAS) meta-analysis. A total of 103,530 HER2-positive breast cancer cases from GWAS were enrolled in our study. The summary GWAS statistics of 731 immune cells (N = 3757) were obtained from the MRCEU open database. Causal associations were evaluated by applying Mendelian randomization (MR) including inverse variance weighting, MR-Egger regression, and weighted-median analysis. Sensitivity analyses were employed, including Cochran Q test, MR-Egger intercept test, MR Pleiotropy Residual Sum and Outlier, and leave-one-out analysis, to examine the robustness of these findings. Genetic liability to critically ill COVID-19 was significantly causally associated with the increased risk of HER2-positive breast cancer (odds ratios = 1.086, 95% confidence intervals = 1.015-1.162, P = .016). No causal associations between severe acute respiratory syndrome coronavirus 2 infection or hospitalized COVID-19 and HER2-positive breast cancer were observed. Additionally, genetic liabilities to critically ill COVID-19 were significantly positively associated with 9 immune cells. IgD- CD27- AC and CD27 on IgD+ CD38- unsw mem (memory B cells) were identified as potential mediators of the causal relationship between critically ill COVID-19 and HER2-positive breast cancer. Genetic liability to critically ill COVID-19 is associated with an increased risk of HER2-positive breast cancer possibly through the immune pathway. Future studies are essential to elucidate the mechanisms underlying this causal relationship, aiming to develop therapeutic strategies to mitigate the immune cell-mediated impact on HER2-positive breast cancer risk.
Collapse
Affiliation(s)
- Jianfeng Chen
- Department of Toxicology II, Hunan Prevention and Treatment Institute for Occupational Diseases (Affiliated Prevention and Treatment Institute for Occupational Diseases of University of South China), Changsha, China
| | - Bingyan Xin
- Department of Toxicology II, Hunan Prevention and Treatment Institute for Occupational Diseases (Affiliated Prevention and Treatment Institute for Occupational Diseases of University of South China), Changsha, China
| | - Wenwen Tan
- Department of Occupational Lung Diseases II, Hunan Institute of Occupational Disease Prevention, Changsha, China
| | - Zhilian Chen
- Department of Toxicology II, Hunan Prevention and Treatment Institute for Occupational Diseases (Affiliated Prevention and Treatment Institute for Occupational Diseases of University of South China), Changsha, China
| | - Lan Zhang
- Department of Toxicology II, Hunan Prevention and Treatment Institute for Occupational Diseases (Affiliated Prevention and Treatment Institute for Occupational Diseases of University of South China), Changsha, China
| | - Xiaochuan Zhu
- Department of Toxicology II, Hunan Prevention and Treatment Institute for Occupational Diseases (Affiliated Prevention and Treatment Institute for Occupational Diseases of University of South China), Changsha, China
| |
Collapse
|
13
|
Hugaboom MB, Wirth LV, Street K, Ruthen N, Jegede OA, Schindler NR, Shah V, Zaemes JP, Ahmar NE, Matar S, Savla V, Choueiri TK, Denize T, West DJ, McDermott DF, Plimack ER, Sosman JA, Haas NB, Stein MN, Alter R, Bilen MA, Hurwitz ME, Hammers H, Signoretti S, Atkins MB, Wu CJ, Braun DA. Presence of Tertiary Lymphoid Structures and Exhausted Tissue-Resident T Cells Determines Clinical Response to PD-1 Blockade in Renal Cell Carcinoma. Cancer Discov 2025; 15:948-968. [PMID: 39992403 PMCID: PMC12048281 DOI: 10.1158/2159-8290.cd-24-0991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/08/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
SIGNIFICANCE We describe a paradigm wherein combined high TLS and low tissue-resident exhausted CD8+ T cells are required for optimal response to PD-1 blockade in RCC. This analysis identifies key determinants of response to PD-1 blockade in advanced RCC and suggests avenues for future immune modulation through rational combination therapy strategies.
Collapse
Affiliation(s)
- Miya B. Hugaboom
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Lena V. Wirth
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Street
- Division of Biostatistics, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Neil Ruthen
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Opeyemi A. Jegede
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Valisha Shah
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacob P. Zaemes
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Nourhan El Ahmar
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sayed Matar
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Varunika Savla
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Toni K. Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Destiny J. West
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Jeffrey A. Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Naomi B. Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark N. Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Robert Alter
- Hackensack University Medical Center, Hackensack, NJ, USA
| | - Mehmet A. Bilen
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael E. Hurwitz
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Hans Hammers
- Department of Internal Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Michael B. Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David A. Braun
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Song J, Pu Q, Chen C, Liu X, Zhang X, Wang Z, Yan J, Wang X, Wang H, Qian Q. Neurological Outcomes of Joint Exposure to Polystyrene Micro/Nanospheres and Silver Nanoparticles in Zebrafish. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:57007. [PMID: 40138633 PMCID: PMC12068508 DOI: 10.1289/ehp14873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Micro/nanoplastics and silver nanoparticles (AgNPs) are emerging environmental contaminants widely detected in aquatic environments. However, previous research has primarily focused on the interactions between micro/nanoplastics and organic substances or heavy metals, whereas the interactions and combined toxic effects of micro/nanoplastics with AgNPs remain unclear. OBJECTIVE Our study aimed to investigate the effects and mechanisms of coexposure to AgNPs and polystyrene micro/nanospheres (PS M/NPs) on the nervous system, comparing the toxicity of AgNPs alone and in combination with PS M/NPs in larval zebrafish. METHODS We investigated the dynamics of AgNPs' (5 nm ) adsorption onto PS M/NPs (5 μ m / 100 nm ) using inductively coupled plasma-mass spectrometry. Zebrafish larvae were coexposed to PS M/NPs (200 μ g / L ) and AgNPs (10 μ g / L ) from 6 h post fertilization (hpf) to 72 hpf to ∼ 120 hpf to evaluate neuroinflammatory effects from multiple perspectives, including developmental abnormalities, oxidative stress, neurobehavioral differences, vascular development, immune responses, differences in gene expression, and differences upon neuroinflammation inhibitor addition. RESULTS Adsorption experiments showed PS M/NPs could stably adsorb AgNPs, with higher adsorption in smaller particles. Zebrafish larvae exposed to combined PS M/NPs and AgNPs demonstrated neurodevelopmental abnormalities, including developmental malformations, lower levels of locomotor activity, delayed response, and abnormal neuronal development. In addition, exposed zebrafish also exhibited disrupted neurodevelopmental markers, including vascular and apoptotic indicators, and oxidative stress and neuroimmune responses. Quantitative real-time polymerase chain reaction analysis showed differences in gene expression within neurotoxic pathways in PS M/NPs and AgNPs-exposed zebrafish, focusing on key genes in immunity, apoptosis, vascular, and neural development. Furthermore, these neurotoxic effects induced by combined exposure were alleviated following the introduction of the neuroinflammation inhibitor curcumin. DISCUSSION Our findings demonstrate that polystyrene nanospheres (PSNPs) intensified AgNPs-induced neurotoxicity in larval zebrafish, whereas polystyrene microspheres (PSMPs) had a lesser effect, indicating distinct gene regulation roles when combined with AgNPs. These findings enhance the assessment of environmental risks in settings with coexisting nanomaterials and microplastics, offering important insights for evaluating combined exposure risks. https://doi.org/10.1289/EHP14873.
Collapse
Affiliation(s)
- Jie Song
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Qian Pu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Chen Chen
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Xingcheng Liu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Xinlei Zhang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Zejun Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Jin Yan
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| | - Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, China
| |
Collapse
|
15
|
Huang D, Dai J, Yu H, Chen W. Immune dysregulation in endometrial tuberculosis: elevated HLA-G and IL-1Ra as key modulators. Front Cell Infect Microbiol 2025; 15:1548238. [PMID: 40375897 PMCID: PMC12078264 DOI: 10.3389/fcimb.2025.1548238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/03/2025] [Indexed: 05/18/2025] Open
Abstract
Endometrial tuberculosis (ETB) is a reproductive system infection caused by Mycobacterium tuberculosis, primarily invading the endometrium through hematogenous dissemination. This study included 10 patients diagnosed with ETB and 10 patients with pulmonary tuberculosis (PTB) to analyze their clinical, pathological, and immunological characteristics. Anatomically, PTB presented the highest prevalence among tuberculosis cases. Compared to PTB imaging, CT scans of ETB showed less distinctive diagnostic features. Pathologically, abscess formation was more frequently observed in ETB patients than in PTB patients, suggesting a more intense local inflammatory response in ETB. However, there were no statistically significant differences in granulomatous lesions, caseous necrosis, coagulative necrosis, inflammatory necrosis, exudation, acute inflammation, or fibrous tissue hyperplasia between the two groups. Immunohistochemical analysis revealed higher infiltration of macrophages (CD68) in ETB lesions compared to PTB, whereas the counts of T cells (CD3+, CD4+, CD8+) and B cells (CD20) showed no significant differences. Notably, the expression levels of HLA-G and IP-10 were significantly elevated in the lesion areas of ETB compared to PTB. Similarly, the expression of HLA-G, IP-10, IL-1Ra, and IL-10 was significantly higher in the ETB group than in the PTB group. Furthermore, HLA-G and IL-1Ra expression levels were markedly elevated in ETB lesion areas compared to surrounding normal endometrial tissue. HLA-G plays a pivotal role in immune tolerance by modulating local immune responses, while IP-10 is involved in chronic inflammatory signaling. IL-1Ra and IL-10 are key regulators of endometrial immune homeostasis, counterbalancing inflammatory responses that could otherwise disrupt reproductive function. These immunoregulatory factors are crucial in maintaining immune tolerance within the endometrium and may influence immune responses associated with endometrial tuberculosis.
Collapse
Affiliation(s)
- Dan Huang
- Graduate School of Hebei North University, Zhangjiakou, China
- Department of Reproductive Medicine Clinic, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinlong Dai
- Department of Pathology, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Haotian Yu
- Department of Reproductive Medicine Clinic, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Sausa M, Paladino L, Scalia F, Zummo FP, Vergilio G, Rappa F, Cappello F, Gratie MI, Proia P, Di Felice V, Marino Gammazza A, Macaluso F, Barone R. Lactobacillus fermentum LF31 Supplementation Reversed Atrophy Fibers in a Model of Myopathy Through the Modulation of IL-6, TNF-α, and Hsp60 Levels Enhancing Muscle Regeneration. Nutrients 2025; 17:1550. [PMID: 40362856 PMCID: PMC12073311 DOI: 10.3390/nu17091550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Recent studies have highlighted the role of the gut-muscle axis, suggesting that modulation of the gut microbiota may indirectly benefit skeletal muscle. This study aimed to evaluate the effects of Lactobacillus fermentum (L. fermentum) supplementation in a model of muscle atrophy induced by chronic ethanol (EtOH) intake, focusing on inflammatory and antioxidant mechanisms. Methods: Sixty 12-month-old female Balb/c mice were divided randomly into three groups (n = 20/group): (1) Ethanol (EtOH) group, receiving ethanol daily for 8 and 12 weeks to induce systemic oxidative stress and inflammation; (2) Ethanol + Probiotic (EtOH + P) group, receiving both ethanol and L. fermentum supplementation for the same durations; and (3) Control (Ctrl) group, receiving only water. Muscle samples were analyzed for the fiber morphology, inflammatory markers, oxidative stress indicators, and satellite cell (SC) activity. All data were tested for normality using the Shapiro-Wilk test before applying a parametric analysis. A statistical analysis was performed using one-way ANOVA followed by a Bonferroni post-hoc test. The level of significance was set at p < 0.05. Results: EtOH exposure caused significant atrophy in all muscle fiber types (type I, IIa, and IIb), with the most pronounced effects on oxidative fibers. L. fermentum supplementation significantly reversed atrophy in type I and IIa fibers, accompanied by a significant reduction in IL-6, TNF-α, and Hsp60 expression levels, indicating the protective effect of L. fermentum against oxidative stress and inflammation. Moreover, the probiotic treatment increased MyoD expression in SCs, suggesting enhanced regenerative activity, without histological evidence of fibrosis. Conclusions: These findings suggest that L. fermentum supplementation could counteract EtOH-induced skeletal muscle damage by reducing inflammation and oxidative stress and promoting muscle repair, indicating its potential as an adjuvant, in the therapeutic strategy of models of muscle degeneration.
Collapse
Affiliation(s)
- Martina Sausa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Letizia Paladino
- Department of Theoretical and Applied Sciences, eCampus University, 22060 Novedrate, Italy;
| | - Federica Scalia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Francesco Paolo Zummo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Giuseppe Vergilio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Francesca Rappa
- The Institute of Translational Pharmacology, National Research Council of Italy (CNR), 90146 Palermo, Italy;
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Melania Ionelia Gratie
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Patrizia Proia
- Department of Psychology, Educational Science and Human Movement, University of Palermo, 90128 Palermo, Italy;
| | - Valentina Di Felice
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90133 Palermo, Italy
| | - Filippo Macaluso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| | - Rosario Barone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy; (M.S.); (F.S.); (F.P.Z.); (G.V.); (F.C.); (M.I.G.); (V.D.F.); (A.M.G.); (R.B.)
| |
Collapse
|
17
|
Demetriou A, Lindqvist B, Ali HG, Shamekh MM, Varshney M, Inzunza J, Maioli S, Nilsson P, Nalvarte I. ERβ mediates sex-specific protection in the App-NL-G-F mouse model of Alzheimer's disease. Biol Sex Differ 2025; 16:29. [PMID: 40302001 PMCID: PMC12039102 DOI: 10.1186/s13293-025-00711-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Menopausal loss of neuroprotective estrogen is thought to contribute to the sex differences in Alzheimer's disease (AD). Activation of estrogen receptor beta (ERβ) can be clinically relevant since it avoids the adverse systemic effects of ERα activation. However, very few studies have explored ERβ-mediated neuroprotection in AD, and no information on its contribution to the sex differences in AD exists. In the present study, we specifically explored the role of ERβ in mediating sex-specific protection against AD pathology in the AppNL-G-F knock-in mouse model of amyloidosis, and if surgical menopause (ovariectomy) modulates pathology in this model. METHODS We treated male and female AppNL-G-F knock-in mice with the clinically relevant and selective ERβ agonist LY500307. A subset of the females was ovariectomized prior to treatment. Y-maze and contextual fear conditioning tests were used to assess memory performance, and biochemical assays such as qPCR, immunohistochemistry, Western blot, and multiplex immunoassays, were used to evaluate amyloid pathology. RESULTS We found that Female AppNL-G-F mice had higher soluble Aβ levels in cortex and hippocampus than males and more activated microglia. ERβ activation protected against amyloid pathology and cognitive decline in both male and female AppNL-G-F mice. Although ovariectomy increased soluble amyloid beta (Aβ) in cortex and insoluble Aβ in hippocampus, as well as sustained neuroinflammation after ERβ activation, it had otherwise limited effects on pathology. We further identified that ERβ did not alter APP processing, but rather exerted its protection at least partly via microglia activation in a sex-specific manner. CONCLUSION Combined, we provide new understanding to the sex differences in AD by demonstrating that ERβ protects against AD pathology differently in males and females, warranting reassessment of ERβ in combating AD.
Collapse
Affiliation(s)
- Aphrodite Demetriou
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
| | - Birgitta Lindqvist
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Heba G Ali
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mohamed M Shamekh
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Biochemistry, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Mukesh Varshney
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Jose Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52, Huddinge, Sweden
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden
| | - Ivan Nalvarte
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Visionsgatan 4, J9:20, 171 64, Solna, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 57, Huddinge, Sweden.
| |
Collapse
|
18
|
Álvarez-Mercado AI, López-Plaza B, Plaza-Diaz J, Arcos-Castellanos L, Ruiz-Ojeda FJ, Brandimonte-Hernández M, Feliú-Batlle J, Hummel T, Palma-Milla S, Gil Á. Miraculin Can Contribute to a Reduction in Inflammatory Biomarkers and Cachexia in Malnourished Patients with Cancer and Taste Disorders. Pharmaceuticals (Basel) 2025; 18:622. [PMID: 40430444 PMCID: PMC12115146 DOI: 10.3390/ph18050622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background: In 2022, there were an estimated 20 million new cancer cases and 9.7 million deaths. The number of new cancer cases is expected to rise to over 35 million by 2050, marking a 75% increase from 2022 levels. Twenty to eighty-six percent of cancer patients suffer from taste disorders (TD), which are associated with an increased risk of malnutrition. Cachectic syndrome is linked to the presence and growth of tumors and leads to systemic inflammation. Synsepalum dulcificum is a plant whose berries contain miraculin, a glycoprotein that transforms sour tastes into sweet and can ameliorate TD. Objectives: To evaluate the effect of the regular intake of dried miracle berries (DMBs), a novel food containing miraculin, on biomarkers of inflammation and cachexia in malnourished patients with cancer and TD receiving systemic antineoplastic therapy. Methods: we conducted a triple-blind, randomized, placebo-controlled pilot clinical trial. Thirty-one patients with cancer of various etiologies who received chemotherapy were enrolled in this pilot study and divided into three groups. The first group received a tablet containing 150 mg of DMB (standard dose), the high-dose group received a tablet of 300 mg of DMB, and the third group received a tablet with 300 mg of the placebo for three months before each main meal. The plasma levels of several molecules associated with inflammation and cancer cachexia were measured using the X-MAP Luminex multiplexing platform. Results: We found decreased plasma levels of IFN-γ in the standard-dose group. In addition, our results suggest a downtrend of IL-1β levels in the three groups after three months of intervention (p = 0.093). Moreover, the three groups showed a reduction in tumor-derived molecule proteolysis-inducing factor/dermcidin (p = 0.021). It is important to highlight the positive correlation between IL-6 and IL-10 in the standard group, which suggests a better balance between proinflammatory and anti-inflammatory cytokines. Regardless of DMB consumption, soluble TNF receptor type II tended to decrease with treatment in patients who responded well to the antineoplastic treatment (p = 0.011). We did not find significant correlations between cytokines and sensory variables or dietary and nutritional status. Conclusions: Our results suggest that the regular consumption of a standard dose of DMB along with a systemic antineoplastic treatment could contribute to reducing inflammation and cachexia biomarkers in malnourished patients with cancer exhibiting TD. In this sense, nutritional support is crucial in the treatment of cancer cachexia. In our view, it should be considered a coadjuvant of therapeutics. Future studies on the molecular signaling pathways and specific mechanisms of action of bioactive compounds within food supplements, such as miraculin, will allow them to be used to target pathogenic mechanisms of cancer cachexia and malnutrition: NCT05486260.
Collapse
Affiliation(s)
- Ana Isabel Álvarez-Mercado
- Department of Pharmacology, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain (Á.G.)
- Centre of Biomedical Research, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
| | - Bricia López-Plaza
- Food, Nutrition and Health Platform, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Medicine Department, Faculty of Medicine, Complutense University of Madrid, Plaza de Ramón y Cajal, s/n, 28040 Madrid, Spain
| | - Julio Plaza-Diaz
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain (Á.G.)
- Department of Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain
- School of Health Sciences, Universidad Internacional de La Rioja, Avenida de la Paz, 137, 26006 Logrono, Spain
| | - Lucía Arcos-Castellanos
- Food, Nutrition and Health Platform, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Medicine Department, Faculty of Medicine, Complutense University of Madrid, Plaza de Ramón y Cajal, s/n, 28040 Madrid, Spain
| | - Francisco Javier Ruiz-Ojeda
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain (Á.G.)
- Centre of Biomedical Research, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
- RU Adipocytes and Metabolism, Helmholtz Diabetes Center at Helmholtz Munich, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Marco Brandimonte-Hernández
- Centre of Biomedical Research, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain
| | - Jaime Feliú-Batlle
- Oncology Department, Hospital La Paz Institute for Health Research—IdiPAZ, Hospital Universitario La Paz, 28029 Madrid, Spain
- CIBERONC (CIBER Cancer), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Medicine Department, Faculty of Medicine, Autonomous University of Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Thomas Hummel
- Medicine Department, Faculty of Medicine, Autonomous University of Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain
- Smell & Taste Clinic, Department of Otorhinolaryngology, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Samara Palma-Milla
- Food, Nutrition and Health Platform, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Nutrition Department, Hospital University La Paz, 28046 Madrid, Spain
| | - Ángel Gil
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain (Á.G.)
- Centre of Biomedical Research, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
19
|
Fisher JG, Bartlett LG, Kashyap T, Walker CJ, Khakoo SI, Blunt MD. Modulation of anti-tumour immunity by XPO1 inhibitors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002310. [PMID: 40291981 PMCID: PMC12022495 DOI: 10.37349/etat.2025.1002310] [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: 01/13/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Exportin-1 (XPO1) is a nuclear export protein that, when overexpressed, can facilitate cancer cell proliferation and survival and is frequently overexpressed or mutated in cancer patients. As such, selective inhibitors of XPO1 (XPO1i) function have been developed to inhibit cancer cell proliferation and induce apoptosis. This review outlines the evidence for the immunomodulatory properties of XPO1 inhibition and discusses the potential for combining and sequencing XPO1i with immunotherapy to improve the treatment of patients with cancer. Selinexor is a first-in-class XPO1i that is FDA-approved for the treatment of patients with relapsed and refractory (RR) multiple myeloma and RR diffuse large B cell lymphoma. In addition to the cancer cell intrinsic pro-apoptotic activity, increasing evidence suggests that XPO1 inhibition has immunomodulatory properties. In this review, we describe how XPO1i can lead to a skewing of macrophage polarisation, inhibition of neutrophil extracellular traps, modulation of immune checkpoint expression, blockade of myeloid-derived suppressor cells (MDSCs) and sensitisation of cancer cells to T cell and NK (natural killer) cell immunosurveillance. As such, there is an opportunity for selinexor to enhance immunotherapy efficacy and thus a need for clinical trials assessing selinexor in combination with immunotherapies such as immune checkpoint inhibitors, direct targeting monoclonal antibodies, chimeric antigen receptor (CAR)-T cells and cereblon E3 ligase modulators (CELMoDs).
Collapse
Affiliation(s)
- Jack G. Fisher
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | - Laura G. Bartlett
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | | | | | - Salim I. Khakoo
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | - Matthew D. Blunt
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| |
Collapse
|
20
|
Jolink TA, Feldman MJ, Antenucci NM, Cardenas MN, West TN, Nakamura ZM, Muscatell KA. Effects of a mild inflammatory challenge on cytokines and sickness behavior: A randomized controlled trial using the influenza vaccine. Brain Behav Immun 2025; 128:429-439. [PMID: 40239903 DOI: 10.1016/j.bbi.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/27/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025] Open
Abstract
The influenza vaccine has reliably been associated with mild, within-person increases in inflammation. However, the field lacks rigorous experimental work comparing the effects of the influenza vaccine to a placebo control on changes in plasma inflammatory cytokines and self-reported sickness behavior. In a double-blind, randomized, placebo-controlled trial, 102 participants received either the influenza vaccine or saline placebo. Four cytokines were measured in plasma 24-hours following injection; participants also reported on psychosocial outcomes, specifically sickness behavior, positive/negative affect, sleep, and subjective social disconnection. All cytokines-IL-6, IL-10, TNF-α, IFN-γ-were significantly increased in the influenza vaccine condition compared to placebo. None of the psychosocial outcomes differed by condition. This study fills a gap in the literature by presenting critical causal evidence that the influenza vaccine leads to elevated levels of four inflammatory cytokines, compared to placebo control. However, a more robust increase in inflammation or a larger sample size may be necessary to observe differences in self-reported sickness behavior and other psychosocial outcomes.
Collapse
Affiliation(s)
- Tatum A Jolink
- University of North Carolina at Chapel Hill, Department of Psychology & Neuroscience, United States; University of Michigan, Department of Psychology, United States.
| | - Mallory J Feldman
- University of North Carolina at Chapel Hill, Department of Psychology & Neuroscience, United States.
| | - Natalie M Antenucci
- University of North Carolina at Chapel Hill, Department of Psychology & Neuroscience, United States
| | - Megan N Cardenas
- University of North Carolina at Chapel Hill, Department of Psychology & Neuroscience, United States
| | - Taylor N West
- University of North Carolina at Chapel Hill, Department of Psychology & Neuroscience, United States
| | - Zev M Nakamura
- University of North Carolina at Chapel Hill, Department of Psychiatry, United States
| | - Keely A Muscatell
- University of North Carolina at Chapel Hill, Department of Psychology & Neuroscience, United States; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, United States; Carolina Population Center, University of North Carolina at Chapel Hill, United States
| |
Collapse
|
21
|
Gheorghiu M, Trandafir MF, Savu O, Pasarica D, Bleotu C. Unexpectedly High and Difficult-to-Explain Regenerative Capacity in an 82-Year-Old Patient with Insulin-Requiring Type 2 Diabetes and End-Stage Renal Disease. J Clin Med 2025; 14:2556. [PMID: 40283387 PMCID: PMC12027714 DOI: 10.3390/jcm14082556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The case we present is part of a large study that we conducted on hemodialysis patients with type 2 diabetes mellitus (T2DM) and which set the following objectives: studying changes in the intestinal microbiota, innate and acquired immune response capacity, and tissue regeneration. Methods: (1) For the genetic study of the gut microbiota, special techniques that are not based on cultivation were used since most of the species in the intestinal flora are not cultivable. (2) The immunological study had two targets: innate immunity (inflammation) and adaptive immunity (we chose to address the cellular immune response because, unlike the humoral one, it is insufficiently studied in this category of associated pathologies). As markers for innate immunity (inflammation), the following were determined: IL-6, sIL-6R, IL-1β, TNFα, IL-10, and NGAL. TNFβ/LTα was determined as a marker for adaptive immunity (the cellular immune response). (3) The study of tissue regeneration capacity was performed using NT-3 (this is the first study to do so) and VEGFβ (another marker that is scarce in this category of patients) as markers. All the aforementioned compounds were determined from serum samples, utilizing Merck Millipore ELISA kits for IL-6, IL-1β, IL-10, NT-3, and VEGF β, and Elabscience ELISA kits for IL-6R, TNFα, TNFβ, and NGAL. Results: We were very surprised to find unexpected immunological changes and tissue regenerative capacity in one of the patients studied, an 82-year-old female patient diagnosed with insulin-dependent T2DM with multiple complications, including end-stage renal disease (ESRD). The patient showed a huge capacity for tissue regeneration, combined with amplification of immunological capacity, in comparison to patients in the same group (T2DM and ESRD) and to those in the control group (ESRD). Thus, extremely elevated serum concentrations of IL-1β, IL-6, IL-10, and TNF-β, as well as the tissue regeneration indicators NT-3 and VEGFβ, were obtained in comparison to all other members of the patient group. At the same time, serum levels of the soluble IL-6 receptor (sIL6-R) and TNFα were greatly reduced compared to the test group's mean. Conclusions: All the data obtained during our research were corroborated with those from the specialized literature and entitle us to support the hypothesis that the cause of these unexpected behaviors is the genetically conditioned overproduction (possibly acquired post-infection) of IL-6, along with its predominant anti-inflammatory and pro-regenerative signaling through the membrane-bound receptor IL-6R.
Collapse
Affiliation(s)
- Mihaela Gheorghiu
- Pathophysiology and Immunology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.-F.T.); (O.S.); (D.P.)
| | - Maria-Florina Trandafir
- Pathophysiology and Immunology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.-F.T.); (O.S.); (D.P.)
| | - Octavian Savu
- “N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 020475 Bucharest, Romania
- Doctoral School of “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Daniela Pasarica
- Pathophysiology and Immunology Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.-F.T.); (O.S.); (D.P.)
| | - Coralia Bleotu
- “Stefan S. Nicolau” Institute of Virology, 030304 Bucharest, Romania;
| |
Collapse
|
22
|
Marchesi N, Allegri M, Bruno GM, Pascale A, Govoni S. Exploring the Potential of Dietary Supplements to Alleviate Pain Due to Long COVID. Nutrients 2025; 17:1287. [PMID: 40219044 PMCID: PMC11990457 DOI: 10.3390/nu17071287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Long COVID, characterized by persistent symptoms following COVID-19 infection, significantly impacts individuals' health and daily functioning due to fatigue and pain. Focusing on pain, this review addresses nociplastic and chronic pain conditions. Interventions designed to reduce inflammation, oxidative stress, and enhance vagal activity may offer a promising approach to managing post-pandemic pain. This review presents individual components of food supplements with demonstrated efficacy in one or more pain conditions, focusing on their proposed mechanisms and clinical activity in pain, including their use in post-COVID-19 pain when available. Many of these substances have a long history of safe use and may offer an alternative to long-term analgesic drug treatment, which is often associated with potential side effects. This review also explores the potential for synergistic effects when combining these substances with each other or with conventional analgesics, considering the advantages for both patients and the healthcare system in using these substances as adjunctive or primary therapies for pain symptoms related to long COVID. While preclinical scientific literature provides a mechanistic basis for the action of several food supplements on pain control mechanisms and signaling pathways, clinical experience, particularly in the field of long COVID-associated pain, is still limited. However, the reviewed literature strongly suggests that the use of food supplements in long COVID-associated pain is an attainable goal, provided that rigorous clinical trials are conducted.
Collapse
Affiliation(s)
- Nicoletta Marchesi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
- RedyNeuheart s.r.l., Start-Up, Via Santa Marta 19, 20123 Milan, Italy
| | - Massimo Allegri
- Centre Lémanique de Neuromodulation et Thérapie de la Douleur, Hôpital de Morges, Ensemble Hospitalier de la Côte (EHC), 1110 Morges, Switzerland;
| | - Giacomo Matteo Bruno
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
- Center of Research, SAVE Studi—Health Economics and Outcomes Research, 20123 Milan, Italy
- CEFAT (Center of Pharmaceuticals Economics and Medical Technologies Evaluation), University of Pavia, 27100 Pavia, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, 27100 Pavia, Italy; (G.M.B.); (A.P.); (S.G.)
- CEFAT (Center of Pharmaceuticals Economics and Medical Technologies Evaluation), University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
23
|
Giácomo ALD, Azcurra LN, García GR, Raviolo JM, Grosso MC, Dogi CA, Pereyra MLG. Bacillus velezensis MFF 2.2-derived lipopeptides modulate gut immune response in BALB/c mice maintaining mucosal integrity and systemic safety. Vet Res Commun 2025; 49:159. [PMID: 40192874 DOI: 10.1007/s11259-025-10730-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025]
Abstract
Bacillus-derived lipopeptides, particularly surfactin (SF), are known for their antimicrobial and immunomodulatory properties, making them promising candidates for applications in veterinary medicine. This study investigated the immunomodulatory, cytotoxic, and genotoxic effects of a lipopeptide extract (LPE) derived from Bacillus velezensis MFF 2.2, containing SF, using an in vivo murine model. Four groups of BALB/c mice (n = 6 per group) were orally administered daily doses of: (1) saline solution (control), (2) 60 μg/mL SF standard, (3) 40 μg/mL LPE, and (4) 60 μg/mL LPE, over a 10-day period. Assessments included changes in body weight, histopathological analysis of liver, kidney, and small intestine tissues, as well as evaluation of cytotoxic and genotoxic effects. LPE treatment was well-tolerated, with no significant changes in body weight, organ histopathology, or bone marrow toxicity. Both SF and LPE significantly increased goblet cell counts (P < 0.05), while LPE reduced IFN-γ levels and increased the IL- 10/IFN-γ ratio, suggesting an anti-inflammatory response (P < 0.05). TNF-α and IL- 10 levels remained unchanged, and no significant differences were observed in intraepithelial lymphocyte counts. These findings suggest that LPE derived from B. velezensis MFF 2.2 is safe and promotes immunomodulatory effects, supporting its potential as a therapeutic agent in inflammatory conditions.
Collapse
Affiliation(s)
- Ana Lucía Di Giácomo
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
- Instituto de Ciencias Veterinarias (INCIVET-CONICET-UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
| | - Lorena Nadia Azcurra
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
| | - Gisela Romina García
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
- Instituto de Ciencias Veterinarias (INCIVET-CONICET-UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
| | - José María Raviolo
- Instituto de Ciencias Veterinarias (INCIVET-CONICET-UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
- Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
| | - María Carolina Grosso
- Instituto de Ciencias Veterinarias (INCIVET-CONICET-UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
- Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
| | - Cecilia Ana Dogi
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
- Instituto de Ciencias Veterinarias (INCIVET-CONICET-UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina
| | - María Laura González Pereyra
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina.
- Instituto de Ciencias Veterinarias (INCIVET-CONICET-UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ruta N 36 Km 601, Río Cuarto City, 5800, Córdoba, Argentina.
| |
Collapse
|
24
|
Morisaki M, Rezaali F, Lau LC, Boche D, Khandaker GM, Turecki G, Sinclair LI. Depression as a risk factor for Alzheimer's disease: A human post-mortem study. PLoS One 2025; 20:e0320561. [PMID: 40179065 PMCID: PMC11967934 DOI: 10.1371/journal.pone.0320561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Depression is associated with persistent low mood. In mid to late life, it has been identified as a risk factor for Alzheimer's disease (AD) with evidence that depression might be an early manifestation of AD. Although the underlying mechanisms by which depression enhances AD development remain unknown, there are several features commonly seen in both diseases such as the presence of neuroinflammation. In this study, we aimed to identify whether neuroinflammation is increased in depression as observed in the early stages of AD by examining post-mortem human brain tissue. Post-mortem human brain tissue from 54 cases with depression and 37 controls without depression were retrieved from the Douglas Bell Canada Brain Bank. Sixteen early-stage AD cases defined as a Braak stage III-IV and 15 controls were sourced from the South West Dementia Brain Bank. Frozen tissue from the dorsal prefrontal cortex was obtained for all cases in order to measure inflammatory proteins (IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, and TNF-α) and endothelial markers (ICAM-1, VCAM) using ELISA and MesoScale Multiplex Assays. In the depression group, increase of IL-6 and IL-10, and decrease of IL-1β were observed compared to controls, with no changes detected for the other cytokines and the endothelial markers. In early-stage AD cases, only increased ICAM-1 expression was found compared to controls, indicating endothelial activation as an early feature of AD. None of the cytokines measured showed alteration of their expression in early-stage AD cases. Depression, but not AD, was associated with evidence of neuroinflammation. Depression may increase AD risk through different mechanism(s) than inflammation.
Collapse
Affiliation(s)
- Mizuki Morisaki
- Dementia Research Group, University of Bristol, Bristol, United Kingdom
| | - Farnoosh Rezaali
- Dementia Research Group, University of Bristol, Bristol, United Kingdom
| | - Laurie C. Lau
- Clinical and Experimental Sciences, Faculty of Medicine, Sir Henry Wellcome Laboratories, University of Southampton, Southampton, United Kingdom
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Golam M. Khandaker
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, Bristol, United Kingdom
| | - Gustavo Turecki
- Douglas Bell Brain Bank, McGill University, Montreal, Canada
| | | |
Collapse
|
25
|
Ernst LM, Freedman AA, Odom‐Konja RM, Keenan‐Devlin L, Miller GE, Cole S, Crockett A, Borders A. Associations of Serum Inflammatory Biomarkers During Pregnancy With Placental Pathology and Placental Gene Expression at Delivery. Am J Reprod Immunol 2025; 93:e70062. [PMID: 40173254 PMCID: PMC11964289 DOI: 10.1111/aji.70062] [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: 09/09/2024] [Revised: 02/05/2025] [Accepted: 02/19/2025] [Indexed: 04/04/2025] Open
Abstract
PROBLEM We sought to investigate whether maternal inflammatory cytokines during pregnancy are associated with histologic inflammatory or vascular lesions in the placenta and/or correlated with gene expression patterns in the placenta. METHOD OF STUDY We leveraged data from a large randomized controlled trial (RCT) at a single site. Maternal serum was collected in the second and third trimesters, and a composite inflammatory score was created using five measured biomarkers (CRP, IL-6, IL-1ra, IL-10, and TNF-α). Placentas were collected at delivery for histological analysis and four major patterns of placental injury were characterized. Fresh small chorionic villous biopsies were collected for placental genome-wide mRNA profiling. Transcripts showing >2-fold differential expression over the 4-SD range of circulating inflammatory biomarkers were reported, adjusting for potential confounders. RESULTS The primary analysis included 601 participants. A one standard deviation increase in the third-trimester inflammatory composite was associated with increased odds of chronic inflammation in the placenta (OR: 1.23, 95% CI 1.01, 1.51;). This was driven primarily by elevations in IL-10 (OR: 1.37; 99% CI: 1.06, 1.77). Higher maternal IL-10 in circulation was associated with bioinformatic indications of reduced pro-inflammatory gene regulation pathways in the placenta (AP1 decreased 25%, p = 0.003; NF-kB decreased 53%, p = 0.003) and indications of increased STAT family signaling pathways which mediate signaling through the IL-10 receptor (increased 73%, p = 0.002). CONCLUSIONS Our results indicate that elevated maternal circulating IL-10 during pregnancy is associated with chronic inflammatory lesions in the placenta at delivery. Additionally, higher levels of circulating IL-10 are associated with upregulated STAT signaling pathways in placental tissues.
Collapse
Affiliation(s)
- Linda M. Ernst
- Department of Pathology and Laboratory MedicineEndeavor HealthEvanstonIllinoisUSA
- Department of PathologyUniversity of Chicago Pritzker School of MedicineChicagoIllinoisUSA
| | - Alexa A. Freedman
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | | | - Lauren Keenan‐Devlin
- Department of Obstetrics and GynecologyEndeavor HealthEvanstonIllinoisUSA
- Department of Obstetrics and GynecologyUniversity of Chicago Pritzker School of MedicineChicagoIllinoisUSA
| | - Gregory E. Miller
- Department of Psychology and Institute for Policy ResearchNorthwestern UniversityEvanstonIllinoisUSA
| | - Steve Cole
- Department of Medicine, Division of Hematology‐Oncology, and Department of Psychiatry and Biobehavioral SciencesUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Amy Crockett
- Department of Obstetrics and GynecologyPrisma HealthGreenvilleSouth CarolinaUSA
| | - Ann Borders
- Department of Obstetrics and GynecologyEndeavor HealthEvanstonIllinoisUSA
- Department of Obstetrics and GynecologyUniversity of Chicago Pritzker School of MedicineChicagoIllinoisUSA
| |
Collapse
|
26
|
Zhang X, Xu Z, Liu Y. The role of serun lipid, cytokine production in sudden sensorineural hearing loss. Cytotechnology 2025; 77:67. [PMID: 40012925 PMCID: PMC11850693 DOI: 10.1007/s10616-025-00722-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Sudden sensorineural hearing loss (SSNHL) has serious harm to human hearing health, where blood lipid and inflammatory levels may play a key role in it. Thus, the purpose of this investigation was to assess the connection between inflammatory and lipid variables and SSNHL. Patients diagnosed with SSNHL had an analysis of serum lipid parameters, such as total cholesterol (TC), triglycerides, HDL-C, LDL-C, apolipoprotein A (apo A), apolipoprotein B (apo B), and lipoprotein A (Lp(a)), as well as inflammatory factors like TNF-α and IL-10. After that, risk factor analysis was carried out utilizing univariate, multivariate regression, and LASSO retrospective modeling. In all, 72 SSNHL patients and 67 healthy control individuals were involved. The LDL/HDL, total cholesterol, ApoB, LP(a), IL-10, TNF-α, and IFN-γ considerably higher in the SSNHL group than in the healthy control group, however, nervonic acid and coenzyme Q were decreased significantly in SSNHL than Control group. The multivariate logistic regression model's analysis using multifactorial retrospective modeling revealed significant changes in LDL, LDL/HDL, IL-10, and TNF-α. In addition, in the LASSO regression model, the model demonstrated high discrimination, as evidenced by the C-index for the cohort's prediction nomogram, which was 0.998 (95% CI, 0.154-1.115) and confirmed to be 0.925 following bootstrapping validation. Finally, IL-10 and LDL/HDL were the main risk factors in SSNHL. LDL/HDL and IL-10 may be closely related to SSNHL's progress and should be evaluated promptly before treating patients with SSNHL.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- Department of Otorhinolaryngology—Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Otorhinolaryngology, The Third Affiliated Hospital of Anhui Medical
University (The First People’s Hospital of Hefei), Hefei, P.R. China
| | - Zhihua Xu
- Department of Otorhinolaryngology, The Third Affiliated Hospital of Anhui Medical
University (The First People’s Hospital of Hefei), Hefei, P.R. China
| | - Yehai Liu
- Department of Otorhinolaryngology—Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Jarneborn A, Kopparapu PK, Jin T. The Dual-Edged Sword: Risks and Benefits of JAK Inhibitors in Infections. Pathogens 2025; 14:324. [PMID: 40333091 PMCID: PMC12030494 DOI: 10.3390/pathogens14040324] [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: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025] Open
Abstract
Janus kinase inhibitors (JAKis) represent a relatively new class of immunomodulatory drugs with potent effects on various cytokine signalling pathways. They have revolutionized the treatment landscape for autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, and ulcerative colitis. However, their ability to modulate immune responses presents a dual-edged nature, influencing both protective immunity and pathological inflammation. This review explores the complex role of JAKis in infectious settings, highlighting both beneficial and detrimental effects. On the one hand, experimental models suggest that JAK inhibition can impair host defence mechanisms, increasing susceptibility to certain bacterial and viral infections. For example, tofacitinib-treated mice exhibited more severe joint erosions in Staphylococcus aureus (S. aureus) septic arthritis and showed impaired viral clearance in herpes simplex encephalitis. Additionally, clinical data confirm an increased risk of herpes zoster in patients receiving JAKis, underscoring the need for rigorous monitoring. On the other hand, JAK inhibition has demonstrated protective effects in certain infectious and hyperinflammatory conditions. In sepsis models, including cecal ligation and puncture (CLP) and S. aureus bacteraemia, tofacitinib improved survival by attenuating excessive inflammation. Furthermore, JAKis, particularly baricitinib, have shown substantial efficacy in mitigating cytokine storms during severe COVID-19 infections, leading to improved clinical outcomes and reduced mortality. These observations suggest that JAKis have a role in modulating hyperinflammatory responses in select infectious contexts. In conclusion, JAKis present a complex interplay between immunosuppression and immunomodulation. While they increase the risk of certain infections, they also show potential in managing hyperinflammatory conditions such as cytokine storms. The key challenge is determining which patients and situations benefit most from JAKis while minimizing risks, requiring a careful and personalized treatment approach.
Collapse
Affiliation(s)
- Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (P.K.K.); (T.J.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (P.K.K.); (T.J.)
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (P.K.K.); (T.J.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| |
Collapse
|
28
|
Xu J, Wang B, Ao H. Corticosterone effects induced by stress and immunity and inflammation: mechanisms of communication. Front Endocrinol (Lausanne) 2025; 16:1448750. [PMID: 40182637 PMCID: PMC11965140 DOI: 10.3389/fendo.2025.1448750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The body instinctively responds to external stimuli by increasing energy metabolism and initiating immune responses upon receiving stress signals. Corticosterone (CORT), a glucocorticoid (GC) that regulates secretion along the hypothalamic-pituitary-adrenal (HPA) axis, mediates neurotransmission and humoral regulation. Due to the widespread expression of glucocorticoid receptors (GR), the effects of CORT are almost ubiquitous in various tissue cells. Therefore, on the one hand, CORT is a molecular signal that activates the body's immune system during stress and on the other hand, due to the chemical properties of GCs, the anti-inflammatory properties of CORT act as stabilizers to control the body's response to stress. Inflammation is a manifestation of immune activation. CORT plays dual roles in this process by both promoting inflammation and exerting anti-inflammatory effects in immune regulation. As a stress hormone, CORT levels fluctuate with the degree and duration of stress, determining its effects and the immune changes it induces. The immune system is essential for the body to resist diseases and maintain homeostasis, with immune imbalance being a key factor in the development of various diseases. Therefore, understanding the role of CORT and its mechanisms of action on immunity is crucial. This review addresses this important issue and summarizes the interactions between CORT and the immune system.
Collapse
Affiliation(s)
- Jingyu Xu
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baojuan Wang
- Department of Reproductive Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haiqing Ao
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
29
|
Gapińska N, Wlaź P, Wyska E, Świerczek A, Kamiński K, Jakubiec M, Abram M, Ciepiela K, Latacz G, Słowik T, Krokowski D, Jarosz Ł, Ciszewski A, Socała K. Effect of SSR504734, a Selective Glycine Transporter Type 1 Inhibitor, on Seizure Thresholds, Neurotransmitter Levels, and Inflammatory Markers in Mice. ACS Chem Neurosci 2025; 16:1210-1226. [PMID: 40012256 PMCID: PMC11926788 DOI: 10.1021/acschemneuro.5c00039] [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: 01/17/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
Studies have revealed that inhibition of glycine transporter type 1 (GlyT1) may provide a balanced regulation between excitation and inhibition in some brain structures and, thereby, modulate seizure activity. Data on the role of GlyT1 in epilepsy are, however, very limited. Here, we examined the effect of SSR504734, a highly selective and reversible GlyT1 inhibitor, on three acute seizure tests in mice. We also evaluated its impact on neurotransmitter levels in the relevant brain structures following seizures, possible adverse effects, and changes in the levels of inflammatory mediators in the serum and liver. In addition, in vivo pharmacokinetic profile and in vitro ADME-Tox properties of SSR504734 were investigated. The results show that SSR504734 significantly increased the threshold for tonic hindlimb extension in the MEST test after acute and repeated treatment but had no influence on seizure thresholds in the 6 Hz and i.v. PTZ seizure tests. SSR504734 did not affect the levels of glutamate, GABA, glycine, or adenosine in brain structures of mice with MES-induced seizures. However, after acute treatment, the concentration of glutamate and adenosine in the brainstem of control animals (i.e., without seizures) decreased. Moreover, SSR504734 increased the levels of inflammatory markers (TNF-α, Il-1β, IL-6, IL-10, and TLR4) in serum. In vivo pharmacokinetic profiling and in vitro ADME-Tox data confirmed suitable drug-like properties of SSR504734, including its notable penetration into brain tissue. However, possible hepatotoxicity at higher doses should be taken into account. Further studies should be considered to better characterize the SSR504734-mediated effects as well as to validate GlyT1 as a potential new molecular target in epilepsy treatment.
Collapse
Affiliation(s)
- Nikola Gapińska
- Department
of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
- Doctoral
School of Quantitative and Natural Sciences, Maria Curie-Skłodowska University, Weteranów 18, 20-038 Lublin, Poland
| | - Piotr Wlaź
- Department
of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Elżbieta Wyska
- Department
of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Artur Świerczek
- Department
of Pharmacokinetics and Physical Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Krzysztof Kamiński
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Marcin Jakubiec
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Michał Abram
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Katarzyna Ciepiela
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
- Selvita
S.A., Bobrzyńskiego
14, 30-348 Cracow, Poland
| | - Gniewomir Latacz
- Department
of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Tymoteusz Słowik
- Experimental
Medicine Center, Medical University, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Dawid Krokowski
- Department
of Molecular Biology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Łukasz Jarosz
- Department
of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary
Medicine, University of Life Sciences in
Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Artur Ciszewski
- Department
of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary
Medicine, University of Life Sciences in
Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Katarzyna Socała
- Department
of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| |
Collapse
|
30
|
Matei C, Diaconu LS, Tampa M. Interleukins in the Pathogenesis of Warts: Insight from the Last Decade-A Narrative Review. J Clin Med 2025; 14:2057. [PMID: 40142865 PMCID: PMC11942832 DOI: 10.3390/jcm14062057] [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: 02/20/2025] [Revised: 03/11/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
Human papillomavirus (HPV) is the etiological agent of a wide spectrum of diseases, from benign lesions to neoplasms. In most cases, in the first few years after infection, viral clearance occurs; however, in some cases, the infection remains persistent, allowing the progression of the lesions. The host immune response plays a key role in the resolution of the infection. The immune response to HPV is regulated by the dynamic interaction between numerous interleukins that exert pro- or anti-inflammatory effects. The role of interleukins in malignant lesions caused by HPV has been intensively studied, but in the case of benign lesions including warts, data are limited. This review compiles data from the last 10 years on the involvement of interleukins in the pathogenesis of warts, with the aim of providing new perspectives on this topic. Elucidating the role of interleukins will not only increase our knowledge of the pathogenesis of HPV infection but will also provide the foundation for the development of new therapies.
Collapse
Affiliation(s)
- Clara Matei
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laura Sorina Diaconu
- Department of Internal Medicine III and Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Internal Medicine III and Gastroenterology, Emergency University Hospital, 050098 Bucharest, Romania
| | - Mircea Tampa
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| |
Collapse
|
31
|
El-Shemi AG, Alqurashi A, Abdulrahman JA, Alzahrani HD, Almwalad KS, Felfilan HH, Alomiri WS, Aloufi JA, Madkhali GH, Maqliyah SA, Alshahrani JB, Kamal HT, Daghistani SH, Refaat B, Minshawi F. IL-10-Directed Cancer Immunotherapy: Preclinical Advances, Clinical Insights, and Future Perspectives. Cancers (Basel) 2025; 17:1012. [PMID: 40149345 PMCID: PMC11940594 DOI: 10.3390/cancers17061012] [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: 02/16/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Interleukin-10 (IL-10) is a dimeric cytokine encoded by the IL-10 gene on chromosome 1 [...].
Collapse
Affiliation(s)
- Adel G. El-Shemi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | | | - Jihan Abdullah Abdulrahman
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Hanin Dhaifallah Alzahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Khawlah Saad Almwalad
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Hadeel Hisham Felfilan
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Wahaj Saud Alomiri
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Jana Ahmed Aloufi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Ghadeer Hassn Madkhali
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
- Department of Hematology, Dr. Sulaiman Al-Habib Medical Diagnostic Laboratory, Olaya District, Riyadh 12234-3785, Saudi Arabia
| | - Sarah Adel Maqliyah
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
- Department of Blood Bank and Laboratory, Saudi German Hospital, Makkah 24242, Saudi Arabia
| | - Jood Bandar Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Huda Taj Kamal
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Sawsan Hazim Daghistani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Bassem Refaat
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| | - Faisal Minshawi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah 21955, Saudi Arabia; (A.G.E.-S.); (J.A.A.); (H.D.A.); (K.S.A.); (H.H.F.); (W.S.A.); (J.A.A.); (G.H.M.); (S.A.M.); (J.B.A.); (H.T.K.); (S.H.D.); (B.R.)
| |
Collapse
|
32
|
Araújo R, Ramalhete L, Von Rekowski CP, Fonseca TAH, Calado CRC, Bento L. Cytokine-Based Insights into Bloodstream Infections and Bacterial Gram Typing in ICU COVID-19 Patients. Metabolites 2025; 15:204. [PMID: 40137168 PMCID: PMC11944015 DOI: 10.3390/metabo15030204] [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: 02/17/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Timely and accurate identification of bloodstream infections (BSIs) in intensive care unit (ICU) patients remains a key challenge, particularly in COVID-19 settings, where immune dysregulation can obscure early clinical signs. Methods: Cytokine profiling was evaluated to discriminate between ICU patients with and without BSIs, and, among those with confirmed BSIs, to further stratify bacterial infections by Gram type. Serum samples from 45 ICU COVID-19 patients were analyzed using a 21-cytokine panel, with feature selection applied to identify candidate markers. Results: A machine learning workflow identified key features, achieving robust performance metrics with AUC values up to 0.97 for BSI classification and 0.98 for Gram typing. Conclusions: In contrast to traditional approaches that focus on individual cytokines or simple ratios, the present analysis employed programmatically generated ratios between pro-inflammatory and anti-inflammatory cytokines, refined through feature selection. Although further validation in larger and more diverse cohorts is warranted, these findings underscore the potential of advanced cytokine-based diagnostics to enhance precision medicine in infection management.
Collapse
Affiliation(s)
- Rúben Araújo
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Luís Ramalhete
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- IPST—Instituto Português do Sangue e da Transplantação, Alameda das Linhas de Torres 117, 1769-001 Lisbon, Portugal
- iNOVA4Health—Advancing Precision Medicine, RG11: Reno-Vascular Diseases Group, NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Cristiana P. Von Rekowski
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Tiago A. H. Fonseca
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Cecília R. C. Calado
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
- Institute for Bioengineering and Biosciences (iBB), The Associate Laboratory Institute for Health and Bioeconomy-i4HB, Instituto Superior Técnico (IST), Universidade de Lisboa (UL), Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Luís Bento
- NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria 130, 1169-056 Lisbon, Portugal; (R.A.)
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal
- Intensive Care Department, ULSSJ—Unidade Local de Saúde São José, Rua José António Serrano, 1150-199 Lisbon, Portugal
- Integrated Pathophysiological Mechanisms, CHRC—Comprehensive Health Research Centre, NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
| |
Collapse
|
33
|
Tukaj S, Sitna M, Sitko K. The impact of the mRNA COVID-19 vaccine on the Th-like cytokine profile in individuals with no history of COVID-19: insights into autoimmunity targeting heat shock proteins. Front Immunol 2025; 16:1549739. [PMID: 40160814 PMCID: PMC11949786 DOI: 10.3389/fimmu.2025.1549739] [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: 12/21/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Although some reports suggest that COVID-19 vaccination may exacerbate existing autoimmune diseases or trigger new-onset cases, a definitive causal relationship between the vaccines and these conditions has not been established. Several potential mechanisms have been proposed to explain this association, including: (i) molecular mimicry, which refers to a structural similarity between SARS-CoV-2 and human antigens; (ii) bystander activation, involving both B and T lymphocytes; and (iii) the effects of adjuvants. In this study, we investigated whether two doses of the mRNA COVID-19 vaccine influenced blood cytokine levels associated with major T helper cell populations, which are known to play a significant role in autoimmunity and revisited the role of the humoral autoimmune response directed against heat shock proteins (Hsps) in individuals with no history of COVID-19. While no significant differences were found in the levels of IFN-γ, IL-6, IL-22, IL-4, IL-8, IL-10, and IL-17A, between vaccinated and unvaccinated people, several positive correlations were observed between serum cytokine levels and circulating autoantibodies directed against self-Hsps exclusively in vaccinated individuals. These findings suggest that the mRNA COVID-19 vaccine does not impact cytokines involved in the pathogenesis of autoimmune diseases. Further research is required to evaluate the safety of COVID-19 vaccination in patients with autoimmune conditions, particularly those in whom anti-Hsps autoantibodies are suspected to contribute to disease development.
Collapse
Affiliation(s)
- Stefan Tukaj
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | | | | |
Collapse
|
34
|
Ubaida‐Mohien C, Moaddel R, Spendiff S, MacMillan NJ, Filion M, Morais JA, Candia J, Fitzgerald LF, Taivassalo T, Coen PM, Ferrucci L, Hepple RT. Serum Proteomic and Metabolomic Signatures of High Versus Low Physical Function in Octogenarians. Aging Cell 2025; 24:e70002. [PMID: 40059508 PMCID: PMC12073904 DOI: 10.1111/acel.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/12/2024] [Accepted: 01/10/2025] [Indexed: 05/15/2025] Open
Abstract
Physical function declines with aging, yet there is considerable heterogeneity, with some individuals declining very slowly while others experience accelerated functional decline. To gain insight into mechanisms promoting high physical function with aging, we performed proteomics, targeted metabolomics, and targeted kynurenine-focused metabolomic analyses on serum specimens from three groups of octogenarians: High-functioning master athletes (HF, n = 16), healthy normal-functioning non-athletes (NF, n = 12), and lower functioning non-athletes (LF, n = 11). Higher performance status was associated with evidence consistent with: Lower levels of circulating proinflammatory markers, as well as unperturbed tryptophan metabolism, with the normal function of the kynurenic pathway; higher circulating levels of lysophosphatidylcholines that have been previously associated with better mitochondrial oxidative capacity; lower activity of the integrated stress response; lower levels of circulating SASP protein members; and lower levels of proteins that reflect neurodegeneration/denervation. Extending the observations of previous studies focused on the biomarkers of aging that predict poor function, our findings show that many of the same biomarkers associated with poor function exhibit attenuated changes in those who maintain a high function. Because of the cross-sectional nature of this study, results should be interpreted with caution, and bidirectional causality, where physical activity behavior is both a cause and outcome of differences in the biomarker changes, remains a possible interpretation.
Collapse
Affiliation(s)
- Ceereena Ubaida‐Mohien
- Intramural Research Program, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Ruin Moaddel
- Intramural Research Program, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Sally Spendiff
- Children's Hospital of Eastern Ontario Research InstituteOttawaCanada
| | - Norah J. MacMillan
- Research Institute of the McGill University Health CentreMcGill UniversityMontrealCanada
| | - Marie‐Eve Filion
- Research Institute of the McGill University Health CentreMcGill UniversityMontrealCanada
| | - Jose A. Morais
- Research Institute of the McGill University Health CentreMcGill UniversityMontrealCanada
| | - Julián Candia
- Intramural Research Program, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Liam F. Fitzgerald
- Department of Physical TherapyUniversity of FloridaGainesvilleFloridaUSA
| | - Tanja Taivassalo
- Department of Physiology and AgingUniversity of FloridaGainesvilleFloridaUSA
| | - Paul M. Coen
- Translational Research InstituteAdvent HealthOrlandoFloridaUSA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Russell T. Hepple
- Department of Physical TherapyUniversity of FloridaGainesvilleFloridaUSA
- Department of Physiology and AgingUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
35
|
Reznik SE, Kashou A, Ward D, Yellon SM. N,N-dimethylacetamide blocks inflammation-induced preterm birth and remediates maternal systemic immune responses. Sci Rep 2025; 15:8234. [PMID: 40065144 PMCID: PMC11893883 DOI: 10.1038/s41598-025-93282-0] [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: 07/15/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
The common excipient, N,N-dimethylacetamide (DMA), prevents imminent endotoxin-induced preterm birth in mice. The present study hypothesized that DMA forestalls preterm birth to term (defined as day 18.5 or later) by attenuating bacterial endotoxin lipopolysaccharide (LPS)-induced maternal systemic inflammatory responses and cervix remodeling. Accordingly, LPS (i.p.) on day 15 postbreeding stimulated preterm delivery within 24 h while mice treated with DMA 2 h preceding and 9 h following LPS administration remained pregnant, comparable to saline and DMA controls, to deliver viable pups at term. Irrespective of LPS or DMA + LPS treatment, maternal plasma pro- and anti-inflammatory cytokines on day 15.5 (12 h post-LPS) increased tenfold compared to baseline concentrations in controls. On day 16 of pregnancy, plasma concentrations of G-CSF and TNFα were statistically significantly reduced in the prepartum LPS + DMA group compared to those in postpartum mice given LPS. By day 18 of pregnancy, all cytokines returned to baseline-equivalent to low systemic levels throughout the study in saline and DMA controls that gave birth at term. In addition, maternal plasma progesterone declined within 12 h in prepartum LPS-treated mice to postpartum concentrations on day 16. Although a similar transient decrease occurred by 12 h in DMA + LPS mice, plasma progesterone returned to baseline concentrations in controls. Contemporaneously, the progression of prepartum cervix remodeling leading to preterm delivery was acutely forestalled by DMA without impeding birth at term. These findings support the hypothesis that DMA not only prevents inflammation-driven preterm birth, but rescues pregnancy for birth to occur at term. The results raise the possibility that maternal signals can forecast risk of preterm birth while selective suppression of systemic inflammation can mitigate adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Sandra E Reznik
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA.
- Departments of Pathology and Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Alexander Kashou
- Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Daylan Ward
- Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Steven M Yellon
- Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| |
Collapse
|
36
|
Laurido-Soto OJ, Tan G, Nielsen SS, Huguenard AL, Donovan K, Xu I, Giles J, Dhar R, Adeoye O, Lee JM, Leuthardt E. Transcutaneous Auricular Vagus Nerve Stimulation Reduces Inflammatory Biomarkers and May Improve Outcomes after Large Vessel Occlusion Strokes: Results of the Randomized Clinical Trial NUVISTA. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.06.25323500. [PMID: 40093228 PMCID: PMC11908276 DOI: 10.1101/2025.03.06.25323500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Background Inflammation plays a critical role in brain injury following acute ischemic stroke (AIS). Transcutaneous auricular vagus nerve stimulation (taVNS) has shown anti-inflammatory properties in various conditions, but its efficacy in AIS remains unexplored. We investigated if taVNS could mitigate post-AIS inflammation and its safety. Methods In this randomized, sham-controlled trial with blinded outcomes assessment, patients with anterior circulation large vessel occlusion (LVO) AIS were assigned to twice-daily taVNS or sham stimulation for five days or until discharge. Key inclusion criteria included age ≥18 years, National Institutes of Health Stroke Scale (NIHSS) ≥6, anterior circulation LVO, and enrollment within 36 hours of last known normal. Primary endpoints were changes in inflammatory biomarkers (white blood cells and cytokines including interleukins (IL)-1β, 6, 10, 17α, and tumor necrosis factor alpha (TNFα) measured at baseline and Days 1, 3, 5, and 7, and taVNS safety. Secondary exploratory endpoints included change in NIHSS, 90 day modified Rankin score (mRS), and safety (bradycardia, hypotension, infection, and death). Results Thirty-five patients (17 taVNS, 18 sham) were enrolled. The taVNS group showed a significant rate of change in normalized aggregate pro-inflammatory cytokines and interleukin-6 levels compared to sham (p=0.04 and p<0.001, respectively). Each 1 pg/mL reduction in interleukin-6 correlated with a 0.798-point improvement in NIHSS in the taVNS group (95% confidence interval [0.077, 1.518], p = 0.031]), with no significant correlation in the sham group. IL-1β, 10, 17α, and TNFα showed reduction in cytokine levels, but did not reach statistical significance. There were no statistically significant differences amongst mRS and safety outcomes between both groups. Conclusions taVNS safely reduced post-AIS inflammation in anterior circulation LVO stroke patients, demonstrating biological effects. Secondary analyses also found potential effects in NIHSS improvements. These promising findings warrant further investigation in larger trials.
Collapse
Affiliation(s)
- Osvaldo J Laurido-Soto
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Gansheng Tan
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Division of Neurotechnology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Susan Searles Nielsen
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Anna L Huguenard
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Division of Neurotechnology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Kara Donovan
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Division of Neurotechnology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Isabella Xu
- Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - James Giles
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rajat Dhar
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Opeolu Adeoye
- Department of Emergency Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Eric Leuthardt
- Department of Neurosurgery, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Division of Neurotechnology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
37
|
Baek H, Yang SW, Kim S, Lee Y, Park H, Park M, Jeon BJ, Park H, Hwang HS, Kim JY, Kim JH, Kang YS. Development of Anti-Inflammatory Agents Utilizing DC-SIGN Mediated IL-10 Secretion in Autoimmune and Immune-Mediated Disorders: Bridging Veterinary and Human Health. Int J Mol Sci 2025; 26:2329. [PMID: 40076949 PMCID: PMC11901132 DOI: 10.3390/ijms26052329] [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: 01/17/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin) is a C-type lectin receptor expressed on dendritic cells and M2 macrophages, playing a key role in immune regulation and pathogen recognition. Its ability to mediate anti-inflammatory effects by interacting with specific ligands triggers pathways that suppress pro-inflammatory responses and promote tissue repair, making it a potential therapeutic target for inflammatory and autoimmune diseases. DC-SIGN homologs in various animal species share structural similarities and perform comparable immune functions, offering valuable insights into its broader application across species. By recognizing carbohydrate ligands on pathogens, DC-SIGN facilitates immune modulation, which can be harnessed for developing therapies aimed at controlling inflammation. In veterinary medicine, autoimmune and inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel disease, represent significant challenges, and the anti-inflammatory properties of DC-SIGN could provide new therapeutic options to improve disease management and enhance animal health. Future investigations should focus on the structural and functional analysis of DC-SIGN homologs in various species, as well as the development of preclinical models to translate these findings into clinical interventions bridging veterinary and human health.
Collapse
Affiliation(s)
- Hayeon Baek
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
| | - Seung-Woo Yang
- Sanford Consortium for Regenerative Medicine, School of Medicine, University of California, San Diego, CA 92037, USA;
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Seulki Kim
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| | - Yunseok Lee
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| | - Hwi Park
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
| | - Byung-Ju Jeon
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Hanwool Park
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Joon-Young Kim
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| |
Collapse
|
38
|
Bermudez‐Lekerika P, Tseranidou S, Kanelis E, Nüesch A, Crump KB, Alexopoulos LG, Wuertz‐Kozak K, Noailly J, Le Maitre CL, Gantenbein B. Ex Vivo and In Vitro Proteomic Approach to Elucidate the Relevance of IL-4 and IL-10 in Intervertebral Disc Pathophysiology. JOR Spine 2025; 8:e70048. [PMID: 39931581 PMCID: PMC11808320 DOI: 10.1002/jsp2.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/08/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
Background This study investigates the native presence and potential anabolic effects of interleukin (IL)-4 and IL-10 in the human intervertebral disc (IVD). Methods Human nucleus pulposus (NP) cells cultured in 3D from trauma and degenerate IVDs and NP explants were stimulated with 10 ng/mL IL-4, IL-10, or each in combination with 1 ng/mL IL-1β stimulation. The role of IL-4 and IL-10 in the IVD was evaluated using immunohistochemistry, gene expression, and Luminex multiplex immunoassay proteomics (73 secreted) and phosphoproteomics (21 phosphorylated proteins). Results IL-4, IL-4R, and IL-10R expression and localization in human cartilage endplate tissue were demonstrated for the first time. No significant gene expression changes were noted under IL-4 or IL-10 stimulation. However, IL-1β stimulation significantly increased MMP3, COX2, TIMP1, and TRPV4 expression in NP cells from trauma IVDs. Combined IL-4 and IL-1β treatment induced a significant increase in protein secretion of IL-1α, IL-7, IL-16, IL-17F, IL-18, IFNγ, TNF, ST2, PROK1, bFGF2, and stem cell factor exclusively in NP cells from degenerated IVDs. Conversely, the secretome profile of explants revealed an IL-4-mediated decrease in CXCL13 following treatment with IL-1β. Combined IL-10 and IL-1β treatment increased neurotrophic growth factor secretion compared with IL-10 baseline. Conclusions The NP cell phenotype affects the pleiotropic role of IL-4, which can induce a pro-inflammatory response in the presence of catabolic stimuli and enhance the effects of IL-1β in degenerated IVDs. Environmental factors, including 3D culture and hypoxia, may alter IL-4's role. Finally, IL-10's potential neurotrophic effects under catabolic stimuli warrant further investigation to clarify its role in IVD degeneration.
Collapse
Affiliation(s)
- Paola Bermudez‐Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Faculty of MedicineUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical Sciences (GCB)University of BernBernSwitzerland
| | | | - Exarchos Kanelis
- Testing ServicesProtavio Ltd, Demokritos Science ParkAthensGreece
- School of Mechanical EngineeringNational Technical University of AthensZografouGreece
| | - Andrea Nüesch
- Division of Clinical Sciences, School of Medicine and Population HealthUniversity of SheffieldSheffieldEngland
| | - Katherine B. Crump
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Faculty of MedicineUniversity of BernBernSwitzerland
- Graduate School for Cellular and Biomedical Sciences (GCB)University of BernBernSwitzerland
| | - Leonidas G. Alexopoulos
- Testing ServicesProtavio Ltd, Demokritos Science ParkAthensGreece
- School of Mechanical EngineeringNational Technical University of AthensZografouGreece
| | - Karin Wuertz‐Kozak
- Department of Biomedical EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Spine Center, Schön Klinik München Harlaching Academic Teaching Hospital and Spine ResearchInstitute of the Paracelsus Private Medical University Salzburg (Austria)MunichGermany
| | - Jérôme Noailly
- Department of EngineeringUniversitat Pompeu FabraBarcelonaSpain
| | - Christine L. Le Maitre
- Division of Clinical Sciences, School of Medicine and Population HealthUniversity of SheffieldSheffieldEngland
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone and Joint Program, Department for BioMedical Research (DBMR), Faculty of MedicineUniversity of BernBernSwitzerland
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Faculty of MedicineUniversity of BernBernSwitzerland
| |
Collapse
|
39
|
Anh DD, Vugrek L, The NT, Hafza N, My TN, Linh LTK, Loc DH, Schmidt-Chanasit J, Toan NL, Kremsner PG, Song LH, Velavan TP. Characterization of dengue patients in Vietnam: Clinical, virological, and IL-10 profiles during 2021- 2022 outbreaks. PLoS Negl Trop Dis 2025; 19:e0012954. [PMID: 40153381 PMCID: PMC11977983 DOI: 10.1371/journal.pntd.0012954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 04/08/2025] [Accepted: 02/28/2025] [Indexed: 03/30/2025] Open
Abstract
BACKGROUND The pathogenesis of dengue is attributed to a complex interaction between the dengue virus (DENV) and the host immune system. The aim of this study is to investigate the clinical, virological, and Interleukin-10 (IL-10) profiles of dengue patients in Vietnam from two consecutive outbreaks in 2021 and 2022. METHODS A total of n=306 dengue patients were examined, who were clinically stratified according to dengue without warning signs (DF; n=178), dengue with warning signs (DWS; n=115) and severe dengue (SD; n=13). Patients were screened for dengue, Zika and chikungunya viruses. DENV were subjected to serotype specific real-time RT-PCR. Interleukin-10 (IL-10) levels were measured by ELISA, and IL-10 promoter variants (-1082G/A; -819C/T; -592C/A) were genotyped by direct Sanger sequencing to determine a possible association with susceptibility to dengue and disease severity. RESULTS No chikungunya or Zika viruses were detected. Patients were infected by one of the three different DENV serotypes (DENV-1, -2, -4). Plasma IL-10 levels were significantly elevated in patients (DF vs. DWS, p=0.004; DF vs. SD, p=0.001; DWS vs. SD, p=0.015). While the IL-10 allele -819C contributed to an increased risk of dengue (OR = 1.5, 95% CI = 1.1-2.0, p=0.04), genotype -1082GA showed a protective role against the disease (OR = 0.45, 95% CI = 0.27-0.72, p=0.009), and allele -1082G showed a protective role against DWS (OR = 0.44, 95% CI = 0.22-0.81, p=0.049). Also, the IL-10 GTA (-1082G/-819T/-592A) haplotype was observed to confer protection (OR = 0.31, 95% CI = 0.14-0.67, p< 0.003). CONCLUSION While DENV-1 and DENV-2 were the predominant serotypes in circulation, plasma IL-10 levels and IL-10 promoter variants were also significantly associated with dengue and its severity.
Collapse
Affiliation(s)
- Do Duc Anh
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Lara Vugrek
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Nguyen Trong The
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- 108 Military Central Hospital, Hanoi, Vietnam
| | - Nourhane Hafza
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Truong Nhat My
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Le Thi Kieu Linh
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Do Huy Loc
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
| | - Jonas Schmidt-Chanasit
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Faculty of Mathematics, Informatics and Natural Sciences, University of Hamburg, Hamburg, Germany
| | - Nguyen Linh Toan
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- Vietnam Military Medical University, Hanoi, Vietnam
| | - Peter G. Kremsner
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné (CERMEL), Gabon
| | - Le Huu Song
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- 108 Military Central Hospital, Hanoi, Vietnam
| | - Thirumalaisamy P. Velavan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Vietnamese-German Center for Medical Research (VG-CARE), Hanoi, Vietnam
- Faculty of Medicine, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
40
|
Moradi A, Aslani F, Boskabady MH, Pahlavan Y, Aslani MR. Effect of Zataria multiflora supplementation on pro- or anti-inflammatory markers: a systematic review and meta-analysis of randomized placebo-controlled trials. Inflammopharmacology 2025; 33:1255-1270. [PMID: 39992592 DOI: 10.1007/s10787-025-01668-3] [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] [Accepted: 01/22/2025] [Indexed: 02/26/2025]
Abstract
The aim of this study was to investigate the effectiveness of Zataria multiflora and carvacrol supplementation on inflammatory markers (IL-2, IL-4, IL-5, IL-6, TNF-α, IL-8, IL-10, interferon gamma (IFN-γ), C-reactive protein (CRP), monocyte chemotactic protein 1 (MCP-1), epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF)) by conducting a systematic review and meta-analysis of randomized controlled trials (RCTs).The exploration of literature was conducted until August 2024 on databases like PubMed/Medline, Web of Science, Scopus, Cochrane CENTRAL, and Google Scholar. The current study incorporated trial studies examining how oral supplements of Zataria multiflora and carvacrol impact concentrations of inflammatory markers. By utilizing a random effects model, the mean differences (SMD) and 95% confidence intervals (CI) were pooled for analysis of the results. The Cochrane Q and I2 values were used to evaluate heterogeneity. The meta-analysis included ten cases, with 562 participants in the Zataria multiflora group and 700 in the control group. A significant decrease in the levels of various cytokines including IL-2, IL-4, IL-5, IL-6, IL-8, CRP, EGF, VEGF, and MCP-1 was observed with the consumption of Zataria multiflora along with a noteworthy increase in both IFN-γ and IL-10. However, TNF-α levels remained unaffected by the intervention involving Zataria multiflora and carvacrol. It should be noted that limitations of this study include the fact that it draws from research in Iran, encompasses a range of different diseases, and overlooks potential confounders like smoking, physical activity, and diet. In summary, the results suggested that Zataria multiflora and carvacrol can be beneficial for reducing inflammation.
Collapse
Affiliation(s)
- Alireza Moradi
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, 5615780011, Iran
| | - Farzin Aslani
- Department of Orthopedics, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Yasamin Pahlavan
- Biosensor Sciences and Technologies Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, 5615780011, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Kuan CH, Chang L, Ho CY, Tsai CH, Liu YC, Huang WY, Wang YN, Wang WH, Wang TW. Immunomodulatory hydrogel orchestrates pro-regenerative response of macrophages and angiogenesis for chronic wound healing. Biomaterials 2025; 314:122848. [PMID: 39342917 DOI: 10.1016/j.biomaterials.2024.122848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Chronic wound healing often encounters challenges characterized by prolonged inflammation and impaired angiogenesis. While the immune response plays a pivotal role in orchestrating the intricate process of wound healing, excessive inflammation can hinder tissue repair. In this study, a bilayer alginate hydrogel system encapsulating polyelectrolyte complex nanoparticles (PCNs) loaded with anti-inflammatory cytokines and angiogenic growth factors is developed to address the challenges of chronic wound healing. The alginate hydrogel is designed using two distinct crosslinking methods to achieve differential degradation, thereby enabling precise spatial and temporal controlled release of PCNs. Initially, interleukin-10 (IL-10) is released to mitigate inflammation, while unsaturated PCNs bind and remove accumulated pro-inflammatory cytokines at the wound site. Subsequently, angiogenic growth factors, including vascular endothelial growth factor and platelet-derived growth factor, are released to promote vascularization and vessel maturation. Our results demonstrate that the bilayer hydrogel exhibits distinct degradation kinetics between the two layers, facilitating the staged release of multiple signaling molecules. In vitro experiments reveal that IL-10 can activate the Jak1/STAT3 pathway, thereby suppressing pro-inflammatory cytokines and chemokines while down-regulating inflammation-related genes. In vivo studies demonstrate that application of the hydrogel in chronic wounds using diabetic murine model promotes healing by positively modulating multiple integral reparative mechanisms. These include reducing inflammation, promoting macrophage polarization towards a pro-regenerative phenotype, enhancing keratinocyte migration, stimulating angiogenesis, and expediting wound closure. In conclusion, our hydrogel system effectively mitigates inflammatory responses and provides essential physiological cues by inducing a synergistic angiogenic effect, thus offering a promising approach for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taiwan.
| | - Ling Chang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Yu Ho
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan; Department of Bioengineering, Rice University, Houston, USA
| | - Chia-Hsuan Tsai
- Division of Plastic Surgery, Department of Surgery, Chang Gung Memorial Hospital, Keelung Branch, Taiwan
| | - Yu-Chung Liu
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan; Department of Biomedical Engineering, University of Michigan-Ann Arbor, Michigan, USA
| | - Wei-Yuan Huang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ning Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Tzu-Wei Wang
- Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
42
|
Lizhen M, Shuhua C, Yong Z, Weijia Z, Xuelong Z, Mingzhao Z, Zhifeng Y, Xiangjun H, Lifeng W. Microwaves Activate Immune Response and Promote Lymphocytes Proliferation of Wistar Rats. APMIS 2025; 133:e70017. [PMID: 40083229 DOI: 10.1111/apm.70017] [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: 10/23/2024] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
The potential effects of microwave radiation have gained increasing attention due to its widespread application in daily life. This study aimed to investigate its effects on rat immune function. Male Wistar rats were subjected to 2.856 GHz microwave radiation for 20 min. Cytokine levels in serum were measured by ELISA at 0 and 7 days' post-exposure. HE staining was used to observe the structure of the spleen. Morphology and proliferation of spleen mixed lymphocytes were detected after 24 h of culture, and proliferation was evaluated by the CCK-8 assay. Expression of HSP70 and calreticulin (CRT) was assessed by WB. Rats showed an increased secretion of inflammatory factors in serum. Spleen tissue revealed no significant damage. Compared to the control group, irradiated groups (R-0d and R-7d) showed a significant increase and thickening of the white pulp. The boundary between the red and white medulla in the R-7d group appeared blurred. CCK-8 assay showed that splenic mixed lymphocytes in the irradiated group showed a significant proliferation. There was no significant difference in HSP70 and CRT expression between the exposed and control groups. Microwave exposure might activate the immune system of rats, potentially triggering a T cell-mediated inflammatory response.
Collapse
Affiliation(s)
- Ma Lizhen
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Cao Shuhua
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zou Yong
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhi Weijia
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhao Xuelong
- Beijing Institute of Radiation Medicine, Beijing, China
| | | | - Yan Zhifeng
- First Medical Center of Chinses PLA General Hospital, Beijing, China
| | - Hu Xiangjun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Wang Lifeng
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
43
|
Patat AS, Nalbantoğlu ÖU. Enhancing Functional Protein Design Using Heuristic Optimization and Deep Learning for Anti-Inflammatory and Gene Therapy Applications. Proteins 2025. [PMID: 39985803 DOI: 10.1002/prot.26810] [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: 08/29/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/24/2025]
Abstract
Protein sequence design is a highly challenging task, aimed at discovering new proteins that are more functional and producible under laboratory conditions than their natural counterparts. Deep learning-based approaches developed to address this problem have achieved significant success. However, these approaches often do not adequately emphasize the functional properties of proteins. In this study, we developed a heuristic optimization method to enhance key functionalities such as solubility, flexibility, and stability, while preserving the structural integrity of proteins. This method aims to reduce laboratory demands by enabling a design that is both functional and structurally sound. This approach is particularly valuable for the synthetic production of proteins with anti-inflammatory properties and those used in gene therapy. The designed proteins were initially evaluated for their ability to preserve natural structures using recovery and confidence metrics, followed by assessments with the AlphaFold tool. Additionally, natural protein sequences were mutated using a genetic algorithm and compared with those designed by our method. The results demonstrate that the protein sequences generated by our method exhibit much greater similarity to native protein sequences and structures. The code and sequences for the designed proteins are available at https://github.com/aysenursoyturk/HMHO.
Collapse
Affiliation(s)
- Ayşenur Soytürk Patat
- Department of Bioinformatics Systems Biology, Erciyes University, Kayseri, Turkey
- Department of Bioinformatics, Necmettin Erbakan University, Konya, Turkey
| | | |
Collapse
|
44
|
Brimacombe M, Jadhav A, Lawrence DA, Carson K, Lee WT, Hogan AH, Herbst KW, Lynes MA, Salazar JC. The Detection of COVID-19-Related Multivariate Biomarker Immune Response in Pediatric Patients: Statistical Aspects. Viruses 2025; 17:297. [PMID: 40143228 PMCID: PMC11945793 DOI: 10.3390/v17030297] [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: 01/27/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
The development of new point-of-care diagnostic testing tools for the detection of infectious diseases such as COVID-19 are a key aspect of clinical care and research. Accurate predictive classification methods are required to correctly identify and treat patients. Here, the onset of multisystem inflammatory syndrome in children (MIS-C), a more serious form of COVID-19, was predicted in a pediatric population using a set of multivariate immunological biomarker expression values. A first-stage bivariate detection of statistically significant biomarkers was obtained from a chosen set of standard cytokines and chemokine biomarkers considered relevant to COVID-19-related infection and disease. To incorporate the observed correlation structure among the resulting set of significant biomarkers, dimension reduction was then applied in the form of principal components. A second-stage logistic regression model using a small number of the principal component variables provided a highly predictive classification model for MIS-C. The resulting model was shown to compare favorably with an artificial neural network-based predictive model.
Collapse
Affiliation(s)
- Michael Brimacombe
- Connecticut Children’s Medical Center, Hartford, CT 06106, USA
- Department of Pediatrics, UConn Health, Farmington, CT 06030, USA
| | - Aishwarya Jadhav
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - David A. Lawrence
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
- Department of Biomedical Sciences, College of Integrated Health Sciences, University at Albany, Rensselaer, NY 12144, USA
| | - Kyle Carson
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
- Department of Biomedical Sciences, College of Integrated Health Sciences, University at Albany, Rensselaer, NY 12144, USA
| | - William T. Lee
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
- Department of Biomedical Sciences, College of Integrated Health Sciences, University at Albany, Rensselaer, NY 12144, USA
| | - Alexander H. Hogan
- Connecticut Children’s Medical Center, Hartford, CT 06106, USA
- Department of Pediatrics, UConn Health, Farmington, CT 06030, USA
| | | | - Michael A. Lynes
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Juan C. Salazar
- Connecticut Children’s Medical Center, Hartford, CT 06106, USA
- Department of Pediatrics, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
45
|
Floyd BM, Schmidt EL, Till NA, Yang JL, Liao P, George BM, Flynn RA, Bertozzi CR. Mapping the nanoscale organization of the human cell surface proteome reveals new functional associations and surface antigen clusters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637979. [PMID: 40027624 PMCID: PMC11870420 DOI: 10.1101/2025.02.12.637979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The cell surface is a dynamic interface that controls cell-cell communication and signal transduction relevant to organ development, homeostasis and repair, immune reactivity, and pathologies driven by aberrant cell surface phenotypes. The spatial organization of cell surface proteins is central to these processes. High-resolution fluorescence microscopy and proximity labeling have advanced studies of surface protein associations, but the spatial organization of the complete surface proteome remains uncharted. In this study, we systematically mapped the surface proteome of human T-lymphocytes and B-lymphoblasts using proximity labeling of 85 antigens, identified from over 100 antibodies tested for binding to surface-exposed proteins. These experiments were coupled with an optimized data-independent acquisition mass spectrometry workflow to generate a robust dataset. Unsupervised clustering of the resulting interactome revealed functional modules, including well-characterized complexes such as the T-cell receptor and HLA class I/II, alongside novel clusters. Notably, we identified mitochondrial proteins localized to the surface, including the transcription factor TFAM, suggesting previously unappreciated roles for mitochondrial proteins at the plasma membrane. A high-accuracy machine learning classifier predicted over 6,000 surface protein associations, highlighting functional associations such as IL10RB's role as a negative regulator of type I interferon signaling. Spatial modeling of the surface proteome provided insights into protein dispersion patterns, distinguishing widely distributed proteins, such as CD45, from localized antigens, such as CD226 pointing to active mechanisms of regulating surface organization. This work provides a comprehensive map of the human surfaceome and a resource for exploring the spatial and functional dynamics of the cell membrane proteome.
Collapse
Affiliation(s)
- Brendan M Floyd
- Sarafan ChEM-H and Department of Chemistry, Stanford University, Stanford, CA, USA
- Lead contact
| | - Elizabeth L Schmidt
- Sarafan ChEM-H and Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Nicholas A Till
- Sarafan ChEM-H and Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Jonathan L Yang
- Sarafan ChEM-H and Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Pinyu Liao
- Sarafan ChEM-H and Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Benson M George
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Ryan A Flynn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Carolyn R Bertozzi
- Sarafan ChEM-H and Department of Chemistry, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
- Lead contact
| |
Collapse
|
46
|
Mukherjee D, Satyavolu S, Thomas A, Cioffi S, Li Y, Chan ER, Wen K, Huang AY, Jain MK, Dubyak GR, Nayak L. Neutrophil KLF2 regulates inflammasome-dependent neonatal mortality from endotoxemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637657. [PMID: 39990480 PMCID: PMC11844471 DOI: 10.1101/2025.02.11.637657] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Preterm neonates die at a significantly higher rate from sepsis than full-term neonates, attributable to their dysregulated immune response. In addition to tissue destruction caused directly by bacterial invasion, an overwhelming cytokine response by the immune cells to bacterial antigens also results in collateral damage. Sepsis leads to decreased gene expression of a critical transcription factor, Krüppel-like factor-2 (KLF2), a tonic repressor of myeloid cell activation. Using a murine model of myeloid- Klf2 deletion, we show that loss of KLF2 is associated with decreased survival after endotoxemia in a developmentally dependent manner, with increased mortality at postnatal day 4 (P4) compared to P12 pups. This survival is significantly increased by neutrophil depletion. P4 knockout pups have increased pro-inflammatory cytokine levels after endotoxemia compared to P4 controls or P12 pups, with significantly increased levels of IL-1β, a product of the activation of the NLRP3 inflammasome complex. Loss of myeloid-KLF2 at an earlier postnatal age leads to a greater increase in NLRP3 priming and activation and greater IL-1β release by BMNs. Inhibition of NLRP3 inflammasome activation by MCC950 significantly increased survival after endotoxemia in P4 pups. Transcriptomic analysis of bone marrow neutrophils showed that loss of myeloid-KLF2 is associated with gene enrichment of pro-inflammatory pathways in a developmentally dependent manner. These data suggest that targeting KLF2 could be a novel strategy to decrease the pro-inflammatory cytokine storm in neonatal sepsis and improve survival in neonates with sepsis. Summary sentence KLF2 regulates the developmental response to endotoxin in neonatal mice through the NLRP3 inflammasome signaling pathway.
Collapse
|
47
|
Wang D, Zou Y, Zhao T, Cao W, Han J, Wu Q, Li Z, Li X, Liu P, Bai L, Ren G. Fibroblast growth factor 21 alleviated atopic march by inhibiting the differentiation of type 2 helper T cells. Int Immunopharmacol 2025; 147:114055. [PMID: 39798471 DOI: 10.1016/j.intimp.2025.114055] [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: 09/27/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND The blood FGF21 expression has been previously suggested to increase in patients developing atopic dermatitis (AD) and asthma. However, its impact on atopic march is rarely analyzed. The present work focused on investigating the role of Fibroblast Growth Factor 21(FGF21) in atopic march mice and its underlying mechanisms. METHODS The models were induced with Diisononyl phthalate (DINP) and OVA in wild-type C57BL/6 and FGF21-/- mice. RAW264.7 cells were induced by LPS with/without FGF21 or KLB-SiRNA for in vitro analyses. RESULTS The data indicated that there were more severe allergic reactions including IgE levels and the proportion of mast cells in the blood of FGF21-/- mice in relative to WT model mice during the progression from AD to asthma. However, exogenous administration of FGF21 inhibited allergies. In this study, we reported that FGF21 mitigated AD-like lesions and Th1/2 or Th17/Treg cell imbalance in AD mice, and significantly decreased TSLP, IL-33, IL-4, IL-5, IL-13 and IL-17A expression on skin. During the asthma phase, FGF21 improved airway remodeling by downgrading inflammatory factors IL-4, IL-5, IL-13 and IL-17A; fibrotic markers α-SMA and Collagen I; and oxidative products MDA and ROS in wild-type model mice. Compared with WT model mice, the adverse consequences were aggravated in FGF21-/- asthmatic mice. From the mechanistic perspective, FGF21 suppressed NF-κB/NLRP3, TGF-β1/Smad3 and JNK signaling pathways and increased Nrf2 expression in vivo and in vitro. In addition, β-Klotho knockdown attenuated the ameliorative impact of FGF21 on cellular damage. Blocking AMPKα in the LPS-treated RAW264.7 cells inhibited the reduction of FGF21 and the phosphorylation of JNK. CONCLUSIONS To conclude, FGF21 alleviated atopic march by inhibiting Th2/17 immune response, and reduced airway remodeling by regulating NF-κB/NLRP3, TGF-β1/Smad3 and AMPKα/JNK pathways. Moreover, this study provides a rationale and novel ideas for applying FGF21 in treating AD and asthma.
Collapse
Affiliation(s)
- Dan Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yimeng Zou
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Tianqi Zhao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Weiyue Cao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jiachi Han
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Qing Wu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhitong Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyu Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Peijing Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Lin Bai
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Research Center of Genetic Engineering of Pharmaceuticals of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
48
|
Peng H, Song X, Chen J, Xiong X, Yang L, Yu C, Qiu M, Zhang Z, Hu C, Zhu S, Xia B, Wang J, Xiong Z, Du L, Yang C. Soybean bioactive peptide supplementation improves gut health and metabolism in broiler chickens. Poult Sci 2025; 104:104727. [PMID: 39729732 PMCID: PMC11741984 DOI: 10.1016/j.psj.2024.104727] [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: 09/18/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/29/2024] Open
Abstract
This study aimed to investigate the effects of soybean bioactive peptide (SBP) on the growth performance and intestinal health of yellow-feathered broilers and to further elucidate the regulatory mechanisms of intestinal health using multi-omics analysis. A total of 320 1-day-old yellow-feathered broilers were randomly divided into two groups, with 10 replicates per group and 16 birds per replicate. Broilers in the control group received the basal diet, and those in the experimental group (SBPG) received the basal diet with 0.2 % SBP replacing the same amount of soybean meal. The experiment lasted for 70 d. The results showed that, compared with those in the control group, the final body weight and average daily gain of SBPG broilers were significantly higher (P < 0.05), and the feed conversion ratio was significantly lower (P < 0.05). Notably, SBP significantly improved gut health in chickens, including increased intestinal villus height, decreased levels of proinflammatory factors, such as IL-1β and interferon-γ, and upregulated expression of tight junction proteins, such as ZO-1 and occludin. In addition, transcriptome sequencing results revealed that broilers in the SBP group exhibited significant enrichment in multiple metabolic pathways, including fatty acid metabolism, fatty acid degradation, and the biosynthesis of unsaturated fatty acids (P < 0.05). Cecal 16S rRNA sequencing showed that SBPG increased the abundance of the butyrate-producing beneficial bacteria Muribaculaceae. Subsequent cecal metabolome analysis also revealed that SBPG enhanced lipid-related metabolic pathways, such as alpha-linolenic acid metabolism and GPI-anchor biosynthesis. In conclusion, SBP is a potential feed additive that can improve intestinal morphology, enhance intestinal immunity and barrier function, optimize the structure of the intestinal microbiota, and enhance metabolic function.
Collapse
Affiliation(s)
- Han Peng
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Xiaoyan Song
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Jialei Chen
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Xia Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Li Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Mohan Qiu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Zengrong Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Chenming Hu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Shiliang Zhu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Bo Xia
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Jiangxian Wang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Zhuxiang Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Longhuan Du
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, PR China.
| |
Collapse
|
49
|
Yeung JC, Koike T, Wagnetz D, Machuca TN, Bonato R, Liu M, Juvet S, Cypel M, Keshavjee S. Ex vivo delivery of recombinant IL-10 to human donor lungs. JHLT OPEN 2025; 7:100192. [PMID: 40144859 PMCID: PMC11935390 DOI: 10.1016/j.jhlto.2024.100192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background The immunoregulatory cytokine interleukin-10 (IL-10) has been shown to be a promising therapy for donor lung injuries before transplantation. However, the very short half-life of IL-10 in vivo (∼2 hours) has necessitated the use of gene therapy in almost all animal models of lung transplantation. Because isolation of the donor lung on the ex vivo lung perfusion (EVLP) circuit removes it from the influence of renal and hepatic clearance mechanisms, a much-prolonged half-life of IL-10 is anticipated. Thus, we hypothesized that delivery of recombinant IL-10 (rIL-10) to injured donor lungs isolated on EVLP could be a clinically relevant and a logistically simpler method of employing IL-10 therapy in lung transplantation. Methods Injured human donor lungs clinically rejected for transplantation were split into single lungs and the better of the 2 subjected to 12 hours of EVLP and randomized (n = 5/group) to receive either saline (control), rIL-10 (5 µg in 2-liter perfusate), or rIL-10 (25 µg) aerosolized into the airways. Results Perfusate and intratracheal delivery of rIL-10 did not provide the therapeutic anti-inflammatory action that has been traditionally achieved with gene therapy. It appears that intratracheally delivered rIL-10 moves into the perfusate where it seems to be biologically inactive. Conclusions Gene therapy remains superior as it allows for continued production of IL-10 within the alveoli where it has the potential to continuously act on alveolar macrophages and epithelial cells in a paracrine fashion.
Collapse
Affiliation(s)
- Jonathan C. Yeung
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Terumoto Koike
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Dirk Wagnetz
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Tiago N. Machuca
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Riccardo Bonato
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Schwab AD, Wyatt TA, Schanze OW, Nelson AJ, Gleason AM, Duryee MJ, Mosley DD, Thiele GM, Mikuls TR, Poole JA. Lung-delivered IL-10 mitigates Lung inflammation induced by repeated endotoxin exposures in male mice. Physiol Rep 2025; 13:e70253. [PMID: 39980189 PMCID: PMC11842461 DOI: 10.14814/phy2.70253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Therapies capable of resolving inflammatory lung disease resulting from high-consequence occupational/environmental hazards are lacking. This study seeks to determine the therapeutic potential of direct lung-delivered interleukin (IL)-10 following repeated lipopolysaccharide exposures. C57BL/6 mice were intratracheally instilled with LPS (10 μg) and treated with IL-10 (1 μg) or vehicle control for 3 days. Lung cell infiltrates were enumerated by flow cytometry. Lung sections were stained for myeloperoxidase (MPO), CCR2, vimentin, and post-translational protein citrullination (CIT) and malondialdehyde-acetaldehyde (MAA) modifications. Lung function testing and longitudinal in vivo micro-CT imaging were performed. Whole lungs were profiled using bulk RNA sequencing. IL-10 treatment reduced LPS-induced weight loss, pentraxin-2, and IL-6 serum levels. LPS-induced lung proinflammatory and wound repair mediators (i.e., TNF-α, IL-6, CXCL1, CCL2, MMP-8, MMP-9, TIMP-1, fibronectin) were decreased with IL-10. IL-10 reduced LPS-induced influx of lung neutrophils, CD8+ T cells, NK cells, recruited monocyte-macrophages, monocytes, and tissue expression of CCR2+ monocytes-macrophages, MPO+ neutrophils, vimentin, CIT, and MAA. IL-10 reduced LPS-induced airway hyperresponsiveness and improved lung compliance. Micro-CT imaging confirmed the reduction in LPS-induced lung density by IL-10. Lung-delivered IL-10 therapy administered after daily repeated endotoxin exposures strikingly reduces lung inflammatory and wound repair processes to decrease lung pathologic changes and mitigate airway dysfunction.
Collapse
Affiliation(s)
- Aaron D. Schwab
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Todd A. Wyatt
- Division of Pulmonary, Critical Care & SleepUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Department of Environmental, Agricultural and Occupational Health, College of Public HealthUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Oliver W. Schanze
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Amy J. Nelson
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Angela M. Gleason
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Michael J. Duryee
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Division of Rheumatology & Immunology, Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Deanna D. Mosley
- Division of Pulmonary, Critical Care & SleepUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
| | - Geoffrey M. Thiele
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Division of Rheumatology & Immunology, Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Ted R. Mikuls
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemResearch ServiceOmahaNebraskaUSA
- Division of Rheumatology & Immunology, Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jill A. Poole
- Division of Allergy & ImmunologyUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|