1
|
Vella VR, Ainsworth-Cruickshank G, Luft C, Wong KE, Parfrey LW, Vogl AW, Holman PJ, Bodnar TS, Raineki C. Dysregulation of immune system markers, gut microbiota and short-chain fatty acid production following prenatal alcohol exposure: A developmental perspective. Neurochem Int 2025; 185:105952. [PMID: 39988283 DOI: 10.1016/j.neuint.2025.105952] [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: 11/04/2024] [Revised: 01/25/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Prenatal alcohol exposure (PAE) can severely impact fetal development, including alterations to the developing immune system. Immune perturbations, in tandem with gut dysbiosis, have been linked to brain and behavioral dysfunction, but this relationship is poorly understood in the context of PAE. This study takes an ontogenetic approach to evaluate PAE-induced alterations to brain and serum cytokine levels and both the composition and metabolic output of the gut microbiota. Using a well-established rat model of PAE, cytokine levels in the serum, prefrontal cortex, amygdala, and hypothalamus as well as gut microbiota composition and short-chain fatty acid (SCFA) levels were assessed at three postnatal (P) timepoints: P8 (infancy), P22 (weaning), and P38 (adolescence). Male PAE rats had increased cytokine levels in the amygdala and hypothalamus, but not prefrontal cortex, at P8. This altered neuroimmune function was not seen in the PAE females. The effect of PAE on central cytokine levels was reduced at P22/38, the same age at which PAE-induced alterations in serum cytokine levels emerge in both sexes. PAE reduced bacterial diversity in both sexes at P8, but only in females at P38, where a PAE-induced unique community composition emerged. Both sexes had alterations to specific bacterial taxa (e.g., Firmicutes), some of which are important in producing the SCFA butyric acid, which was decreased in PAE animals at P22. These results demonstrate that PAE leads to sex- and age-specific alterations in immune function, gut microbiota and SCFA production, highlighting the need to consider both age and sex in future work.
Collapse
Affiliation(s)
- Victoria R Vella
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | | | - Carolina Luft
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Kingston E Wong
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Laura W Parfrey
- Department of Botany, University of British Columbia, British Columbia, Canada
| | - A Wayne Vogl
- Life Sciences Centre, Department of Cellular and Physiological Sciences, University of British Columbia, British Columbia, Canada
| | - Parker J Holman
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Tamara S Bodnar
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada.
| |
Collapse
|
2
|
Le PM, Pal-Ghosh S, Stepp MA, Menko AS. Shared Phenotypes of Immune Cells Recruited to the Cornea and the Surface of the Lens in Response to Formation of Corneal Erosions. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:960-981. [PMID: 39889825 PMCID: PMC12016862 DOI: 10.1016/j.ajpath.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Injuries to the cornea can lead to recurrent corneal erosions, compromising its barrier function and increasing the risk of infection. Vital as corneal integrity is to the eye's optical power and homeostasis, the immune response to corneal erosions remains poorly understood. It is also unknown whether there is coordinated immune activation between the cornea and other regions of the anterior segment to protect against microbial invasion and limit the spread of inflammation when corneal erosions occur. Herein, a corneal debridement wounding model was used to characterize the immune cell phenotypes populating the cornea in response to erosion formation, and whether and which immune cells are concurrently recruited to the surface of the lens was investigated. The formation of corneal erosions induced an influx of myeloid lineage phenotypes, both M2 macrophages associated with tissue healing and wound repair, and Ly6G+ Ly6C+ myeloperoxidase+ cells resembling neutrophils/polymorphonuclear-myeloid-derived suppressor cells (PMN-MDSCs), with few regulatory T cells, into the corneal stroma under erosion sites. This leukocyte migration into the cornea when erosions develop was paralleled by the recruitment of immune cells, predominantly neutrophils/PMN-MDSCs, to the anterior, cornea-facing lens capsule. Both cornea-infiltrating and lens capsule-associated neutrophil/PMN-MDSC-like immune cells produce the anti-inflammatory cytokine IL-10. These findings suggest a collaborative role for the lens capsule-associated immune cells in preventing infections, controlling inflammation, and maintaining homeostasis of the anterior segment during recurrent corneal erosions.
Collapse
Affiliation(s)
- Phuong M Le
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - A Sue Menko
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
3
|
Shen Y, Lin P. The Role of Cytokines in Postherpetic Neuralgia. J Integr Neurosci 2025; 24:25829. [PMID: 40302252 DOI: 10.31083/jin25829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 05/02/2025] Open
Abstract
Nerve injury is a significant cause of postherpetic neuralgia (PHN). It is marked by upregulated expression of cytokines secreted by immune cells such as tumor necrosis factor alpha, interleukin 1 beta (IL-1β), IL-6, IL-18, and IL-10. In neuropathic pain (NP) due to nerve injury, cytokines are important for the induction of neuroinflammation, activation of glial cells, and expression of cation channels. The release of chemokines due to nerve injury promotes immune cell infiltration, recruiting inflammatory cytokines and further amplifying the inflammatory response. The resulting disequilibrium in neuroimmune response and neuroinflammation leads to a reduction of nerve fibers, altered nerve excitability, and neuralgia. PHN is a typical NP and cytokines may induce PHN by promoting central and peripheral sensitization. Currently, treating PHN is challenging and research on the role of cytokine signaling pathways in PHN is lacking. This review summarizes the potential mechanisms of cytokine-mediated PHN and discusses the cytokine signaling pathways associated with the central and peripheral sensitization of PHN. By elucidating the mechanisms of cytokines, the cells and molecules that regulate cytokines, and their signaling systems in PHN, this review reveals important research developments regarding cytokines and their signaling pathways mediating PHN, highlighting new targets of action for the development of analgesic drugs.
Collapse
Affiliation(s)
- Yunyan Shen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Ping Lin
- Department of Geriatrics, Hangzhou Third People's Hospital, 310009 Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Xi Y, Ma H, Liu X, Mu Q, An X, Li S, Liang H, Sun D, Ma R, Deng H, Wu Z, Zhang C, Liu G, Liu C. Epigenetically Reprogrammed Nanovesicles as Inverse Vaccines for Antigen-Specific Immune Tolerance in Autoimmune Diseases. NANO LETTERS 2025; 25:6725-6734. [PMID: 40213869 DOI: 10.1021/acs.nanolett.5c00986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The development of antigen-specific immunotherapy for autoimmune diseases constitutes an important unmet clinical need. Here we present an innovative inverse vaccine platform leveraging epigenetic reprogramming to induce durable antigen-specific immune tolerance. This inverse vaccine (mDCNVreg) is constructed using artificial cell membrane nanovesicles derived from IFN-γ-primed regulatory dendritic cells subjected to epigenetic modulation. The engineered mDCNVreg features upregulated MHC-II expression enabling targeted antigen presentation, suppressed costimulatory molecules expression, and an enhanced coinhibitory molecules display. Through coordinated mechanisms involving enhanced lymphoid trafficking and phenotype stabilization, this platform significantly enhances antigen delivery to secondary lymphoid organs while maintaining tolerogenic potency. Crucially, mDCNVreg directly induces CD4+ T cell clonal anergy through epitope-specific interactions, establishing long-lasting immune tolerance. This work demonstrates a promising epigenetic engineering approach for reverse vaccine design in personalized autoimmune disease therapy.
Collapse
Affiliation(s)
- Yue Xi
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Huifeng Ma
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xue Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qianwen Mu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiaoyu An
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Shuo Li
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Hao Liang
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Di Sun
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Rongrong Ma
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Haolan Deng
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhengyu Wu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Chenhao Zhang
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chao Liu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| |
Collapse
|
5
|
Camarano Eula MA, Bayona-Serrano JD, Nishiyama-Jr MY, Squaiella-Baptistão CC, Santoro ML, Junqueira-de-Azevedo IDLM. The underestimated local effects of Micrurus corallinus venom revealed by gene expression and histopathological alterations. Toxicon 2025; 259:108368. [PMID: 40268250 DOI: 10.1016/j.toxicon.2025.108368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
The mechanisms of action of elapid neurotoxins have been widely studied; however, the pathophysiological effects of these venoms, particularly from coral snakes, have not been extensively investigated. To gain a deeper understanding of the mechanisms involved in the local and systemic toxicity of Micrurus corallinus venom and their genomic responses, we injected mice with 2.70 μg of venom, corresponding to a sub-lethal dose (50 % of the LD50), and evaluated the effects using transcriptomic and histopathological approaches. mRNA was extracted from the liver, spleen, kidney, heart, brain, diaphragm, and both right and left gastrocnemius muscles of control and treated animals and subjected to RNA sequencing (RNA-Seq) to perform functional analyses of differentially expressed genes (DEGs). In the right gastrocnemius, the site of venom injection, we observed significant histopathological changes characterized by a pronounced local inflammatory response. Consistent with these findings, enrichment analyses revealed 2454 DEGs in the right gastrocnemius, mostly involved in inflammatory pathways. Systemically, the liver emerged as the most affected non-local organ, showing over 400 DEGs containing several up-regulated genes involved in the production of acute phase proteins. These results underscore that inflammation possibly induced by the sub-lethal amounts of venom typically injected during human envenomation, and not only the neurotoxicity, could be a potentially deleterious effect of venom and should not be ruled out when diagnosing envenomation caused by coral snakes.
Collapse
|
6
|
Stacchiotti C, Mazzella di Regnella S, Cinotti M, Spalloni A, Volpe E. Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. Int J Mol Sci 2025; 26:3854. [PMID: 40332510 PMCID: PMC12028049 DOI: 10.3390/ijms26083854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neuroinflammation is an inflammatory response occurring within the central nervous system (CNS). The process is marked by the production of pro-inflammatory cytokines, chemokines, small-molecule messengers, and reactive oxygen species. Microglia and astrocytes are primarily involved in this process, while endothelial cells and infiltrating blood cells contribute to neuroinflammation when the blood-brain barrier (BBB) is damaged. Neuroinflammation is increasingly recognized as a pathological hallmark of several neurological diseases, including amyotrophic lateral sclerosis (ALS), and is closely linked to neurodegeneration, another key feature of ALS. In fact, neurodegeneration is a pathological trigger for inflammation, and neuroinflammation, in turn, contributes to motor neuron (MN) degeneration through the induction of synaptic dysfunction, neuronal death, and inhibition of neurogenesis. Importantly, resolution of acute inflammation is crucial for avoiding chronic inflammation and tissue destruction. Inflammatory processes are mediated by soluble factors known as cytokines, which are involved in both promoting and inhibiting inflammation. Cytokines with anti-inflammatory properties may exert protective roles in neuroinflammatory diseases, including ALS. In particular, interleukin (IL)-10, transforming growth factor (TGF)-β, IL-4, IL-13, and IL-9 have been shown to exert an anti-inflammatory role in the CNS. Other recently emerging immune regulatory cytokines in the CNS include IL-35, IL-25, IL-37, and IL-27. This review describes the current understanding of neuroinflammation in ALS and highlights recent advances in the role of anti-inflammatory cytokines within CNS with a particular focus on their potential therapeutic applications in ALS. Furthermore, we discuss current therapeutic strategies aimed at enhancing the anti-inflammatory response to modulate neuroinflammation in this disease.
Collapse
Affiliation(s)
- Costanza Stacchiotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Mazzella di Regnella
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| | - Miriam Cinotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
| | - Alida Spalloni
- Molecular Neurobiology Unit, Santa Lucia Foundation, 00143 Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| |
Collapse
|
7
|
Lee B, Kwon JT, Jeong Y, Caris H, Oh D, Feng M, Davila Mejia I, Zhang X, Ishikawa T, Watson BR, Moffitt JR, Chung K, Huh JR, Choi GB. Inflammatory and anti-inflammatory cytokines bidirectionally modulate amygdala circuits regulating anxiety. Cell 2025; 188:2190-2202.e15. [PMID: 40199321 DOI: 10.1016/j.cell.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 11/17/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025]
Abstract
Patients with autoimmune or infectious diseases can develop persistent mood alterations after inflammatory episodes. Peripheral immune molecules, like cytokines, can influence behavioral and internal states, yet their impact on the function of specific neural circuits in the brain remains unclear. Here, we show that cytokines act as neuromodulators to regulate anxiety by engaging receptor-expressing neurons in the basolateral amygdala (BLA). Heightened interleukin-17A (IL-17A) and IL-17C levels, paradoxically induced from treatment with anti-IL-17 receptor A (IL-17RA) antibodies, promote anxiogenic behaviors by increasing the excitability of IL-17RA/RE-expressing BLA neurons. Conversely, the anti-inflammatory IL-10, acting on the same population of BLA neurons via its receptor, exerts opposite effects on neuronal excitability and behavior. These findings reveal that inflammatory and anti-inflammatory cytokines bidirectionally modulate anxiety by engaging their respective receptors in the same BLA population. Our results highlight the role of cytokine signaling in shaping internal states through direct modulation of specific neural substrates.
Collapse
Affiliation(s)
- Byeongjun Lee
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeong-Tae Kwon
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yire Jeong
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hannah Caris
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dongsun Oh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mengyang Feng
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Irene Davila Mejia
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xiaoying Zhang
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomoe Ishikawa
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brianna R Watson
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kwanghun Chung
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Bio2Q, Keio University, Tokyo, Japan; Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA, USA.
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
8
|
Maes M, Vasupanrajit A, Jirakran K, Zhou B, Tunvirachaisakul C, Almulla AF. Simple dysmood disorder, a mild subtype of major depression, is not an inflammatory condition: Depletion of the compensatory immunoregulatory system. J Affect Disord 2025; 375:75-85. [PMID: 39848470 DOI: 10.1016/j.jad.2025.01.101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/13/2024] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND A recent study conducted by the laboratory of the first author revealed that major depression is composed of two distinct subtypes: major dysmood disorder (MDMD) and simple dysmood disorder (SDMD). The latter is a less severe phenotype with fewer aberrant biological pathways. MDMD, but not SDMD, patients were identified to have highly sensitized cytokine/growth factor networks using stimulated whole blood cultures. However, no information regarding serum cytokines/chemokines/growth factors in SDMD is available. OBJECTIVES This case-control study compares 48 serum cytokines/chemokines/growth factors in academic students with SDMD (n = 64) and first episode (FE)-SDMD (n = 47) to those of control students (n = 44) using a multiplex assay. FINDINGS Both FE-SDMD and SDMD exhibited a notable inhibition of immune profiles, such as the compensatory immunoregulatory response system (CIRS) and alternative M2 macrophage and T helper-2 (Th-2) profiles. We observed a substantial reduction in the serum concentrations of five proteins: interleukin (IL)-4, IL-10, soluble IL-2 receptor (sIL-2R), IL-12p40, and macrophage colony-stimulating factor. A considerable proportion of the variability observed in suicidal behaviors (26.7 %) can be accounted for by serum IL-4, IL-10, and sIL-2R (all decreased), CCL11 (eotaxin) and granulocyte CSF (both increased). The same biomarkers (except for IL-10), accounted for 25.5 % of the variance in SDMS severity. A significant correlation exists between decreased levels of IL-4 and elevated ratings of the brooding type of rumination. CONCLUSIONS SDMD is characterized by the suppression of the CIRS profile, which signifies a disruption of immune homeostasis and tolerance, rather than the presence of an inflammatory response.
Collapse
Affiliation(s)
- Michael Maes
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Fitness and Biopsychological Technology Research Unit, Faculty of Medicine Chulalongkorn University, Bangkok 10330, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University of Plovdiv, Plovdiv, Bulgaria; Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Asara Vasupanrajit
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ketsupar Jirakran
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Center of Excellence for Maximizing Children's Developmental Potential, Department of Pediatric, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Bo Zhou
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China
| | - Chavit Tunvirachaisakul
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Ph.D. Program in Mental Health, Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Abbas F Almulla
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China; Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu 610072, China; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq.
| |
Collapse
|
9
|
Bai R, Ma L, Li F, Pan L, Bao Y, Li X, Wang S, Yue H, Zheng F. Total ginsenosides from wild ginseng improve immune regulation in a rat model of spleen qi deficiency by modulating fecal-bacteria-associated short-chain fatty acids and intestinal barrier integrity. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1256:124554. [PMID: 40081219 DOI: 10.1016/j.jchromb.2025.124554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/17/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
For thousands of years, traditional Chinese medicine (TCM) has made extensive use of wild ginseng. It is thought to provide vital energy effects and to boost immunity. This study aimed to clarify the processes by which short-chain fatty acids (SCFAs) metabolites and the intestinal barrier are used by total ginsenosides wild ginseng (TWG) to modulate immunity. In this study, we analyzed and identified ginsenosides in the colon using UPLC-Q-TOF-MSE methods. In the meantime, a rat model of spleen qi deficiency (SQD) was created using reserpine, and the effects of TWG on intestinal barrier function and short-chain fatty acids in the feces of SQD-affected rats were examined. 28 ginsenosides were found in the colon during this experiment, and the main components were measured. TWG considerably increased fecal concentrations of acetic, propionic and 6 others, according to SCFAs analysis. According to serum immunological markers, TWG reduced IL-17 and IL-1β levels, increased IL-10, IL-22, and TGF-β concentrations, balanced Th17/Treg ratios, and reduced toxicants such DAO and LPS in rats with SQD. TWG improved barrier function, reduced permeability, increased tight junction protein expression, and lessened intestinal injury. A favorable correlation between intestinal barrier proteins and fatty acids was shown by correlation studies. The gut barrier and SCFAs perspectives helped to clarify the mechanism by which TWG controls immune activity. This study offers a fresh theoretical framework for TWG's future advancement and application.
Collapse
Affiliation(s)
- Ruobing Bai
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Liting Ma
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Fangtong Li
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Lijia Pan
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Yuwen Bao
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Xinze Li
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Shen Wang
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China
| | - Hao Yue
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China.
| | - Fei Zheng
- Changchun University of Chinese Medicine, 130117 Changchun, Jilin, PR China.
| |
Collapse
|
10
|
Wang Y, Ming Y, Yu Z, Xu Z, Zou M, Chen C, Luo F, Huang D, Wang N, Lin Z, Weng Z. ROS-Responsive Cationic Polymers with Intrinsic Anti-Inflammatory Activity for Intracellular Protein Delivery. Biomacromolecules 2025; 26:2268-2281. [PMID: 40114391 DOI: 10.1021/acs.biomac.4c01593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The intracellular delivery of protein drugs via nanocarriers offers significant potential for expanding their therapeutic applications. However, the unintended activation of innate immune responses and inflammation triggered by the carriers presents a major challenge, often compromising therapeutic efficacy. Here, we present oligoethylenimine-thioketal (OEI-TK), a reactive oxygen species-responsive cationic polymer with intrinsic anti-inflammatory properties, to overcome this challenge. OEI-TK self-assembles electrostatically with bovine serum albumin (BSA) to form stable nanoparticles (OTB NPs) with excellent encapsulation efficiency. In vitro studies confirmed that OTB NPs retained OEI-TK's antioxidant and anti-inflammatory properties, enhanced biocompatibility, and efficiently delivered BSA into cells. Furthermore, OEI-TK facilitated the intracellular delivery of β-galactosidase while preserving its enzymatic activity, demonstrating its potential for functional protein transport. These findings highlight OEI-TK as a promising platform with dual benefits of inflammation modulation and intracellular protein delivery, holding potential for the synergistic treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Yongming Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yangcan Ming
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, Hubei 430022, China
| | - Zhichao Yu
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zhenjin Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Minglang Zou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Cuiping Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Fang Luo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Na Wang
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, Hubei 430022, China
| | - Zhenyu Lin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zuquan Weng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| |
Collapse
|
11
|
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:S0022-0302(25)00217-6. [PMID: 40221035 DOI: 10.3168/jds.2024-26074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [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 of MP/kg DM) or high (113 g of MP/kg DM) level of estimated MP. Postpartum diets were formulated to contain either a control (104 g of MP/kg DM) or high (131 g of 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), interleukin-10 (IL-10), and interferon-γ (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 polymorphonuclear leukocyte (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, respectfully. 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, while 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
|
12
|
Masmoudi D, Villalba M, Alix-Panabières C. Natural killer cells: the immune frontline against circulating tumor cells. J Exp Clin Cancer Res 2025; 44:118. [PMID: 40211394 PMCID: PMC11983744 DOI: 10.1186/s13046-025-03375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Natural killer (NK) play a key role in controlling tumor dissemination by mediating cytotoxicity towards cancer cells without the need of education. These cells are pivotal in eliminating circulating tumor cells (CTCs) from the bloodstream, thus limiting cancer spread and metastasis. However, aggressive CTCs can evade NK cell surveillance, facilitating tumor growth at distant sites. In this review, we first discuss the biology of NK cells, focusing on their functions within the tumor microenvironment (TME), the lymphatic system, and circulation. We then examine the immune evasion mechanisms employed by cancer cells to inhibit NK cell activity, including the upregulation of inhibitory receptors. Finally, we explore the clinical implications of monitoring circulating biomarkers, such as NK cells and CTCs, for therapeutic decision-making and emphasize the need to enhance NK cell-based therapies by overcoming immune escape mechanisms.
Collapse
Affiliation(s)
- Doryan Masmoudi
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, Montpellier, IRD, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- LCCRH, Site Unique de Biologie (SUB), 641, Avenue du Doyen Gaston Giraud, Montpellier, 34093, France.
| |
Collapse
|
13
|
Jonsson A, Korsgren O, Hedin A. Transcriptomic characterization of human pancreatic CD206- and CD206 + macrophages. Sci Rep 2025; 15:12037. [PMID: 40199933 PMCID: PMC11978877 DOI: 10.1038/s41598-025-96313-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: 11/26/2024] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Macrophages reside in all organs and participate in homeostatic- and immune regulative processes. Little is known about pancreatic macrophage gene expression. In the present study, global gene expression was characterized in human pancreatic macrophage subpopulations. CD206- and CD206 + macrophages were sorted separately from pancreatic islets and exocrine tissue to high purity using flow cytometry, followed by RNA-seq analysis. Comparing CD206- with CD206 + macrophages, CD206- showed enrichment in histones, proliferation and cell cycle regulation, glycolysis and SPP1-associated immunosuppressive polarization while CD206 + showed enrichment in complement and coagulation-, IL-10 and IL-2RA immune regulation, as well as scavenging-related gene sets. Comparing islet CD206- with exocrine CD206-, enrichments in islet samples included two sets involved in immune regulation, while enrichments in exocrine samples included sets related to extracellular matrix and immune activation. Fewer differences were found between CD206 + macrophages, with enrichments in islet samples including two IL2-RA related gene sets, while enrichments in exocrine samples included sets related to extracellular matrix and immune activation. Comparing macrophages between individuals with normoglycemia, elevated HbA1c or type 2 diabetes, only a few diverse differentially expressed genes were identified. This work characterizes global gene expression and identifies differences between CD206- and CD206 + macrophage populations within the human pancreas.
Collapse
Affiliation(s)
- Alexander Jonsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anders Hedin
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Moradi K, Moghaddami R, Ghaffari-Nasab A, Khordadmehr M, Pagheh AS, Mosajakhah H, Rezaei S, Gharepapagh E, Ahmadi M, Montazeri M, Pazoki H, Ahmadpour E. Toxoplasma gondii modulates immune responses and mitigates type 1 diabetes progression in a streptozotocin-induced rat model. Cell Commun Signal 2025; 23:172. [PMID: 40200271 PMCID: PMC11980074 DOI: 10.1186/s12964-025-02168-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/22/2025] [Indexed: 04/10/2025] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by the destruction of insulin-producing β-cells in the pancreas. Emerging evidence suggests that infections, including Toxoplasma gondii (T. gondii), may modulate immune responses and influence disease outcomes. This study aimed to investigate the effects of T. gondii infection on the development of T1DM in a Streptozotocin (STZ)-induced rat model, with an emphasis on immune modulation, cytokine profiles, and organ inflammation. In rats experimentally infected with pathogenic and non-pathogenic Toxoplasma strains, diabetes was induced via STZ injection and compared to a control group. Blood glucose levels and the expression of IL-10, IL-1β, and TNF-α at both gene and protein levels were assessed. Histopathological examinations of the pancreas and kidneys were conducted, alongside small-animal PET scans to evaluate metabolic activity in these organs. The T. gondii-infected diabetic groups showed reduced blood glucose levels, increased IL-10, and decreased TNF-α and IL-1β levels compared to the STZ group. Histopathological and PET imaging analyses revealed improved pancreatic and renal tissues and reduced metabolic activity, indicating improvement effects associated with decreased inflammation and immune modulation. T. gondii infection seems to influence immune responses and slow the progression of T1DM in a rat model. These results suggest a potential therapeutic role for parasitic infections in autoimmune diseases, offering valuable insights into the complex relationship between infections, immune regulation, and metabolic health.
Collapse
Affiliation(s)
- Kimia Moradi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Moghaddami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arshad Ghaffari-Nasab
- School of Nursing and Allied Medical Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Monir Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Abdol Sattar Pagheh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Mosajakhah
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Rezaei
- Department of Nuclear Medicine, Medical School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Gharepapagh
- Department of Nuclear Medicine, Medical School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Ahmadi
- School of Medical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Mahbobeh Montazeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Pazoki
- Faculty of Medicine, Department of Medical Microbiology, Infectious Diseases Research Center, Gonabad University of Medical Science, Gonabad, Iran
| | - Ehsan Ahmadpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Lin X, Li X, Zhai Z, Zhang M. JAK-STAT pathway, type I/II cytokines, and new potential therapeutic strategy for autoimmune bullous diseases: update on pemphigus vulgaris and bullous pemphigoid. Front Immunol 2025; 16:1563286. [PMID: 40264772 PMCID: PMC12011800 DOI: 10.3389/fimmu.2025.1563286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Autoimmune Bullous Diseases (AIBDs), characterized by the formation of blisters due to autoantibodies targeting structural proteins, pose significant therapeutic challenges. Current treatments, often involving glucocorticoids or traditional immunosuppressants, are limited by their non-specificity and side effects. Cytokines play a pivotal role in AIBDs pathogenesis by driving inflammation and immune responses. The JAK-STAT pathway is central to the biological effects of various type I and II cytokines, making it an attractive therapeutic target. Preliminary reports suggest that JAK inhibitors may be a promising approach in PV and BP, but further clinical validation is required. In AIBDs, particularly bullous pemphigoid (BP) and pemphigus vulgaris (PV), JAK inhibitors have shown promise in modulating pathogenic cytokine signaling. However, the safety and selectivity of JAK inhibitors remain critical considerations, with the potential for adverse effects and the need for tailored treatment strategies. This review explores the role of cytokines and the JAK-STAT pathway in BP and PV, evaluating the therapeutic potential and challenges associated with JAK inhibitors in managing these complex disorders.
Collapse
Affiliation(s)
| | | | - Zhifang Zhai
- Department of Dermatology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Mingwang Zhang
- Department of Dermatology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
16
|
Yeshna, Singh M, Monika, Kumar A, Garg V, Jhawat V. Pathophysiology and emerging therapeutic strategies for cervical spondylosis: The role of pro-inflammatory mediators, kinase inhibitors, and Organogel based drug delivery systems. Int Immunopharmacol 2025; 151:114350. [PMID: 40010157 DOI: 10.1016/j.intimp.2025.114350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Cervical spondylosis is a prevalent ailment characterized by chronic wear and degenerative changes affecting the cervical spine, leading to various clinical syndromes such as axial neck pain, cervical myelopathy, and cervical radiculopathy. The pathophysiology of the development of cervical alterations is multifaceted, with alterations in the normal physiology and pathogenesis of intervertebral disc degeneration. The involvement of pro-inflammatory mediators, such as interleukin-1, tumor necrosis factor-α, interleukin-4, interleukin-6, and interleukin-10, in the pathological processes associated with intervertebral disc degeneration offers potential therapeutic targets. The review also introduces kinase inhibitors as potential treatments for cervical spondylosis. Protein kinase inhibitors, including mitogen-activated protein kinase (MAPK), Janus kinase (JAK), and spleen tyrosine kinase (SYK), are explored for their anti-inflammatory properties. The article discusses their potential in modulating inflammatory signaling cascades and presents them as attractive candidates for treating immune-mediated disorders. Inhibitors of Nuclear Factor-κB, p38 MAPK, Jun-N terminal kinase (JNK), and Extracellular signal-regulated kinase (ERK) have shown efficacy in suppressing inflammatory responses, offering potential avenues for intervention in this prevalent condition. Organogels are semi-solid materials formed by trapping an organic solvent within a three-dimensional cross-linked network. They hold considerable potential in drug delivery, especially in enhancing drug solubility, facilitating controlled release, and improving skin penetration. These properties of organogels can help treat or alleviate the symptoms of cervical spondylosis.
Collapse
Affiliation(s)
- Yeshna
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika Singh
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Ashok Kumar
- Faculty of Pharmacy, Kalinga University, Naya Raipur, Chhattisgarh, India
| | - Vandana Garg
- Department of Pharmaceutical Science, MD University, Rohtak, India
| | - Vikas Jhawat
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India.
| |
Collapse
|
17
|
Chang Z, Guo X, Li X, Wang Y, Zang Z, Pei S, Lu W, Li Y, Huang JD, Xiao Y, Liu C. Bacterial immunotherapy leveraging IL-10R hysteresis for both phagocytosis evasion and tumor immunity revitalization. Cell 2025; 188:1842-1857.e20. [PMID: 40037354 DOI: 10.1016/j.cell.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 12/07/2024] [Accepted: 02/05/2025] [Indexed: 03/06/2025]
Abstract
Bacterial immunotherapy holds promising cancer-fighting potential. However, unlocking its power requires a mechanistic understanding of how bacteria both evade antimicrobial immune defenses and stimulate anti-tumor immune responses within the tumor microenvironment (TME). Here, by harnessing an engineered Salmonella enterica strain with this dual proficiency, we unveil an underlying singular mechanism. Specifically, the hysteretic nonlinearity of interleukin-10 receptor (IL-10R) expression drives tumor-infiltrated immune cells into a tumor-specific IL-10Rhi state. Bacteria leverage this to enhance tumor-associated macrophages producing IL-10, evade phagocytosis by tumor-associated neutrophils, and coincidently expand and stimulate the preexisting exhausted tumor-resident CD8+ T cells. This effective combination eliminates tumors, prevents recurrence, and inhibits metastasis across multiple tumor types. Analysis of human samples suggests that the IL-10Rhi state might be a ubiquitous trait across human tumor types. Our study unveils the unsolved mechanism behind bacterial immunotherapy's dual challenge in solid tumors and provides a framework for intratumoral immunomodulation.
Collapse
Affiliation(s)
- Zhiguang Chang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xuan Guo
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xuefei Li
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongsheng Zang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiqi Lu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yang Li
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jian-Dong Huang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China; Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chenli Liu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
18
|
Sim M, Hong S, Jung MH, Choi EY, Hwang GS, Shin DM, Kim CS. Gut microbiota links vitamin C supplementation to enhanced mental vitality in healthy young adults with suboptimal vitamin C status: A randomized, double-blind, placebo-controlled trial. Brain Behav Immun 2025; 128:179-191. [PMID: 40187667 DOI: 10.1016/j.bbi.2025.03.032] [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: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
The intricate relationship between nutrition, gut microbiome, and mental health has gained increasing attention. We aimed to determine how vitamin C supplementation improves mental vitality through the gut microbiome and associated neurological and immunological changes. We used 16S rRNA sequencing to analyze gut microbiota profiles of participants from our previous trial, in which healthy young adults (20-39 years) with inadequate serum vitamin C levels (< 50 μM) received 500 mg vitamin C or a placebo twice daily for 4 weeks (vitamin C, n = 21; placebo, n = 19). We examined whether changes in gut microbiota correlated with previously determined mental vitality indices, including Stroop test performance, work engagement, and serum brain-derived neurotrophic factor (BDNF) levels. Serum concentrations of microbial-derived molecules, cytokines, and neurotransmitters were analyzed using enzyme-linked immunosorbent assay, electrochemiluminescence-based immunoassay, or ultra-high-performance liquid chromatography-mass spectrometry. Monocyte subpopulations in peripheral blood were quantified using fluorescence-activated cell sorting analysis. Vitamin C supplementation increased the relative abundance of Bacillaceae and Anaerotruncus, while decreasing Desulfovibrio, with the Desulfovibrio reduction correlating with Stroop test performance. Moreover, participants showing a substantial Desulfovibrio reduction ("responders") demonstrated greater BDNF increases and stronger correlations between serum L-DOPA levels and work engagement scores than did non-responders. In addition, vitamin C supplementation suppressed inflammatory responses with concurrent reduction in serum lipopolysaccharide levels, and responders showed greater decreases in IL-10 levels and classical monocyte frequencies than non-responders. In conclusion, vitamin C supplementation modulates gut microbiota composition, particularly by reducing Desulfovibrio abundance, with the extent of reduction correlating with mental vitality improvements and decreased inflammation. This study provides insights into vitamin C supplementation as a critical dietary intervention, as it may modulate mental health through its influence on the gut-brain-immune axis.
Collapse
Affiliation(s)
- Minju Sim
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Sehwa Hong
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Min Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Republic of Korea
| | - Chong-Su Kim
- Department of Food and Nutrition, Seowon University, Cheongju 28674, Republic of Korea.
| |
Collapse
|
19
|
Alrefaee SH, Aljohani FS, El-Khatib M, Shahin YH, Elwakil BH, Shahin SH, Akl SH, Moneer EA, Darwish AAE. Evaluation of potential antiparasitic effect of ZnO nanoparticles on experimental cryptosporidiosis in immunosuppressed mice. Biometals 2025; 38:647-661. [PMID: 40009283 DOI: 10.1007/s10534-025-00669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025]
Abstract
Cryptosporidium is a food and water-borne enteric protozoan that infects a wide range of vertebrates, causing life-threatening complications, particularly in immunocompromised hosts. The absence of effective anti-cryptosporidial medications could be attributed to the parasite's specific intestinal location, as well as the lack of research into the mechanism by which the protozoan impairs intestine cellular function. The present work aimed to evaluate the in vivo efficacy of zinc nanoparticles in the treatment of experimental cryptosporidiosis infection in immunosuppressed mice. Small-sized ZnO-NPs revealed better treatment efficacy than Large-sized ZnO-NPs in all studies. Nitazoxanide-treated group revealed the highest percentage reduction of the oocyst's counts followed by the small-sized ZnO-NPs treated group. The small-sized ZnO-NPs treated mice group showed a minimal inflammatory effect in all examined treated tissues when compared to the infected non-treated group. The morphological structure of the oocysts was examined using SEM indicating variable degrees of morphological changes in the treated mice. Moreover, the levels of biochemical analyses were significantly lower in the treated group. The histopathological study revealed the significant effect of small-sized ZnO-NPs in treating cryptosporidiosis.
Collapse
Affiliation(s)
- Salhah Hamed Alrefaee
- Department of Chemistry, College of Science, Taibah University, 30799, Yanbu, Saudi Arabia
| | - Faizah S Aljohani
- Department of Chemistry, College of Science, Taibah University, 30002, Al-Madinah Al-Munawarah, Saudi Arabia
| | - M El-Khatib
- Department of Basic Sciences, Faculty of Computer Science and Artificial Intelligence, Pharos University, Alexandria, Egypt
| | - Yahya H Shahin
- Department of Medical Laboratory Technology, Faculty of Health and Medical Techniques, Almaaqal University, Basrah, Iraq
| | - Bassma H Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt.
| | - Sendianah H Shahin
- Department of Economics and Agribusiness, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| | - Sara H Akl
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt
| | - Esraa Abdelhamid Moneer
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt
| | - Amira Abd-Elfattah Darwish
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, 21526, Egypt
| |
Collapse
|
20
|
O'Garra A. From Cytokines to Tuberculosis and Back: My Journey to Understanding the Immune Response to Infection. Annu Rev Immunol 2025; 43:1-28. [PMID: 40279305 DOI: 10.1146/annurev-immunol-010824-041601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
I felt honored by the invitation to write this autobiography, although it was an arduous task to describe my journey through science: first bacterial adhesion, then cytokine function, and then immune responses in tuberculosis. Since only seven women had been authors of autobiographies for the Annual Review of Immunology, I felt I couldn't refuse to contribute to Volume 43 of the journal. Moreover, this was a good occasion to record my appreciation to all the lab members and collaborators for their contributions over the last 40 years, to remember the exciting times, and to reflect on the obstacles we faced. I often reflect on this line that is commonly attributed to Winston Churchill: Success is not final; failure is not fatal: It is the courage to continue that counts. What kept me going was a burning desire to know how things work and find enjoyment in the discovery. This passion to understand immune responses to infection remains with me to this day. I thank all those I have interacted with for the support and friendship they provided.
Collapse
Affiliation(s)
- Anne O'Garra
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
21
|
Islam H, Jackson GS, Boultbee J, Tsai SH, Moreno-Cabañas A, Teixeira AADS, Wright DC, Mui AL, Little JP. Interleukin-10 resistance in type 2 diabetes is associated with defective STAT3 signaling in human blood leukocytes. Am J Physiol Cell Physiol 2025; 328:C1293-C1302. [PMID: 40080435 DOI: 10.1152/ajpcell.00124.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Chronic inflammation is strongly implicated in the pathophysiology of type 2 diabetes (T2D), highlighting the need to better understand inflammatory processes in people living with T2D. Hyperglycemia blunts the anti-inflammatory actions of interleukin-10 (IL-10)-the most potent anti-inflammatory cytokine-but the mechanistic basis remains unclear. To test the hypothesis that signaling defects underpin this hyporesponsiveness to IL-10 action, fasted blood samples were obtained from individuals living with T2D (n = 17, age: 64 ± 9 yr, HbA1c: 7.2 ± 1.1%) and their age-matched counterparts without diabetes (n = 19, 65 ± 8 yr, 5.5 ± 0.3%). Blood leukocytes were analyzed for IL-10-mediated signaling, gene expression, and cytokine secretion using flow cytometry, qPCR, and whole blood cultures, respectively. Despite no overt elevations in circulating pro- and anti-inflammatory cytokine concentrations, blood leukocytes from individuals with T2D exhibited exaggerated cytokine secretion when exposed to lipopolysaccharide (LPS) (P < 0.05). IL-10's ability to activate its canonical transcription factor signal transducer and activator of transcription 3 (STAT3) was blunted in CD14 monocytes and CD4 lymphocytes from people with T2D (P < 0.01)-a defect associated with lower IL-10 receptor expression on both cell types (P < 0.05). This upstream signaling defect was accompanied by attenuated suppressor of cytokine signaling 3 mRNA levels in IL-10-treated mononuclear cells (P = 0.059) and higher lipopolysaccharide (LPS)-stimulated cytokine secretion from blood leukocytes exposed to IL-10 (P < 0.01). Our findings identify defective IL-10-mediated signaling and gene expression as a potential mechanism underpinning IL-10 resistance in T2D, highlighting the need for further investigation into therapeutic approaches targeting IL-10.NEW & NOTEWORTHY Our findings demonstrate that immune cells from people with type 2 diabetes (T2D) are less responsive to the anti-inflammatory actions of interleukin-10 (IL-10), which may drive chronic inflammation in this population. We identify T2D-associated defects at multiple steps of the IL-10 cascade-including IL-10 receptor expression, STAT3 signaling, SOCS3 mRNA, and cytokine secretion. Our findings highlight defective IL-10 action as a potential therapeutic target to ameliorate inflammation in T2D.
Collapse
Affiliation(s)
- Hashim Islam
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Garett S Jackson
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Jordan Boultbee
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| | - Shun-Hsi Tsai
- Office of Physical Education, National Taipei University, New Taipei City, Taiwan
| | | | | | - David C Wright
- School of Kinesiology, The University of British Columbia, Vancouver, British Columbia, Canada
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Alice L Mui
- Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan P Little
- School of Health and Exercise Sciences, The University of British Columbia, Kelowna, British Columbia, Canada
| |
Collapse
|
22
|
Hua Q, Li Z, Weng Y, Wu Y, Zheng L. Myeloid cells: key players in tumor microenvironments. Front Med 2025; 19:265-296. [PMID: 40048137 DOI: 10.1007/s11684-025-1124-8] [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: 08/14/2024] [Accepted: 12/16/2024] [Indexed: 05/04/2025]
Abstract
Cancer is the result of evolving crosstalk between neoplastic cell and its immune microenvironment. In recent years, immune therapeutics targeting T lymphocytes, such as immune checkpoint blockade (ICB) and CAR-T, have made significant progress in cancer treatment and validated targeting immune cells as a promising approach to fight human cancers. However, responsiveness to the current immune therapeutic agents is limited to only a small proportion of solid cancer patients. As major components of most solid tumors, myeloid cells played critical roles in regulating the initiation and sustentation of adaptive immunity, thus determining tumor progression as well as therapeutic responses. In this review, we discuss emerging data on the diverse functions of myeloid cells in tumor progression through their direct effects or interactions with other immune cells. We explain how different metabolic reprogramming impacts the characteristics and functions of tumor myeloid cells, and discuss recent progress in revealing different mechanisms-chemotaxis, proliferation, survival, and alternative sources-involved in the infiltration and accumulation of myeloid cells within tumors. Further understanding of the function and regulation of myeloid cells is important for the development of novel strategies for therapeutic exploitation in cancer.
Collapse
Affiliation(s)
- Qiaomin Hua
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhixiong Li
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yulan Weng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yan Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Limin Zheng
- Guangdong Provincial Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
23
|
Nakamura S, Minato KI, Mizuno M. Interleukin 10 and hydrogen peroxide mediate anti-allergic activity of polysaccharide from Pyropia yezoensis f. narawaensis. Int J Biol Macromol 2025; 308:142547. [PMID: 40157665 DOI: 10.1016/j.ijbiomac.2025.142547] [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: 12/06/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
In recent years, anti-allergic effects of food factors have been attracting attention as a treatment for allergic diseases with fewer side effects. Previous study demonstrated that oral administration of the polysaccharide from Pyropia yezoensis f. narawaensis (PPY) to mice improved allergic responses, and that the suppressive effect involved an increase in IL-10 secretion into the blood. However, it was not clear how IL-10 secretion was related to the anti-allergic activity of oral administration of PPY. The aim of the present study was to investigate the mechanism of anti-allergic effect of PPY. PPY inhibited allergic responses without suppressing the increase in serum IgE levels in mice model of active cutaneous anaphylaxis, suggesting that PPY suppressed allergy by affecting post-sensitization phase. PPY did not inhibit cell degranulation when RBL-2H3 cells were treated directly with IL-10 alone, but coexistence of IL-10 and hydrogen peroxide (H2O2) inhibited its degranulation. Furthermore, PPY increased H2O2 release from HT-29 cells, but pretreatment with DPI, an NADPH oxidase inhibitor, suppressed H2O2 production. Therefore, when N-acetylcysteine, a reactive oxygen species scavenger, was administered orally at the simultaneous time with PPY, the PPY-induced suppression of ear edema was eliminated. In conclusion, the results of the present study indicate that PPY exerts its anti-allergic effects through IL-10 production from intestinal epithelial cells with NADPH oxidase-mediated H2O2 production.
Collapse
Affiliation(s)
- Shunya Nakamura
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
| | - Ken-Ichiro Minato
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Meijo University, 1-501, Shiogamaguchi, Nagoya 468-8502, Japan
| | - Masashi Mizuno
- Department of Health and Nutrition, Faculty of Human Science, Osaka Aoyama University, 2-11-1 Niina, Minoh, Osaka 562-8580, Japan.
| |
Collapse
|
24
|
Jooss T, Maier K, Reichardt LM, Hindelang B, Süberkrüb L, Hamberger KL, Bülow JM, Schuetze K, Gebhard F, Mannes M, Halbgebauer R, Wohlgemuth L, Huber-Lang M, Relja B, Bergmann CB. Dynamic functional assessment of T cells reveals an early suppression correlating with adverse outcome in polytraumatized patients. Front Immunol 2025; 16:1538516. [PMID: 40196124 PMCID: PMC11973370 DOI: 10.3389/fimmu.2025.1538516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Most trauma patients require intensive care treatment and are susceptible to developing persistent inflammation and immunosuppression, potentially leading to multi organ dysfunction syndrome (MODS) and dependence on long term care facilities. T cells undergo changes in numbers and function post trauma. T cell dysfunction in polytraumatized patients was characterized using functional immunomonitoring to predict individual clinical outcome. Moreover, the potential to reverse T cell dysfunction using Interleukin (IL)-7 was examined. Methods Blood samples were drawn from healthy individuals and prospectively enrolled polytrauma patients (Injury Severity Score ≥ 18) on admission, 8, 24 and 48 hours, 5 and 10 days after. CD3/28-stimulated cytokine production of T cells in whole blood was assessed via Enzyme Linked Immuno Spot (ELISpot). T cell subsets were quantified via counting and flow cytometry. Unfavorable physical performative outcome was defined as death or new functional disability necessitating long term care. Secondary outcomes were the development of MODS and in-hospital mortality. IL-7 was added ex vivo to test reversibility of cytokine disturbances. Results 34 patients were enrolled. The different outcome groups showed no difference in injury severity. Patients with favorable physical performative outcome revealed higher functional T cell specific Interferon γ (IFN-γ) and IL-17 (8 hours) and lower IL-10 production (day 5) and higher CD8 T cell concentrations. Patients without MODS development showed a higher IFN-γ (day 10), higher IL-2 (8 hours) and higher IL-17 production (admission, day 5). There were no differences regarding in-hospital mortality. Systemic blood IFN-γ, IL-2 and IL-10 concentrations only correlated with MODS (24 hours). Systemic CD8 T cell numbers correlated with functional IFN-γ production. Whole blood stimulation with IL-7 increased functional T cell IFN-γ release. Discussion Our study reveals an early characteristic overall T cell dysfunction of pro-inflammatory (IFN-γ, IL-2, IL-17) and immunosuppressive (IL-10) subtypes in polytraumatized patients. Our data indicates that rather the functional capacity of T cells to release cytokines, but not systemic cytokine concentrations can be used to predict outcome post trauma. We assume that the early stimulation of pro- and anti-inflammatory T cells benefits polytraumatized patients. Potentiation of functional IFN-γ release might be achieved by IL-7 administration.
Collapse
Affiliation(s)
- Tobias Jooss
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Katharina Maier
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Lena-Marie Reichardt
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Bianca Hindelang
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Lönna Süberkrüb
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Kim Lena Hamberger
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Maria Bülow
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Konrad Schuetze
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Florian Gebhard
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Medical Center, Ulm, Germany
| | - Borna Relja
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| | - Christian B. Bergmann
- Translational and Experimental Trauma Research, Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
25
|
Gong G, Yun M, Kwon O, Kim B. Therapeutic and Pharmaceutical Potential of Scutellaria baicalensis-Derived Exosomes for Oily Skin Disorders. Antioxidants (Basel) 2025; 14:364. [PMID: 40227405 PMCID: PMC11939588 DOI: 10.3390/antiox14030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Fine dust exposure worsens oily skin by disrupting lipid metabolism and triggering oxidative inflammation. Scutellaria baicalensis extract-induced exosomes (SBEIEs) have shown anti-inflammatory effects by suppressing reactive oxygen species (ROS) and lipid-regulating properties, making them potential therapeutic agents. METHODS Exosomes from fibroblasts treated with SBEIEs and PM10 were tested on macrophages, adipose-derived stem cells (ASCs), and T lymphocytes. ELISA, flow cytometry, and PCR measured cytokines and gene expression. A 10-day clinical trial evaluated skin hydration, oiliness, and inflammation. RESULTS SBEIEs increased IRF3 (1.6 times) and suppressed PPARγ in ASCs while enhancing lipolysis markers. Sebaceous gland activity (squalene synthase) decreased by 10%. Macrophages showed increased IRF3, IFN-β, and IL-10 (2.1 times). T cells secreted IL-4 and IL-22 (2-2.33 times). Clinically, SBEIEs improved hydration (21%), reduced oiliness (1.6 times), and decreased inflammation (2.2 times). CONCLUSIONS SBEIEs effectively regulate lipid metabolism, cytokines, and immune responses, showing promise to treat oily and inflamed skin caused by fine dust exposure. Further studies are needed for clinical applications.
Collapse
Affiliation(s)
- Guybin Gong
- Department of Management of Beauty and Design, College of Design, Hansung University, Seoul 02876, Republic of Korea; (G.G.); (O.K.)
| | - Mihae Yun
- Department of Dental Hygiene, Andong Science College, Andong 36729, Republic of Korea;
| | - Ohhyuk Kwon
- Department of Management of Beauty and Design, College of Design, Hansung University, Seoul 02876, Republic of Korea; (G.G.); (O.K.)
| | - Boyong Kim
- EVERBIO, 131, Jukhyeon-gil, Gwanghyewon-myeon, Jincheon-gun 27809, Republic of Korea
| |
Collapse
|
26
|
Sun S, Ding Y, Yang D, Shen J, Zhang T, Song G, Chen X, Lin Y, Chen R. Identification of potential hub genes and drugs in septic kidney injury: a bioinformatic analysis with preliminary experimental validation. Front Med (Lausanne) 2025; 12:1502189. [PMID: 40166075 PMCID: PMC11955678 DOI: 10.3389/fmed.2025.1502189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/13/2025] [Indexed: 04/02/2025] Open
Abstract
Background Sepsis-associated kidney injury (SAKI) is a prevalent complication in intensive care unit (ICU) patients with sepsis. Diagnosis currently relies on clinical assessment, urine output, and serum creatinine levels, yet effective clinical treatments remain scarce. Our objectives are to explore prospective, targeted medications for the treatment of septic kidney injury and to employ bioinformatics to identify key genes and pathways that may be implicated in the pathogenesis of SAKI. Methods We utilized the GEO database for differential gene screening. Related genes of septic kidney injury were identified through Pubmed2Ensembl, followed by annotation and visualization of gene ontology biological processes and KEGG pathways using DAVID. Protein-protein interactions were analyzed with the STRING database, and hub genes were identified using Cytoscape software. Candidate genes were further validated through Metascape. The CTD database was employed to uncover the relationship between hub genes and acute kidney injury (AKI). CIBERSORT was applied to evaluate the infiltration of immune cells and their association with hub genes. Hub genes were experimentally verified through qPCR detection. Lastly, the Drug-Gene Interaction Database (DGIdb) was utilized to identify drug-gene interactions. Results Six genes, including TNF, CXCL8, IL-6, IL-1β, IL-2, and IL-10, were associated with three major signaling pathways: the COVID-19 adverse outcome pathway, an overview of pro-inflammatory and pro-fibrotic mediators, and the interleukin-10 signaling pathway. Additionally, 12 targeted drugs were identified as potential therapeutic agents.
Collapse
Affiliation(s)
- Shujun Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Dong Yang
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiwei Shen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Guobin Song
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yun Lin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Rui Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Anesthesiology, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
27
|
Wu F, Liu D, Xia X, Yang X, Huang S, Jiang X, LuLi. Association of IL-1RAcP rs16865597 gene polymorphism with susceptibility to essential hypertension: a case-control study in the Chinese Han population. BMC Cardiovasc Disord 2025; 25:172. [PMID: 40075282 PMCID: PMC11900220 DOI: 10.1186/s12872-025-04629-4] [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: 10/02/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The IL-33/ST2 pathway plays a crucial role in the development of essential hypertension (EH). This study aimed to investigate the relationship between EH and genetic variations in this pathway in the Chinese Han population. METHODS A total of 1,151 EH patients and 1,135 healthy controls were included in the study. Sixteen single nucleotide polymorphisms (SNPs) in the interleukin-33 (IL-33) and interleukin-1receptor associated protein (IL-1RAcP) genes were genotyped using the Sequenom MassArray and TaqMan assays. Genotype and allele frequencies were compared between the EH patients and controls using logistic regression analysis. RESULTS The rs16865597 SNP in the IL-1RAcP gene was found to be associated with the risk of EH. Specifically, the presence of the C allele of rs16865597 was negatively correlated with EH susceptibility in both the additive model (P = 0.014, OR = 0.75, 95% CI = 0.59-0.94) and the recessive model (P = 0.011, OR = 0.72, 95% CI = 0.56-0.93). Additionally, rs16865597 was linked to a reduced risk of EH in specific subgroups, including males (OR add = 0.73, 95% CI = 0.56-0.94, P = 0.015), nonsmokers (OR add = 0.72, 95% CI = 0.54-0.96, P = 0.023), nondrinkers (OR add = 0.70, 95% CI = 0.53-0.93, P = 0.013), and individuals with low BMI (OR add = 0.69, 95% CI = 0.51-0.92, P = 0.013). Moreover, the C genotype of rs16865597 was strongly associated with higher interleukin-10 levels in vivo. CONCLUSION The rs16865597 SNP is significantly associated with a reduced risk of EH in the Chinese Han population, potentially due to its role in immune regulation.
Collapse
Affiliation(s)
- Fangqin Wu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, 1Minde Road, Nanchang, China
| | - Dongchen Liu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xin Xia
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, 1Minde Road, Nanchang, China
| | - Xinlei Yang
- Biobank Center, The Second Affiliated Hospital of Nanchang University, No. 1 MinDe Road, Nanchang, China
| | - Suli Huang
- Department of Environment and Health, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xinghua Jiang
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, 1Minde Road, Nanchang, China
| | - LuLi
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China.
| |
Collapse
|
28
|
Yang S, Shi J, Qiao Y, Teng Y, Zhong X, Wu T, Liu C, Ge J, Yang H, Zou J. Harnessing Anti-Inflammatory and Regenerative Potential: GelMA Hydrogel Loaded with IL-10 and Kartogenin for Intervertebral Disc Degeneration Therapy. ACS Biomater Sci Eng 2025; 11:1486-1497. [PMID: 39846724 DOI: 10.1021/acsbiomaterials.4c01864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Intervertebral disc degeneration (IVDD) is a major contributor to chronic back pain and disability, with limited effective therapeutic options. Current treatment options, including conservative management and surgical interventions, often fail to effectively halt disease progression and come with notable side effects. IVDD is characterized by the breakdown of the extracellular matrix (ECM) and the infiltration of inflammatory cells, which exacerbate disc degeneration. This study presents a novel therapeutic strategy aimed at addressing the dual challenges of inflammation and ECM degradation in IVDD. We developed a gelatin methacryloyl (GelMA) hydrogel system loaded with interleukin-10 (IL-10), an anti-inflammatory cytokine, and kartogenin (KGN), a small-molecule compound known for its regenerative properties. The KGN + IL-10@GelMA hydrogel was designed to deliver these agents in a controlled manner directly to the degenerated disc, targeting both the inflammatory microenvironment and the promotion of nucleus pulposus (NP) tissue regeneration. In a puncture-induced IVDD model, this hydrogel system effectively delayed the degenerative progression and facilitated NP regeneration. Our findings suggest that the KGN + IL-10@GelMA hydrogel holds significant potential as a nonsurgical treatment option for IVDD, offering a promising approach to mitigate the progression of IVDD and enhance disc repair.
Collapse
Affiliation(s)
- Shaofeng Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Jinhui Shi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Yusen Qiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Yun Teng
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Xianggu Zhong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Tianyi Wu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Chao Liu
- Department of Orthopaedic Surgery, Clinical Medicine Institute of Soochow University & Suzhou BenQ Medical Center, Soochow University, Suzhou 215010, Jiangsu, China
| | - Jun Ge
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
- Department of Orthopaedic Surgery, Clinical Medicine Institute of Soochow University & Suzhou BenQ Medical Center, Soochow University, Suzhou 215010, Jiangsu, China
| |
Collapse
|
29
|
Fong H, Mendel M, Jascur J, Najmi L, Kim K, Lew G, Garimalla S, Schock S, Hu J, Villegas AG, Conway A, Fontenot JD, Zompi S. A serum- and feeder-free system to generate CD4 and regulatory T cells from human iPSCs. Stem Cells 2025; 43:sxaf001. [PMID: 39878584 DOI: 10.1093/stmcls/sxaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/02/2025] [Indexed: 01/31/2025]
Abstract
iPSCs can serve as a renewable source of a consistent edited cell product, overcoming limitations of primary cells. While feeder-free generation of clinical grade iPSC-derived CD8 T cells has been achieved, differentiation of iPSC-derived CD4sp and regulatory T cells requires mouse stromal cells in an artificial thymic organoid. Here we report a serum- and feeder-free differentiation process suitable for large-scale production. Using an optimized concentration of PMA/Ionomycin, we generated iPSC-CD4sp T cells at high efficiency and converted them to Tregs using TGFβ and ATRA. Using genetic engineering, we demonstrated high, non-viral, targeted integration of an HLA-A2 CAR in iPSCs. iPSC-Tregs ± HLA-A2-targeted CAR phenotypically, transcriptionally and functionally resemble primary Tregs and suppress T-cell proliferation in vitro. Our work is the first to demonstrate an iPSC-based platform amenable to manufacturing CD4 T cells to complement iPSC-CD8 oncology products and functional iPSC-Tregs to deliver Treg cell therapies at scale.
Collapse
Affiliation(s)
- Helen Fong
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Proximity Therapeutics, San Francisco, CA 94107, United States
| | | | - John Jascur
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Proximity Therapeutics, San Francisco, CA 94107, United States
| | - Laeya Najmi
- Sangamo Therapeutics, Richmond, CA 94804, United States
- BioMarin, Novato, CA 94949, United States
| | - Ken Kim
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Garrett Lew
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Swetha Garimalla
- Sangamo Therapeutics, Richmond, CA 94804, United States
- OmniAb, Emeryville, CA 94608, United States
| | | | - Jing Hu
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Andres Gordillo Villegas
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Kodiak Sciences, Palo Alto, CA 94304, United States
| | - Anthony Conway
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Replay, San Diego, CA 92121, United States
| | - Jason D Fontenot
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Stylus Medicine, Cambridge, MA 02139, United States
| | - Simona Zompi
- Sangamo Therapeutics, Richmond, CA 94804, United States
- CARGO Therapeutics, San Carlos, CA 94070, United States
| |
Collapse
|
30
|
DiDonato M, Simpson CT, Vo T, Knuth M, Geierstanger B, Jamontt J, Jones DH, Fathman JW, DeLarosa D, Junt T, Picard D, Sommer U, Bagger M, Peters E, Meeusen S, Spraggon G. A novel interleukin-10 antibody graft to treat inflammatory bowel disease. Structure 2025; 33:475-488.e7. [PMID: 39798572 DOI: 10.1016/j.str.2024.12.010] [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: 07/19/2024] [Revised: 11/19/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025]
Abstract
Inflammatory bowel disease (IBD) consists of chronic conditions that severely impact a patient's health and quality of life. Interleukin-10 (IL-10), a potent anti-inflammatory cytokine has strong genetic links to IBD susceptibility and has shown strong efficacy in IBD rodent models, suggesting it has great therapeutic potential. However, when tested in clinical trials for IBD, recombinant human IL-10 (rhIL-10) showed weak and inconsistent efficacy due to its short half-life and pro-inflammatory properties that counteract the anti-inflammatory efficacy. Here we present an engineered, IL-10, antibody-graft therapeutic (GFT-IL10M) designed to rectify these issues. GFT-IL10M combines the half-life extension properties of a monoclonal IgG antibody with altered IL-10 cell-type selective signaling, retaining desirable signaling on monocytes while reducing unwanted signaling on T, natural killer (NK), and B cells. Our structural and biochemical results indicate that the altered IL-10 topology in GFT-IL10M leads to a predominantly anti-inflammatory profile, potentially altering cell-type specific signaling patterns and extending half-life.
Collapse
Affiliation(s)
- Michael DiDonato
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Carolina Turk Simpson
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Todd Vo
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Mark Knuth
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Bernhard Geierstanger
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | | | - David H Jones
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - John W Fathman
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Donnie DeLarosa
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Tobias Junt
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Damien Picard
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Ulrike Sommer
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Morten Bagger
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Eric Peters
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Shelly Meeusen
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Glen Spraggon
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA.
| |
Collapse
|
31
|
Chen X, Zhang R, Xie H, Li S, Guo J, Wang Y. Association of the IL-10 and IL-18 polymorphisms with nasopharyngeal carcinoma risk. Front Oncol 2025; 15:1543182. [PMID: 40115022 PMCID: PMC11922698 DOI: 10.3389/fonc.2025.1543182] [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: 12/11/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Objective To evaluate the possible association of the cytokine polymorphisms with the risk of nasopharyngeal carcinoma (NPC). Methods We performed a comprehensive search of electronic databases from PubMed, Web of Science, Embase, and CNKI. Articles related to the cytokine polymorphisms in patients with NPC and healthy controls from inception to 1 April 2024 were included. The results were analysed independently by two reviewers using RevMan 5.4 software. Summary odds ratio (OR) and 95% confidence interval (CI) were used to evaluate cancer risk. Results Our results showed that IL-10 1082A>G showed a significant difference only in the Dominant model, but in the Asian population, a significant difference was shown in all models. IL-18 607C>A polymorphism showed significant differences in the Allele model, Heterozygote model, and Homozygote model. In addition, the IL-18 137G>C polymorphism showed significant differences in all models. No statistically significant association was found between IL-8 251A>T, IL-10 819T>C polymorphism, and the risk of NPC. Conclusion Our meta-analysis results suggest that the IL-18 607C>A and IL-18 137G>C polymorphism are associated with the increased risk of NPC, and IL-10-1082 A/G polymorphism is associated with the increased risk of NPC in Asian populations.
Collapse
Affiliation(s)
- Xueru Chen
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Ruibin Zhang
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| | - Hui Xie
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| | - Sha Li
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Jincai Guo
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yan Wang
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
32
|
Izzy S, Yahya T, Albastaki O, Abou-El-Hassan H, Aronchik M, Cao T, De Oliveira MG, Lu KJ, Moreira TG, da Silva P, Boucher ML, Beauchamp LC, S LeServe D, Brandao WN, Carolina Durão A, Lanser T, Montini F, Lee JH, Bernstock JD, Kaul M, Pasquarelli-do-Nascimento G, Chopra K, Krishnan R, Mannix R, Rezende RM, Quintana FJ, Butovsky O, Weiner HL. Nasal anti-CD3 monoclonal antibody ameliorates traumatic brain injury, enhances microglial phagocytosis and reduces neuroinflammation via IL-10-dependent T reg-microglia crosstalk. Nat Neurosci 2025; 28:499-516. [PMID: 40016353 PMCID: PMC11893472 DOI: 10.1038/s41593-025-01877-7] [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: 10/17/2022] [Accepted: 12/20/2024] [Indexed: 03/01/2025]
Abstract
Neuroinflammation plays a crucial role in traumatic brain injury (TBI), contributing to both damage and recovery, yet no effective therapy exists to mitigate central nervous system (CNS) injury and promote recovery after TBI. In the present study, we found that nasal administration of an anti-CD3 monoclonal antibody ameliorated CNS damage and behavioral deficits in a mouse model of contusional TBI. Nasal anti-CD3 induced a population of interleukin (IL)-10-producing regulatory T cells (Treg cells) that migrated to the brain and closely contacted microglia. Treg cells directly reduced chronic microglia inflammation and regulated their phagocytic function in an IL-10-dependent manner. Blocking the IL-10 receptor globally or specifically on microglia in vivo abrogated the beneficial effects of nasal anti-CD3. However, the adoptive transfer of IL-10-producing Treg cells to TBI-injured mice restored these beneficial effects by enhancing microglial phagocytic capacity and reducing microglia-induced neuroinflammation. These findings suggest that nasal anti-CD3 represents a promising new therapeutic approach for treating TBI and potentially other forms of acute brain injury.
Collapse
Affiliation(s)
- Saef Izzy
- Immunology of Brain Injury Program, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Stroke, Cerebrovascular, and Critical Care Neurology, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Taha Yahya
- Immunology of Brain Injury Program, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Stroke, Cerebrovascular, and Critical Care Neurology, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar Albastaki
- Immunology of Brain Injury Program, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Stroke, Cerebrovascular, and Critical Care Neurology, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Aronchik
- Immunology of Brain Injury Program, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tian Cao
- Immunology of Brain Injury Program, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marilia Garcia De Oliveira
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kuan-Jung Lu
- Immunology of Brain Injury Program, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thais G Moreira
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick da Silva
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masen L Boucher
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Leah C Beauchamp
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Danielle S LeServe
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wesley Nogueira Brandao
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana Carolina Durão
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Toby Lanser
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Federico Montini
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joon-Hyuk Lee
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Megha Kaul
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Kusha Chopra
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajesh Krishnan
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebekah Mannix
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Pérez-Castillo IM, Rueda R, Bouzamondo H, Aparicio-Pascual D, Valiño-Marques A, López-Chicharro J, Segura-Ortiz F. Does Lifelong Exercise Counteract Low-Grade Inflammation Associated with Aging? A Systematic Review and Meta-Analysis. Sports Med 2025; 55:675-696. [PMID: 39792347 PMCID: PMC11985631 DOI: 10.1007/s40279-024-02152-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Aging is associated with sustained low-grade inflammation, which has been linked to age-related diseases and mortality. Long-term exercise programs have been shown to be effective to for attenuating this process; however, subsequent detraining might negate some of these benefits. Master athletes, as a model of lifelong consistent exercise practice, have been suggested to present similar inflammatory profiles to untrained young adults. Nonetheless, it is unclear whether maintaining training habits throughout life can completely counteract low-grade inflammation associated with aging. OBJECTIVES We aimed to systematically evaluate comparisons of baseline inflammatory profiles in Master athletes, untrained middle-aged and older adults, and untrained young individuals to elucidate whether lifelong exercise can counteract low-grade inflammation associated with aging. METHODS A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement, and a protocol was prospectively registered in PROSPERO (CRD42024521339). Studies reporting baseline systemic levels of proinflammatory and anti-inflammatory markers in Master athletes and untrained controls were eligible for inclusion. A total of six databases (PubMed [MEDLINE], Embase, Cochrane Central Register of Controlled Trials [CENTRAL], Scopus, SPORTDiscus, and Web of Science [WoS]) were searched in September 2024, and studies were independently screened by two reviewers. Risk of bias was assessed using an adapted version of the Joanna Briggs Institute Critical Appraisal tool for cross-sectional trials, and random-effect meta-analyses of standardized mean differences (SMDs) of inflammatory markers were conducted to evaluate comparisons between Master athletes and age-matched untrained middle-aged and older adults as well as Master athletes and young untrained subjects. Subgroup analyses were performed based on exercise intensity and type, and participants' sex. RESULTS A total of 17 studies (n = 649 participants) were included both in qualitative and quantitative synthesis. Lifelong exercise appears to attenuate increases in baseline C-reactive protein, and to elevate anti-inflammatory interleukin (IL)-10 levels compared with untrained middle-aged and older adults (C-reactive protein: SMD - 0.71, 95% confidence interval - 0.97, - 0.45, I2 0%, p = 0.78; IL-10: SMD 1.44, 95% confidence interval 0.55, 2.32, I2 87%, p < 0.00001). Statistical significance was maintained in C-reactive protein and IL-10 sub-analyses. No difference in tumor necrosis factor-α levels was observed between Master athletes and untrained middle-aged and older adults (SMD 0.40, 95% confidence interval - 0.15, 0.96, I2 72%, p = 0.0008). A trend towards decreased IL-6 levels in Master athletes was shown in pooled analyses comparing untrained middle-aged and older adults, and rendered statistically significant in sub-analyses. However, comparisons with young untrained adults indicated that Master athletes still present with elevated levels of tumor necrosis factor-α and IL-6, along with decreased IL-10. CONCLUSIONS Master athletes might exhibit a more anti-inflammatory profile denoted by decreased baseline circulating levels of C-reactive protein and, potentially, IL-6, along with increased IL-10 compared with healthy age-matched untrained peers. However, lifelong exercise might still be insufficient to completely counteract age-related changes in tumor necrosis factor-α, IL-6, and IL-10, as shown in comparisons with untrained young adults.
Collapse
Affiliation(s)
- Iñigo M Pérez-Castillo
- Research and Development, Abbott Nutrition, 68 Camino de Purchil, 18004, Granada, Spain.
| | - Ricardo Rueda
- Research and Development, Abbott Nutrition, 68 Camino de Purchil, 18004, Granada, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Huang H, Chen B, Feng C, Chen W, Wu D. Exploring the mediating role of immune cells in the pathogenesis of IgA nephropathy through the inflammatory axis of gut microbiota from a genomic perspective. Mamm Genome 2025; 36:306-316. [PMID: 39505739 PMCID: PMC11880094 DOI: 10.1007/s00335-024-10081-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024]
Abstract
IgA nephropathy (IgAN) is a chronic glomerular disease characterized by the deposition of IgA antibodies in the kidney's mesangium. Its pathogenesis involves genetic, immune, and environmental factors, particularly within the mucosal immune system and gut microbiota. Immune cells play a central role in mediating these processes, which this study investigates using Mendelian Randomization (MR) to explore causal relationships among gut microbiota, inflammatory markers, blood cells, and immune cells in IgAN pathogenesis. We conducted a two-sample MR analysis using Genome-Wide Association Study (GWAS) summary data to assess the causal effects of gut microbiota, inflammatory markers, and blood cell traits on IgAN. Data sources included the FinnGen dataset for IgAN and relevant GWAS datasets for immune traits, blood cells, and inflammatory markers. Inverse variance weighting (IVW) was the primary MR method, supported by sensitivity analyses. We particularly examined the mediation effect of immune cells on these exposures' influence on IgAN. Significant associations were found between several factors and IgAN. Gut microbiota traits, such as Firmicutes E and Sporomusales, increased IgAN risk, while Citrobacter A and Herbinix reduced it. Inflammatory markers, including Interleukin-10 and Fibroblast Growth Factor 23, promoted IgAN onset. Blood cell traits like red blood cell perturbation response increased risk, while monocyte perturbation response was protective. Immune traits played a key mediating role, with Transitional %B cells reducing IgAN risk and CD28- CD25 + + CD8br %T cells increasing it. This study highlights the pivotal mediating role of immune cells in the interactions between gut microbiota, inflammatory markers, and IgAN risk. These findings identify potential biomarkers and therapeutic targets, providing new insights into the immune mechanisms underlying IgAN and opportunities for intervention.
Collapse
Affiliation(s)
- Haoxiang Huang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bohong Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Cong Feng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Dapeng Wu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
35
|
Cao JF, Yang GJ, Zhang YA, Chen J. Contribution of interleukins in the regulation of teleost fish immunity: A review from the perspective of regulating macrophages. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110173. [PMID: 39909123 DOI: 10.1016/j.fsi.2025.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Interleukins (ILs) are potent secreted regulators of a wide range of cell types and cellular activities, particularly in the immune system. They are able to participate in intercellular communication in homeostasis and disease, thereby exerting immune functions. Macrophages serve as the innate immune cells of vertebrates and play a pivotal role in defending against and eliminating external pathogens. In mammals, the immune response mounted by macrophages is intricately linked to ILs. Given the fact that teleost fish have evolved an innate immune system that closely resembles those of mammals, particularly in terms of the functionality of macrophages, raises the intriguing possibility that the regulatory function of ILs in macrophage-mediated immunity might be evolutionarily conserved across both mammal and teleost fish lineages. Consequently, from the perspective of interleukin regulation of macrophages, this review outlines the relationship between ILs and macrophages in teleost fish, and elucidates the regulatory role of ILs of immune cell function in teleost fish, thereby contributing to our understanding of the key role of these cytokines in the prevention and control of aquaculture diseases.
Collapse
Affiliation(s)
- Jia-Feng Cao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Guan-Jun Yang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China.
| |
Collapse
|
36
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Ru Su Y, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3+ T-cell: CD14+ monocyte complexes are dynamic and increased with HIV and glucose intolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae054. [PMID: 40073149 PMCID: PMC11952877 DOI: 10.1093/jimmun/vkae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/07/2024] [Indexed: 03/14/2025]
Abstract
Persistent systemic inflammation is associated with an elevated risk of cardiometabolic diseases. However, the characteristics of the innate and adaptive immune systems in individuals who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled human immunodeficiency virus (HIV) as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+ T cell: CD14+ monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+ T-cell: CD14+ monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis. CD3+ T-cell: CD14+ monocyte complexes may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cell-monocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Samuel S Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Christian M Warren
- Veterans Affairs Flow Cytometry Core, Veterans AffairsTennessee Valley Healthcare System, Nashville, TN, United States
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, United States
| | - Leslie M Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, United States
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Erin M Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sara Gianella
- Division of Infectious Diseases, University of California, San Diego, CA, United States
| | - Elizabeth J Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - David G Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Spyros A Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Simon A Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, United States
| | - John R Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Veterans Health Administration, Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
37
|
Abd El-Gawad EA, Zahran E, Youssuf H, Shehab A, Matter AF. Defatted black soldier fly (Hermetia illucens) diets improved hemato-immunological responses, biochemical parameters, and antioxidant activities in Streptococcus iniae-infected Nile tilapia (Oreochromis niloticus). BMC Vet Res 2025; 21:104. [PMID: 40001064 PMCID: PMC11852831 DOI: 10.1186/s12917-025-04484-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/08/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Challenges of limited supply and increasing prices of fishmeal have driven the aquaculture nutritionists to seek alternative sustainable protein rich ingredients to keep manufacturing aquafeeds in a maintainable and cost-effective way. Black soldier fly, Hermetia illucenslarvae meal represent great potential as a sustainable alternative to fishmeal in aquafeeds. METHODS Three replacement diets for fishmeal were prepared at different levels of defatted black soldier fly (Hermetia illucens) meal (DBSFM): Diet 1 (0 g DBSFM /kg diet, control), 33% (DBSFM-33%, 66 g DBSFM /kg diet), and 100% (DBSFM-100%, 200 g DBSFM /kg diet) to investigate their effects on biochemical parameters, immuno-hematological responses, antioxidant activities, and inflammatory gene expression in Nile tilapia, Oreochromis niloticus, a total of 270 (40.0 ± 0.50 g) before and after challenge with Streptococcus iniae (S. iniae). The feeding trial lasted six weeks (pre-challenge) and two weeks (post-challenge). RESULTS The results showed a significant improvement in white blood cell count (P < 0.01), lymphocyte count (P < 0.01), serum lysozyme activity (P < 0.001), and phagocytic activity (P < 0.001), mostly in the DBSFM-100% group following the pre-challenge phase compared to the control group. Post-challenge phase exhibited significant increases in blood indices in the DBSFM-treated groups compared to the control group. Following pre- and post-challenge periods, both DBSFM-supplemented groups experienced significant increases (P < 0.01, P < 0.001), in serum total protein levels. Albumin and globulin levels also experienced similar increases (P < 0.05, P < 0.01), but only post-challenge. Total antioxidant capacity exhibited a significant increase in both DBSFM-supplemented groups following the post-challenge, as did superoxide dismutase, catalase, and glutathione peroxidase in the liver and spleen. Conversely, levels of glucose, cortisol, and malondialdehyde followed the opposite trend. DBSFM-100% inclusion revealed significant (P < 0.05) up-regulation of interleukin 1β (IL-1β) in the pre-challenge phase compared to control, but no significance (P > 0.05) was seen for other genes. Anti-inflammatory-related genes transforming growth factor-β and interleukin-10 mRNA expression levels were up-regulated in DBSFM-supplemented groups compared to the control post-challenge, but the opposite was seen for IL-1β and tumor necrosis factor- α. CONCLUSION These findings suggest that Nile tilapia challenged with S. iniae may experience significant enhancements in hemato-immunological parameters, antioxidant capability, and anti-inflammatory gene expression when fish meal is replaced with DBSFM up to 100%.
Collapse
Affiliation(s)
- Eman A Abd El-Gawad
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Benha University, Toukh, Kalubia, Egypt.
| | - Eman Zahran
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Hadeer Youssuf
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Benha University, Toukh, Kalubia, Egypt
| | - Ahmed Shehab
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Benha University, Toukh, Kalubia, Egypt
| | - Aya F Matter
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Benha University, Toukh, Kalubia, Egypt
| |
Collapse
|
38
|
Fey RM, Billo A, Clister T, Doan KL, Berry EG, Tibbitts DC, Kulkarni RP. Personalization of Cancer Treatment: Exploring the Role of Chronotherapy in Immune Checkpoint Inhibitor Efficacy. Cancers (Basel) 2025; 17:732. [PMID: 40075580 PMCID: PMC11899640 DOI: 10.3390/cancers17050732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/15/2025] [Indexed: 03/14/2025] Open
Abstract
In the era of precision medicine, mounting evidence suggests that the time of therapy administration, or chronotherapy, has a great impact on treatment outcomes. Chronotherapy involves planning treatment timing by considering circadian rhythms, which are 24 h oscillations in behavior and physiology driven by synchronized molecular clocks throughout the body. The value of chronotherapy in cancer treatment is currently under investigation, notably in the effects of treatment timing on efficacy and side effects. Immune checkpoint inhibitor (ICI) therapy is a promising cancer treatment. However, many patients still experience disease progression or need to stop the therapy early due to side effects. There is accumulating evidence that the time of day at which ICI therapy is administered can have a substantial effect on ICI efficacy. Thus, it is important to investigate the intersections of circadian rhythms, chronotherapy, and ICI efficacy. In this review, we provide a brief overview of circadian rhythms in the context of immunity and cancer. Additionally, we outline current applications of chronotherapy for cancer treatment. We synthesize the 29 studies conducted to date that examine the impact of time-of-day administration on the efficacy of ICI therapy, its associated side effects, and sex differences in both efficacy and side effects. We also discuss potential mechanisms underlying these observed results. Finally, we highlight the challenges in this area and future directions for research, including the potential for a chronotherapeutic personalized medicine approach that tailors the time of ICI administration to individual patients' circadian rhythms.
Collapse
Affiliation(s)
- Rosalyn M. Fey
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Avery Billo
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Terri Clister
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Khanh L. Doan
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Elizabeth G. Berry
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
| | - Deanne C. Tibbitts
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA; (R.M.F.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Portland, OR 97239, USA
- Operative Care Division, U.S. Department of Veterans Affairs Portland Health Care System, Portland, OR 97239, USA
| |
Collapse
|
39
|
Su H, Wu Y, Chen B, Cui Y. STANCE: a unified statistical model to detect cell-type-specific spatially variable genes in spatial transcriptomics. Nat Commun 2025; 16:1793. [PMID: 39979358 PMCID: PMC11842841 DOI: 10.1038/s41467-025-57117-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
One of the major challenges in spatial transcriptomics is to detect spatially variable genes (SVGs), whose expression patterns are non-random across tissue locations. Many SVGs correlate with cell type compositions, introducing the concept of cell type-specific SVGs (ctSVGs). Existing ctSVG detection methods treat cell type-specific spatial effects as fixed effects, leading to tissue spatial rotation-dependent results. Moreover, SVGs may exhibit random spatial patterns within cell types, meaning an SVG is not always a ctSVG, and vice versa, further complicating detection. We propose STANCE, a unified statistical model for both SVGs and ctSVGs detection under a linear mixed-effect model framework that integrates gene expression, spatial location, and cell type composition information. STANCE ensures tissue rotation-invariant results, with a two-stage approach: initial SVG/ctSVG detection followed by ctSVG-specific testing. We demonstrate its performance through extensive simulations and analyses of public datasets. Downstream analyses reveal STANCE's potential in spatial transcriptomics analysis.
Collapse
Affiliation(s)
- Haohao Su
- Department of Statistics and Probability, Michigan State University, East Lansing, 48824, MI, USA
| | - Yuesong Wu
- Department of Statistics and Probability, Michigan State University, East Lansing, 48824, MI, USA
| | - Bin Chen
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, 48824, MI, USA
- Department of Computer Science and Engineering, Michigan State University, East Lansing, 48824, MI, USA
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, 49503, MI, USA
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, 48824, MI, USA.
| |
Collapse
|
40
|
Choi W, Moniruzzaman M, Lee S, Bae J, Bai SC, Min T, Lee S. Evaluation of three fish-derived probiotic bacteria replacing antibiotics on growth, immunity, gut morphology and disease resistance in juvenile olive flounder Paralichthys olivaceus fed reduced fish meal diets. Front Nutr 2025; 12:1519140. [PMID: 40018274 PMCID: PMC11864908 DOI: 10.3389/fnut.2025.1519140] [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: 10/29/2024] [Accepted: 01/29/2025] [Indexed: 03/01/2025] Open
Abstract
A basal diet without feed additives was used as a control (CON) and three diets were formulated by supplementing with Bacillus subtilis WB60 at 1 × 108 CFU/g (Pro-A), B. subtilis SJ10 at 1 × 108 CFU/g (Pro-B), Enterococcus faecium SH30 at 1 × 107 CFU/g (Pro-C), and two other diets supplementing with antibiotics such as amoxicillin (AMO) at 4 g/kg and oxytetracycline (OTC) at 4 g/kg of the basal diet. A total of 450 fish averaging 12.1 ± 0.09 g (mean ± SD) were fed one of the six experimental diet groups in triplicates for 8 weeks. In disease resistance test, 45 fish from each group were intraperitoneally injected with the pathogenic bacteria, Edwardsiella tarda, and mortality was recorded for 15 days. At the end of 8-week feeding trial, weight gain, specific growth rate and feed efficiency of fish fed the Pro-A diet were significantly greater than those of fish fed the CON, OTC and AMO diets (p < 0.05). Furthermore, feeding efficiency and protein efficiency ratio of fish fed the Pro-A diet were significantly greater than those of fish fed the CON, OTC and AMO diets. Serum aspartate aminotransferase levels were significantly greater in fish fed the Pro-B diet than in those fed the Pro-A diet. The lysozyme activity of fish fed the Pro-A, Pro-B and Pro-C diets was significantly greater than that of the CON, OTC and AMO diets. The myeloperoxidase activity of fish fed the Pro-A diet was significantly greater than that of the fish fed the CON and AMO diets. The flounder growth hormone levels of fish fed the Pro-A, Pro-B, Pro-C and AMO diets were significantly greater than that of the fish fed the CON diet. The interleukin 1β gene expression levels in fish fed the Pro-B and Pro-C diets were significantly greater than those in fish fed the CON, OTC and AMO diets. The interleukin 10 gene expression levels in fish fed the Pro-A, Pro-B, Pro-C and OTC diets were significantly greater than those of fish fed the CON and AMO diets. Intestinal histology revealed that the average villi length of fish fed the Pro-A, Pro-B, and Pro-C diets were significantly greater than that of fish fed the CON, OTC and AMO diets. The cumulative survival rates of fish fed the Pro-A, Pro-B and Pro-C diets were significantly greater than those of fish fed the CON diet after the 15th day of the challenge test. Overall, the results demonstrated that the supplementation of fish-derived bacteria, B. subtilis (1 × 108 CFU/g diet) or E. faecium (1 × 107 CFU/g diet) in the diet could be the ideal probiotics to replace antibiotics in olive flounder fed FM reduced diet.
Collapse
Affiliation(s)
- Wonsuk Choi
- Feeds and Foods Nutrition Research Center, Pukyong National University, Busan, Republic of Korea
| | - Mohammad Moniruzzaman
- Department of Animal Biotechnology, Jeju International Animal Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju, Republic of Korea
| | - Seunghan Lee
- Department of Aquaculture and Aquatic Science, Kunsan National University, Gunsan, Republic of Korea
| | - Jinho Bae
- Aquafeed Research Center, National Institute of Fisheries Science, Pohang, Republic of Korea
| | - Sungchul C. Bai
- Feeds and Foods Nutrition Research Center, Pukyong National University, Busan, Republic of Korea
- FAO World Fisheries University Pilot Program, Busan, Republic of Korea
| | - Taesun Min
- Department of Animal Biotechnology, Bio-Resources Computing Research Center, Sustainable Agriculture Research Institute (SARI), Jeju National University, Jeju, Republic of Korea
| | - Seunghyung Lee
- Feeds and Foods Nutrition Research Center, Pukyong National University, Busan, Republic of Korea
- Major of Aquaculture and Applied Life Sciences, Division of Fisheries Life Sciences, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
41
|
Qian W, Chen Y, Li C, Li X, Lv C, Jia Y, Hu S, Zhang M, Wang T, Yan W, Qi M. Neospora caninum Inhibits Lewis Cancer and B16f10 Melanoma Lung Metastasis Development by Activating the Immune Response in Murine Models. Acta Parasitol 2025; 70:52. [PMID: 39918646 DOI: 10.1007/s11686-025-00996-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
Malignant tumors are prevalent with high mortality rates in humans, dogs, and cats. Some microorganisms have been shown to inhibit cancer progression. The objective of this study is to evaluate the inhibitory effects of Neospora caninum, a livestock parasite, on three different tumor models in C57BL/6 mice, including Lewis subcutaneous tumors, Lewis and B16F10 melanoma lung metastasis. The results showed that a sufficient amount of N. caninum tachyzoites can significantly inhibit the development of subcutaneous tumors and lung metastasis (P < 0.001), and induce more than 50% tumor cell death in Lewis subcutaneous tumors. N. caninum treatment can significantly increases the infiltration of macrophages, NK cells, and CD8+ T cells (P < 0.0001) in Lewis subcutaneous tumors detected by immunohistochemistry, and the percentage of these immunocytes in the spleen (P < 0.05) of mice bearing B16F10 melanoma metastasis detected by flow cytometry. And with these changes, the mRNA expression levels of IL-12, IFN-γ, IL-2, IL-10, TNF-α and PD-L1 in tumor microenvironment and IL-12, IFN-γ, IL-2 in spleen were also significantly increased (P < 0.05). Altogether, our results indicate that a sufficient amount N. caninum tachyzoites not only inhibits the growth of Lewis subcutaneous tumors, but inhibits the development of Lewis and B16F10 melanomas lung metastatic in mice by activating potent immune responses. N. caninum and its anti-tumor properties may be an effective anti-tumor tool.
Collapse
Affiliation(s)
- Weifeng Qian
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China.
| | - Yaqi Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Chen Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Xiaojin Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
- China Agricultural University, Beijing, 100193, China
| | - Chaochao Lv
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Yanyan Jia
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Suhui Hu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Min Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Tianqi Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Wenchao Yan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, China
| | - Meng Qi
- College of Animal Science, Tarim University, Alar, Xinjiang, 843300, China.
| |
Collapse
|
42
|
Qin H, Wang J, Bai L, Ding H, Ding H, Zhang F, Han Y. Aerosol inhalation of rhIL-10 improves acute lung injury in mice by affecting pulmonary neutrophil phenotypes through neutrophil-platelet aggregates. Int Immunopharmacol 2025; 147:113948. [PMID: 39778276 DOI: 10.1016/j.intimp.2024.113948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025]
Abstract
This study investigates the therapeutic effects of recombinant human IL-10 (rhIL-10) administered via aerosol inhalation in acute lung injury (ALI), with a particular focus on neutrophils. It explores how rhIL-10, in the presence of platelets, modulates neutrophil polarization to ameliorate acute lung injury. Initially, the ALI model established in mice demonstrated that aerosol inhalation of rhIL-10 significantly mitigated the cytokine storm in the lungs, reduced pulmonary edema, and alleviated histopathological damage to lung tissue. Additionally, rhIL-10 administration was found to decrease neutrophil infiltration and platelet activation in the lungs of mice, inhibiting the formation of platelet-neutrophil aggregates (PNAs) and promoting the differentiation of neutrophils toward an anti-inflammatory phenotype in the presence of platelets. Subsequently, primary neutrophils and platelets were isolated from mouse bone marrow and blood to explore the underlying mechanisms. The results indicated that rhIL-10 promotes the expression of the signal transducer and activator of transcription 3 (STAT3) and the suppressor of cytokine signaling 3 (SOCS3) in neutrophils while inhibiting the activation of the nuclear factor kappa B (NF-κB) and the NF-κB inhibitor (IκB), which in turn enhances CD40 expression. This interaction facilitates the formation of PNAs and influences neutrophil phenotype differentiation. Furthermore, the application of the STAT3 phosphorylation inhibitor Stattic and CD40 antibody in vivo provided further validation of this potential mechanism. In conclusion, these results indicate that aerosol inhalation of rhIL-10 effectively ameliorates ALI. The underlying mechanism may involve the modulation of the neutrophil STAT/SOCS-IκB/NF-κB-CD40 signaling pathway, promoting interactions between neutrophils and platelets that facilitate the differentiation of neutrophils toward an anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Huan Qin
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiangang Wang
- School of Basic Medicine, Qingdao University, Qingdao, China; Kanglitai Biopharmaceutical (Qingdao) Co. Ltd., Qingdao, China
| | - Luyuan Bai
- Xianyang Hospital of Yan'an University, Xianyang, China
| | - Huiqin Ding
- School of Basic Medicine, Qingdao University, Qingdao, China; Kanglitai Biopharmaceutical (Qingdao) Co. Ltd., Qingdao, China
| | | | | | - Yantao Han
- School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
43
|
Allison RL, Mangione CC, Suneja M, Gawrys J, Melvin BM, Belous N, LaCroix M, Harmelink M, Burnett BG, Ebert AD. IL-1ra and CCL5, but not IL-10, are promising targets for treating SMA astrocyte-driven pathology. Mol Ther 2025; 33:734-751. [PMID: 39673131 PMCID: PMC11853362 DOI: 10.1016/j.ymthe.2024.12.016] [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: 08/11/2023] [Revised: 10/30/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a pediatric genetic disorder characterized by the loss of spinal cord motor neurons (MNs). Although the mechanisms underlying MN loss are not clear, current data suggest that glial cells contribute to disease pathology. We have previously found that SMA astrocytes drive microglial activation and MN loss potentially through the upregulation of NF-κB-mediated pro-inflammatory cytokines. In this study, we tested the ability of neutralizing C-C motif chemokine ligand 5 (CCL5) while increasing either interleukin-10 (IL-10) or IL-1 receptor antagonist (IL-1ra) to reduce the pro-inflammatory phenotype of SMA astrocytes. While IL-10 was ineffective, IL-1ra ameliorated SMA astrocyte-driven glial activation and MN loss in induced pluripotent stem cell-derived cultures in vitro. In vivo AAV5 delivered IL-1ra overexpression, and miR-30 small hairpin RNA knockdown of CCL5 made modest but significant improvements in lifespan, weight gain, MN number, and motor function of SMNΔ7 mice. These data identify IL-1ra and CCL5 as possible therapeutic targets for SMA and highlight the importance of glial-targeted therapeutics for neurodegenerative disease.
Collapse
Affiliation(s)
- Reilly L Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cecelia C Mangione
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD 20814, USA
| | - Mya Suneja
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jessica Gawrys
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brendan M Melvin
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD 20814, USA
| | - Natalya Belous
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD 20814, USA
| | - Megan LaCroix
- Department of Neurology (Child Neurology), Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew Harmelink
- Department of Neurology (Child Neurology), Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Barrington G Burnett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD 20814, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
44
|
Kong YH, Huang JY, Ding Y, Chen SH, Li QS, Xiong Y. The effect of BMI on survival outcome of breast cancer patients: a systematic review and meta-analysis. Clin Transl Oncol 2025; 27:403-416. [PMID: 39012453 DOI: 10.1007/s12094-024-03563-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/07/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE The main goal of the present research is to explore the potential link of body mass index (BMI) with different survival metrics in breast cancer patients. Our aim is to offer the latest and most thorough meta-analysis, assessing the strength and reliability of the connection that BMI has with prognostic indicators in this disease. PATIENTS AND METHODS As of January 2024, we conducted a systematic literature search across PubMed, Embase, Web of Science, and the Cochrane Library databases. Our search aimed to identify studies examining BMI as an exposure factor, with breast cancer patients constituting the study population, and utilizing adjusted hazard ratio (HR) as the data type of interest. RESULTS The evidence synthesis incorporated a total of 61 eligible articles involving 201,006 patients. Being underweight posed a risk factor for overall survival (OS) in breast cancer patients compared to normal weight (HR 1.15, 95% CI 0.98-1.35; P = 0.08). Overweight or obesity, in comparison to normal weight, was a risk factor for OS (HR 1.18, 95% CI 1.14-1.23; P < 0.00001), disease-free survival (DFS) (HR 1.11, 95% CI 1.08-1.13; P < 0.00001), relapse-free survival (RFS) (HR 1.14, 95% CI 1.06-1.22; P = 0.03), and breast cancer-specific survival (BCSS) (HR 1.18, 95% CI 1.11-1.26; P < 0.00001), but not for progression-free survival (PFS) (HR 0.91, 95% CI 0.76-1.10; P = 0.33). Notably, in subgroup analyses, overweight patients achieved prolonged PFS (HR 0.80, 95% CI 0.64-0.99; P = 0.04), and compared to the obese population, the overweight cohort exhibited a significant difference in OS (HR 1.11, 95% CI 1.05-1.16; P < 0.00001) and DFS (HR 1.06, 95% CI 1.03-1.10; P = 0.0004), with a considerably stronger association. Furthermore, compared to HER- patients, HER + patients exhibited a greater predictive value for OS (HR 1.23, 95% CI 1.10-1.37; P = 0.0004), RFS (HR 1.30, 95% CI 1.03-1.64; P < 0.00001), and DFS (HR 1.10, 95% CI 1.03-1.17; P = 0.003). CONCLUSIONS The results of our meta-analysis reveal a notable association between BMI and various survival measures in breast cancer prognosis. These findings provide a solid basis for predicting breast cancer outcomes and implementing more effective therapeutic approaches.
Collapse
Affiliation(s)
- Yu-Huan Kong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jing-Yi Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Ye Ding
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Shu-Hua Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Qiu-Shuang Li
- Center of Clinical Evaluation and Analysis, Zhejiang Provincial Hospital of Chinese Medicine), The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
45
|
İlhan İ, Asci H, Ozmen O, Buyukbayram Hİ, Arlıoglu M, Kurtbolat O. The renoprotective effects of cannabidiol on lipopolysaccharide-induced systemic inflammation model of rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1841-1851. [PMID: 39180672 DOI: 10.1007/s00210-024-03391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
Sepsis-induced renal damage poses a significant threat, necessitating effective therapeutic strategies. Cannabidiol (CBD) has beneficial effects on tissues and their functions by exhibiting antioxidant and anti-inflammatory effects. This study investigates the potential protective effects of CBD in mitigating lipopolysaccharide (LPS)-induced renal injury in Wistar Albino rats. Thirty-two Wistar Albino rats were categorized into control, LPS (5 mg/kg i.p.), LPS + CBD, and CBD (5 mg/kg i.p.) groups. After the experiment, samples were collected for biochemical, genetic, histopathological, and immunohistochemical analyses. Oxidative stress markers as total oxidant status (TOS) and total antioxidant status (TAS), oxidative stress index (OSI), superoxide dismutase (SOD), glutathione peroxidase (GPx), malondialdehyde (MDA), immune staining as tumor necrosis factor alpha (TNF-α), interleukin-10 (IL-10), caspase-3, gene expressions as nuclear factor erythroid 2-related factor 2 (NRF2), C/EBP homologous protein (CHOP), caspase-9, glucose-regulating protein 78 (GRP78), B-cell leukemia/lymphoma 2 (Bcl2), and tissue histology have been examined. The LPS-exposed group exhibited significant renal abnormalities, mitigated by CBD intervention in the LPS + CBD group. CBD reduced immunoexpression scores for TNF-α, caspase-3, and IL-10. Biochemically, CBD induced a positive shift in the oxidative balance, increasing TAS, SOD, and GPx, while decreasing TOS, OSI, and MDA levels. Genetic analyses highlighted CBD's regulatory impact on NRF2, CHOP, caspase-9, GRP78, and Bcl2, providing molecular insights into its protective role against LPS-induced renal damage. This study underscores CBD as a promising protective agent against sepsis-induced renal damage. Our findings could provide valuable insights into potential therapeutic avenues for addressing renal complications in sepsis.
Collapse
Affiliation(s)
- İlter İlhan
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, 32200, Turkey.
| | - Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Faculty of Veterinary, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Halil İbrahim Buyukbayram
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, 32200, Turkey
| | - Melih Arlıoglu
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Okan Kurtbolat
- Department of Pharmacology, Institute of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
46
|
Lee J, Goo D, Sharma MK, Ko H, Shi H, Paneru D, Choppa VSR, Liu G, Kim WK. Effects of graded yeast cell wall supplementation on growth performance, immunity and intestinal development of broiler chickens raised in floor pens for 42 days. Poult Sci 2025; 104:104695. [PMID: 39721260 PMCID: PMC11732452 DOI: 10.1016/j.psj.2024.104695] [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] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
This study was conducted to evaluate the effects of dietary supplementation of a novel soluble yeast cell wall (YCW) on growth performance, gut health, intestinal morphology, and immune response in broiler chickens for 42 days. A total of 480 one-day-old Cobb 500 male broilers were randomly assigned to four treatments with six replicates and each replicate of twenty broiler chickens: a control group (CON) without feed additive supplementation, and three groups supplemented with YCW at 0.025 % (YCW1), 0.050 % (YCW2), and 0.100 % (YCW3). Results showed that 0.025 % and 0.100 % YCW supplementation significantly increased (P < 0.05) final body weight (BW) and overall body weight gain (BWG) while reducing overall feed conversion ratio (FCR) compared to the CON group. The YCW supplementation also improved (P < 0.05) the balance of gut microbiota by increasing beneficial bacteria (Lactobacillus) and decreasing Salmonella, a potential foodborne pathogen in humans in the ceca. Although intestinal morphology was not significantly affected, YCW supplementation numerically increased the villus height: crypt depth ratio (VH:CD) compared to the CON group. Furthermore, YCW reduced the mRNA expression of pro-inflammatory cytokines (IL-1β and INF-γ) and tight junction protein claudin-1 (CLDN-1) (P < 0.05), suggesting balanced immune response and improved intestinal barrier function. In conclusion, the supplementation of soluble YCW in broiler diets positively influenced growth performance, gut microbiota composition, and immune response, demonstrating its potential as a viable alternative to antibiotics for improving broilers' health.
Collapse
Affiliation(s)
- Jihwan Lee
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | - Doyun Goo
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | - Milan Kumar Sharma
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | - Hanseo Ko
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | - Hanyi Shi
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | - Deependra Paneru
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | | | - Guanchen Liu
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA, 30602 USA.
| |
Collapse
|
47
|
Chen C, Zhang D, Ye M, You Y, Song Y, Chen X. Effects of various exercise types on inflammatory response in individuals with overweight and obesity: a systematic review and network meta-analysis of randomized controlled trials. Int J Obes (Lond) 2025; 49:214-225. [PMID: 39420086 DOI: 10.1038/s41366-024-01649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/25/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE To explore effective exercise types for reducing chronic inflammation in individuals with overweight and obesity (IOO) while accounting for confounders. METHODS A systematic search for RCTs in English between January 2000 and August 2023 was conducted to evaluating exercise effects on inflammatory biomarkers in IOO. A network meta-analysis conducted. RESULTS A total of 123 RCTs were analyzed. Different exercise type yielded distinct effects on various inflammatory biomarkers. Specifically, aerobic exercise combined with resistance training (COM) and aerobic exercise (AE) were the most effective for improving leptin levels. AE exhibited the greatest effectiveness in reducing CRP and increasing adiponectin. High-intensity interval training (HIIT) was identified as the most effective exercise modality for ameliorating IL-6, TNF-α, and IL-10. Resistance training (RT) had the least effect compared to other exercise types. Meta regression and subgroup analyses revealed that high-intensity AE demonstrated a greater effect size compared to moderate-intensity AE. The impact of AE on IL-10 was positively associated with both the training period and the age of participants. Positive correlations were observed between reductions in body fat and the effect sizes of CRP, TNF-α, and IL-10. Gender influenced AE effects on IL-6 and TNF-α, with females responding better. CONCLUSION This study highlights the potential of exercise in alleviating the inflammatory status in IOO, with different exercise types showing various effects on specific inflammatory biomarkers. The intensity and duration of exercise had a dose-response relationship with intervention effectiveness. Changes in body composition correlated with the effectiveness of the intervention. COM, AE, and HIIT are recommended exercise approaches.
Collapse
Affiliation(s)
- Chaofan Chen
- Department of Physical Education, Tsinghua University, 100084, Beijing, China
| | - Dong Zhang
- Institute of Sports Artificial Intelligence, Capital University of Physical Education and Sports, 100084, Beijing, China
| | - Mingyi Ye
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yanwei You
- Department of Physical Education, Tsinghua University, 100084, Beijing, China
| | - Yiling Song
- Department of Physical Education, Tsinghua University, 100084, Beijing, China
| | - Xiaoke Chen
- Department of Physical Education, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
48
|
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
|
49
|
Zhang L, Liang D, Tian Y, Liang J, Li X, Liu C, Liang J, Luo TR, Li X. Classical Swine Fever Virus Envelope Glycoproteins E rns, E1, and E2 Activate IL-10-STAT1-MX1/OAS1 Antiviral Pathway via Replacing Classical IFNα/β. Biomolecules 2025; 15:200. [PMID: 40001503 PMCID: PMC11853677 DOI: 10.3390/biom15020200] [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: 12/31/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Classical swine fever (CSF) is an acute and often fatal disease caused by CSF virus (CSFV) infection. In the present study, we investigated the transcriptional profiles of peripheral blood mononuclear cells (PBMCs) in pigs infected with CSFV. The results revealed that CSFV inhibits IFNα/β production, but up-regulates the expression of signal transducer and activator of transcription 1 (STAT1); this result was verified in vitro. Interestingly, STAT1 is typically a downstream target of IFNα/β, raising the question of how CSFV can inhibit IFNα/β expression, yet up-regulate STAT1 expression. To explore this further, we observed that UV-treated CSFV induced STAT1 expression. Our results demonstrated that CSFV Erns, E1, and E2 could up-regulate STAT1 expression within the host cell cytoplasm and facilitate its translocation into the nucleus. The Erns, E1, and E2 proteins also separately induced the up-regulation of interleukin (IL)-10; IL-10 acts as the communicator connecting Erns, E1, and E2 proteins to STAT1, leading to the subsequent up-regulation, phosphorylation, and nuclear translocation of STAT1. Silencing of IL-10 down-regulated STAT1 expression. Finally, MX1 and OAS1 were identified as downstream targets of the IL-10-STAT1 pathway. In summary, a novel IL-10-STAT1 pathway independent of IFNα/β induced by CSFV Erns, E1, and E2 was identified in this study.
Collapse
Affiliation(s)
- Liyuan Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Dongli Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Yu Tian
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Jiaxin Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Xiaoquan Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Cheng Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
| | - Jingjing Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
| | - Ting Rong Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
| | - Xiaoning Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China; (L.Z.); (D.L.); (Y.T.); (J.L.); (X.L.); (C.L.); (J.L.)
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
| |
Collapse
|
50
|
Hoang Nguyen KH, Le NV, Nguyen PH, Nguyen HHT, Hoang DM, Huynh CD. Human immune system: Exploring diversity across individuals and populations. Heliyon 2025; 11:e41836. [PMID: 39911431 PMCID: PMC11795082 DOI: 10.1016/j.heliyon.2025.e41836] [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: 07/21/2024] [Revised: 12/23/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
The immune response is an intricate system that involves the complex connection of cellular and molecular components, each with distinct functional specialisations. It has a distinct capacity to adjust and mould the immune response in accordance with specific stimuli, influenced by both genetic and environmental factors. The presence of genetic diversity, particularly across different ethnic and racial groups, significantly contributes to the impact of incidence of diseases, disease susceptibility, autoimmune disorders, and cancer risks in specific regions and certain populations. Environmental factors, including geography and socioeconomic status, further modulate the variety of the immune system responses. These, in turn, affect the susceptibility to infectious diseases and development of autoimmune disorders. Despite the complexity of the relationship, there remains a gap in understanding the specificity of immune indices across races, immune reference ranges among populations, highlighting the need for deeper understanding of immune diversity for personalized approaches in diagnostics and therapeutics. This review systematically organizes these findings, with the goal of emphasizing the potential of targeted interventions to address health disparities and advance translational research, enabling a more comprehensive strategy. This approach promises significant advancements in identifying specific immunological conditions, focusing on personalized interventions, through both genetic and environmental factors.
Collapse
Affiliation(s)
| | - Nghi Vinh Le
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
| | | | - Hien Hau Thi Nguyen
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Viet Nam
| | - Duy Mai Hoang
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
| | | |
Collapse
|