1
|
Li S, Liang Q, Qing W, Fang Z, Yuan C, Pan S, Xie H, Li X, Chen M, He Y, Zhou H, Wang Q. Maternal group B Streptococcus decreases infant length and alters the early-life microbiome: a prospective cohort study. Ann Med 2025; 57:2442070. [PMID: 39693119 DOI: 10.1080/07853890.2024.2442070] [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: 03/07/2024] [Revised: 06/18/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Maternal colonization with Group B Streptococcus (GBS) disrupts the vaginal microbiota, potentially affecting infant microbiota assembly and growth. While the gut microbiota's importance in infant growth is recognized, the specific effects of maternal GBS on growth remain unclear. This study aimed to explore the effects of maternal vaginal GBS during pregnancy on early infant growth, microbiome, and metabolomics. METHODS We recruited and classified 453 pregnant women from southern China into GBS or healthy groups based on GBS vaginal colonization. Their infants were categorized as GBS-exposed or GBS-unexposed groups. We comprehensively analyzed infant growth, gut microbiota, and metabolites during early life, along with maternal vaginal microbiota during pregnancy, using 16S rDNA sequencing and targeted metabolomics. RESULTS GBS-exposed infants exhibited lower length-for-age z-scores (LAZ) than GBS-unexposed infants, especially at 2 months. Altered gut microbiota and metabolites in GBS-exposed infants correlated with growth, mediating the impact of maternal GBS on infant LAZ. Changes in the vaginal microbiota of the GBS group during the third trimester correlated with infant LAZ. Additionally, differences in neonatal gut microbiota, metabolites, and vaginal microbiota during pregnancy were identified between infants with overall LAZ<-1 within 8 months after birth and their counterparts, enhancing the discriminatory power of fundamental data for predicting the occurrence of LAZ<-1 during the first 8 months of life. CONCLUSIONS GBS exposure is associated with decreased infant length growth, with altered microbiota and metabolites potentially mediating the effects of maternal GBS on offspring length growth, offering potential targets for predicting and addressing growth impairment.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qijun Liang
- Department of Obstetrics and Gynecology, Boai Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Wei Qing
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhencheng Fang
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunlei Yuan
- Department of Laboratory Medicine, Boai Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Shilei Pan
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hairui Xie
- Department of Paediatrics Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaocong Li
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Guangdong, China
| | - Muxuan Chen
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan He
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Wang
- Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Yang C, Camargo Tavares L, Lee HC, Steele JR, Ribeiro RV, Beale AL, Yiallourou S, Carrington MJ, Kaye DM, Head GA, Schittenhelm RB, Marques FZ. Faecal metaproteomics analysis reveals a high cardiovascular risk profile across healthy individuals and heart failure patients. Gut Microbes 2025; 17:2441356. [PMID: 39709554 DOI: 10.1080/19490976.2024.2441356] [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: 07/30/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
The gut microbiota is a crucial link between diet and cardiovascular disease (CVD). Using fecal metaproteomics, a method that concurrently captures human gut and microbiome proteins, we determined the crosstalk between gut microbiome, diet, gut health, and CVD. Traditional CVD risk factors (age, BMI, sex, blood pressure) explained < 10% of the proteome variance. However, unsupervised human protein-based clustering analysis revealed two distinct CVD risk clusters (low-risk and high-risk) with different blood pressure (by 9 mmHg) and sex-dependent dietary potassium and fiber intake. In the human proteome, the low-risk group had lower angiotensin-converting enzymes, inflammatory proteins associated with neutrophil extracellular trap formation and auto-immune diseases. In the microbial proteome, the low-risk group had higher expression of phosphate acetyltransferase that produces SCFAs, particularly in fiber-fermenting bacteria. This model identified severity across phenotypes in heart failure patients and long-term risk of cardiovascular events in a large population-based cohort. These findings underscore multifactorial gut-to-host mechanisms that may underlie risk factors for CVD.
Collapse
Affiliation(s)
- Chaoran Yang
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Leticia Camargo Tavares
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Han-Chung Lee
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Joel R Steele
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | | | - Anna L Beale
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
| | - Stephanie Yiallourou
- Preclinical Disease and Prevention Unit, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Melinda J Carrington
- Preclinical Disease and Prevention Unit, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - David M Kaye
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- School of Translational Medicine, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Geoffrey A Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Pharmacology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Victorian Heart Institute, Monash University, Clayton, Australia
| |
Collapse
|
3
|
Amaya-Garrido A, Klein J. The role of calprotectin in vascular calcification. Curr Opin Nephrol Hypertens 2025; 34:276-283. [PMID: 40152927 DOI: 10.1097/mnh.0000000000001075] [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: 03/30/2025]
Abstract
PURPOSE OF REVIEW Vascular calcification significantly contributes to cardiovascular morbidity and mortality, particularly in high-risk populations like chronic kidney disease (CKD) patients. Calprotectin, a heterodimeric protein with pro-inflammatory and pro-calcific properties, has emerged as a key molecule in vascular pathology. This review highlights the mechanisms linking calprotectin to vascular calcification, its clinical relevance, and its potential as a therapeutic target. RECENT FINDINGS Vascular calcification is an active, cell-mediated process involving vascular smooth muscle cell (VSMC) dysfunction, inflammation, matrix remodeling, and cellular senescence. Calprotectin is strongly associated with cardiovascular disease and vascular calcification, particularly in CKD. Mechanistic studies reveal that calprotectin promotes calcification through the activation of RAGE and TLR4 signaling pathways, driving inflammatory cascades. Preclinical studies demonstrate that pharmacological inhibition of calprotectin attenuates vascular calcification in animal models, supporting its potential as a therapeutic target. SUMMARY Calprotectin is emerging as a promising biomarker and therapeutic target in vascular calcification, particularly in CKD and aging-related vascular diseases. However, further studies are required to clarify its mechanisms and assess the long-term efficacy and safety of calprotectin-targeted therapies. A deeper understanding of calprotectin's multifaceted role could pave the way for innovative therapeutic strategies targeting both inflammation and mineralization in calcification-related vascular diseases.
Collapse
Affiliation(s)
- Ana Amaya-Garrido
- Department of Nephrology and Transplantation, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease
- Université Toulouse III Paul-Sabatier, Toulouse, France
| |
Collapse
|
4
|
Zheng BX, Yi Y, Wang XW, Li CY, Zhao Y, Tian JZ, Wang LM, Han JY, Pan C, Liu SY, Liu CY, Qin SS, Tang X, Liu MT, Liang AH. Geniposide via enema alleviates colitis by modulating intestinal flora and bile acid metabolites, inhibiting S100A8/S100A9/NF-κB, and promoting TGR5 inhibition of NLRP3 inflammasome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156791. [PMID: 40279965 DOI: 10.1016/j.phymed.2025.156791] [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: 01/14/2025] [Revised: 03/12/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Geniposide (GE) has potential efficacy in treating ulcerative colitis (UC). However, its reactivity can be affected by rapid degradation after oral administration. Furthermore, increasing oral doses may lead to hepatotoxicity. Thus, We used enema administration, characterized by smaller dose and higher localized concentration in the lesion, to improve the above situation. PURPOSE We aimed to confirm that enema administration is a better modality than oral administration for GE against UC and to explore its mechanism. STUDY DESIGN/METHOD We established UC mouse model, monitoring Disease Activity Index (DAI), inflammatory cytokines levels, and histopathology. Macrogenomics and bile acid (BAs) metabolomics analysed the major intestinal flora and BAs. Simultaneouslly, we conducted quantitative proteomics analysis and screened core proteins and pathway. In vitro validation was taken by qPCR, immunofluorescence and immunoblotting experiments. RESULTS GE via enema alleviate UC by inhibiting inflammatory factor production through downregulating S100A8/S100A9/NF-κB pathway. Analysis of the intestinal flora and BAs revealed that the enhanced abundance of Lachnospiraceae, which improves the ratio of primary to secondary BAs, and the reduced abundance of Provocaceae, which increases intestinal permeability and promotes inflammation, favored the restoration of the intestinal barrier. In addition, in vitro experiments confirmed that the key BA metabolites (mainly UDCA, DCA, and LCA) stimulated TGR5 signal to inhibit the assembly of the NLRP3 inflammasome and alleviated inflammation. CONCLUSION We firstly confirmed that GE alleviates UC via the enema route in a better manner than the oral route, through enhancing the intestinal barrier, restoring intestinal flora and BAs homeostasis, and inhibiting inflammatory injury. This study initially revealed that GE can alleviate UC through elevating UDCA, DCA, and LCA levels at the colonic site to activate TGR5 receptor for inhibiting the NLRP3 inflammasome, in addition to downregulating the S100A8/S100A9/-TLR4-NF-κB pathway related inflammatory response directly. The evidences offer a promising strategy and profround meaning for UC treatment.
Collapse
Affiliation(s)
- Bao-Xin Zheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Yi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Xing-Wen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chun-Ying Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yong Zhao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jing-Zhuo Tian
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Lian-Mei Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jia-Yin Han
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chen Pan
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Su-Yan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chen-Yue Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Sha-Sha Qin
- Chongqing University Of Chinese Medicine, Chongging 400060, China.
| | - Xuan Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Mei-Ting Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ai-Hua Liang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
5
|
Zhang Z, Wang HM, Xu ZX, Luan WY, Lin SX, Miao YD. Application of single-cell sequencing in the study of immune cell infiltration in inflammatory bowel disease and colorectal cancer. World J Gastrointest Oncol 2025; 17:107382. [DOI: 10.4251/wjgo.v17.i6.107382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 06/13/2025] Open
Abstract
The rapid advancement of single-cell sequencing (SCS) technology has provided new insights into the relationship between inflammatory bowel disease (IBD) and colorectal cancer (CRC). This technique allows for detailed cellular analysis, enabling researchers to uncover the infiltration patterns of immune cells within the gut microenvironment and their roles in disease progression. This review summarizes significant research findings on the interplay between IBD and CRC, the characteristics of immune cell infiltration, and potential therapeutic targets identified through SCS. The aim is to offer references for future clinical studies and treatment strategies in this field.
Collapse
Affiliation(s)
- Zheng Zhang
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
- Research and Translational Center for Immunological Disorders, Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Hui-Min Wang
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Zhen-Xi Xu
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
- Research and Translational Center for Immunological Disorders, Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Wen-Yu Luan
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
- Research and Translational Center for Immunological Disorders, Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Si-Xiang Lin
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
- Research and Translational Center for Immunological Disorders, Binzhou Medical University, Yantai 264100, Shandong Province, China
| | - Yan-Dong Miao
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The Second Medical College of Binzhou Medical University, Yantai 264100, Shandong Province, China
- Research and Translational Center for Immunological Disorders, Binzhou Medical University, Yantai 264100, Shandong Province, China
- Guangdong Provincial Key Laboratory of Medical Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510000, Guangdong Province, China
- Department of Oncology, Xinhui District People’s Hospital, Jiangmen 529100, Guangdong Province, China
| |
Collapse
|
6
|
Di Tola M, Bontkes HJ, Irure-Ventura J, López-Hoyos M, Bizzaro N. The follow-up of patients with celiac disease. J Transl Autoimmun 2025; 10:100278. [PMID: 39981115 PMCID: PMC11840481 DOI: 10.1016/j.jtauto.2025.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 02/22/2025] Open
Abstract
Celiac disease (CD) is a very common immune-mediated enteropathy resulting from the interaction between dietary gluten and the immune system in genetically predisposed individuals. The immune response leads to intestinal damage, malabsorption and, ultimately, to a broad spectrum of both intestinal and extra-intestinal symptoms. According to current criteria, a proper diagnosis of CD requires an initial phase consisting of clinical case identification and serological screening that, over time, has increased in importance. In most adults and in selected children, the diagnosis is subsequently defined by histological evidence of intestinal damage as a confirmatory test, which usually returns to normal after a suitable period of a gluten-free diet (GFD). The clinical remission and disappearance of circulating antibodies after a GFD further confirm the diagnosis and represent a goal to be achieved to improve the quality of life and reduce the risk of long-term complications. However, although the diagnostic criteria for CD are well defined and described in specific guidelines, the monitoring of CD patients undergoing GFD has been less studied and, consequently, specific guidelines for this phase are still lacking. The aim of this report was to evaluate the classical tools used to monitor the adherence and response to GFD, other non-invasive biomarkers that have been proposed for CD monitoring, and the histological follow-up of CD patients in order to provide a starting point for future discussions on this specific topic.
Collapse
Affiliation(s)
- Marco Di Tola
- UOC Clinical Pathology, San Giovanni - Addolorata Hospital, Rome, Italy
| | - Hetty J. Bontkes
- Department of Laboratory Medicine, Laboratory Specialized Diagnostics and Research, Section Medical Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Juan Irure-Ventura
- Immunology Department, University Hospital Marqués de Valdecilla, Santander, Spain
- Immunopathology Group, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Department, University Hospital Marqués de Valdecilla, Santander, Spain
- Immunopathology Group, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain
| | - Nicola Bizzaro
- Laboratory of Clinical Pathology, Azienda Sanitaria Universitaria Integrata, Udine, Italy
| |
Collapse
|
7
|
Xie J, Xiong S, Yu J, Ma X, Xiang F, Chen Y, Xia B, Li Y, Zhang Z, Liao D, Lin L. Prunella vulgaris polyphenols alleviate liver injury-uveitis comorbidity by regulating acylcarnitine via the S100A9-PP2A-AMPK pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156675. [PMID: 40215815 DOI: 10.1016/j.phymed.2025.156675] [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: 01/22/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Liver injury and uveitis pose severe threats to human health. Owing to the close relationship of physiology and pathology between the liver and the eyes, cases in which both conditions occur simultaneously are not uncommon in clinical settings, significantly complicating treatment. However, no suitable comorbid animal model has been reported, and research on the pathological mechanisms of this comorbidity is lacking. Prunella vulgaris L., a well-known traditional Chinese medicine renowned for its liver-clearing and eye-brightening properties. Prunella vulgaris polyphenols (PVPs) hold promise for improving liver injury and uveitis. However, research exploring their dual therapeutic effects within a single organism remains lacking, leaving the key active components and mechanisms of action largely uninvestigated. PURPOSE This exploratory study aimed to establish a rat model of liver injury combined with uveitis and investigated its pathological mechanisms, evaluating the therapeutic efficacy of PVPs in alleviating liver injury combined with uveitis in rats. Additionally, it explored the mechanism of action and identified key active ingredients of PVPs, offering potential new directions for the development of clinical therapeutic drugs. METHODS A rat model of liver injury with uveitis was established through intraperitoneal d-GalN/LPS injection. Metabolomics and proteomics were applied to investigate pathological mechanisms, followed by validation using acylcarnitine and S100A9 inhibitors. PVPs were administered to evaluate therapeutic effects and explore mechanisms involved in alleviating liver injury and uveitis. Network pharmacology combined with molecular docking identified critical active components in PVPs. Subsequent animal experiments verified the efficacy of the representative component in improving liver injury and uveitis. RESULTS d-GalN/LPS (150 mg/kg : 1 mg/kg) induced significant liver injury and uveitis in rats. Metabolomics analysis pointed to acylcarnitine as a key metabolite, and its inhibition reduced inflammation. Proteomics analysis implicated S100A9 in inflammation and immunity. Then, we intervened with S100A9 inhibitors in the model rats. The results suggested that the pathological mechanism of liver injury and uveitis caused by d-GalN/LPS involved the upregulation of S100A9 expression, an increase in PP2A activity, the inhibition of AMPK phosphorylation, and the downregulation of CPT1A, leading to the accumulation of acylcarnitine and promoting the inflammatory response in the liver and retina. Further, experiments involving PVPs demonstrated dose-dependent improvements in liver injury and uveitis caused by d-GalN/LPS. The underlying mechanism of action involved suppression of S100A9 expression, reduction of PP2A activity, activation of AMPK, upregulation of CPT1A, and subsequent reduction in acylcarnitine accumulation in both the liver and retina. This mechanism effectively alleviated the inflammatory effects induced by d-GalN/LPS. Network pharmacology and molecular docking analyses pinpointed several key active components of PVPs-namely, rosmarinic acid, salviaflaside, esculetin, 2-hydroxycinnamic acid, 3,4-dihydroxybenzaldehyde, and 7,8-dihydroxycoumarin-that play significant roles in mitigating liver injury and uveitis. Follow-up experiments using the representative active component rosmarinic acid in rats confirmed its efficacy in improving symptoms of d-GalN/LPS-induced liver injury and uveitis, further validating the therapeutic potential of these key active components. CONCLUSIONS This study successfully established a rat model of liver injury combined with uveitis and confirmed the efficacy of PVPs in alleviating this condition. Furthermore, it determined that the underlying mechanism involves regulation of the S100A9-PP2A-AMPK pathway, with rosmarinic acid identified as a key active compound. These findings provide a basis for clinical studies on liver-eye comorbidities and offer critical evidence for further research and drug development of PVPs in liver-clearing and eye-brightening.
Collapse
Affiliation(s)
- Jingchen Xie
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Suhui Xiong
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Jiahui Yu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Xinyi Ma
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Feng Xiang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Yang Chen
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Bohou Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Yamei Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Zhimin Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Duanfang Liao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| | - Limei Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
8
|
Li J, Hu R, Liu X, Peng L, Yi J, Zhong X, Huang Q, Sun J, Feng W, Ma W, Zhou X. S100A9/RAGE pathway regulation of mitophagy and the effect of JianPi LiShi YangGan formula in acute-on-chronic liver failure. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119887. [PMID: 40294664 DOI: 10.1016/j.jep.2025.119887] [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: 10/24/2024] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mitophagy regulates cellular homeostasis and liver inflammation; however, it is inhibited in acute-on-chronic liver failure (ACLF), which drives disease progression. The JianPi LiShi YangGan formula (YGF) has the potential to improve inflammatory responses and reduce mortality in patients with ACLF. However, the precise mechanisms underlying these effects remain unknown. AIM OF THE STUDY We investigated the role of S100A9/RAGE signaling in mitophagy and the protective effects of traditional Chinese medicinal compounds on ACLF. MATERIALS AND METHODS An ACLF mouse model was established using carbon tetrachloride, lipopolysaccharide, and d-galactose. Hematoxylin and eosin staining and enzyme-linked immunosorbent assay were employed to evaluate the hepatoprotective effect of YGF in ACLF mice. Mitochondrial damage was assessed using transmission electron microscopy. Protein levels of mitophagy-related indicators were assessed through immunohistochemistry and western blotting, and immunofluorescence staining was performed to observe Lamp2 and COX-IV co-localization. RESULTS The hepatocytes of ACLF mice contained damaged mitochondria, decreased mitophagy-related protein (Pink1, Parkin, and LC3B) expression and activated S100A9/RAGE signaling. Inhibiting S100A9 or RAGE improved liver injury in ACLF mice and enhanced Lamp2-COX-IV co-localization. In alpha mouse liver 12 (AML12) cells overexpressing RAGE, recombinant S100A9 protein inhibited mitophagy induced by 3-chlorocarbonyl benzoyl chloride. YGF reduced mitochondrial damage, increased Pink1, Parkin, and LC3B levels, and enhanced mitophagy while inhibiting S100A9/RAGE activation in the hepatocytes of ACLF mice. CONCLUSIONS This study found that S100A9/RAGE pathway activation impairs mitophagy, and YGF alleviates liver injury by downregulating S100A9 and RAGE signaling, which may be a novel therapeutic strategy for ACLF.
Collapse
Affiliation(s)
- Jing Li
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Rui Hu
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China; Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, China
| | - Xingning Liu
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Lanfen Peng
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Jinyu Yi
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China; Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, China
| | - Xin Zhong
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Qi Huang
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China; Macau University of Science and Technology, Faculty of Chinese Medicine, Taipa, China
| | - Jialing Sun
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Wenxing Feng
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Wenfeng Ma
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China
| | - Xiaozhou Zhou
- Shenzhen Traditional Chinese Medicine Hospital, Department of Liver Disease, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Department of Liver Disease, Shenzhen, 518033, China.
| |
Collapse
|
9
|
Farrera-Soler L, Hu CW, Ricken B, Díaz-Perlas C, Gerhold CB, Heinis C. Structure-Affinity Relationship Analysis and Affinity Maturation of a Calprotectin-Binding Peptide. Chembiochem 2025; 26:e202500071. [PMID: 40237725 DOI: 10.1002/cbic.202500071] [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: 01/24/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/18/2025]
Abstract
Peptide 3 is an 18-amino acid linear peptide binding with sub-micromolar affinity to the inflammation marker calprotectin. Its application in point-of-care diagnostic assays has shown promising results, yet improving its affinity for calprotectin could facilitate the development of sensitive and robust assays. Herein, a detailed structure-activity relationship analysis of Peptide 3 is reported to better understand the importance of each individual amino acid for the binding to calprotectin. Moreover, two different approaches have been followed here, one based on prolonging the peptide with random sequences and phage display selection, and one on screening chemically synthesized peptide variants containing non-canonical amino acids, to increase its binding affinity. Combining several mutations that enhance affinity by small factors yielded Peptide 4 binding human calprotectin with a KD of 39 ± 5 nM measured by surface plasmon resonance spectroscopy, which is a five-fold improvement compared to the previously reported Peptide 3.
Collapse
Affiliation(s)
- Lluc Farrera-Soler
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Che-Wei Hu
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Benjamin Ricken
- BÜHLMANN Laboratories AG, Baselstrasse 55, CH-4124, Schönenbuch, Switzerland
| | - Cristina Díaz-Perlas
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | | | - Christian Heinis
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| |
Collapse
|
10
|
Hou D, Swaminathan A, Borichevsky GM, Frampton CM, Kettle AJ, Gearry RB. Plasma Calprotectin and Myeloperoxidase as Biomarkers in Inflammatory Bowel Disease. Inflamm Bowel Dis 2025:izaf110. [PMID: 40411452 DOI: 10.1093/ibd/izaf110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Indexed: 05/26/2025]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a relapsing-remitting illness requiring proactive monitoring of gut inflammation. We aimed to determine the correlations of plasma myeloperoxidase (pMPO) and calprotectin (pCal), two neutrophil proteins, with existing measures of disease activity. METHODS Adults with IBD undergoing ileocolonoscopy were recruited prospectively. Baseline assessments included blood tests (pMPO, pCal, and C-reactive protein ([CRP]), symptom questionnaires, and endoscopic indices (simple endoscopic score for CD [SES-CD] and UC endoscopic index of severity [UCEIS]). Active IBD was defined as SES-CD > 2 and UCEIS ≥ 2. Spearman's rank correlations assessed the associations between blood markers and endoscopic activity. The area under the receiver operating characteristics curves (AUROC) and univariable logistic regression assessed the ability of blood markers (at optimal thresholds) to identify active disease. RESULTS In total, 170 participants were included (female, n = 92; Crohn's disease [CD], n = 99; median age 46 years, IQR 35-58). Plasma biomarkers more accurately identified active IBD in individuals with UC (AUROCpMPO = 0.76, P < .001; AUROCpCal = 0.66, P < .05; AUROCCRP = 0.73, P < .001) than in CD (AUROCpMPO = 0.62, P > .05; AUROCpCal = 0.65, P < .01; AUROCCRP = 0.66, P < .01). In all patients with IBD, the addition of pCal (AUROC = 0.73, P < .001) and pMPO (AUROC = 0.73, P < .001) to CRP added benefit compared to CRP (AUROC = 0.70, P < .001) alone. Plasma myeloperoxidase > 13.86 ng/mL (odds ratio [OR] = 10.13, 3.4-30.16) and pCal > 961.72 ng/mL (OR = 3.38, 1.21-9.4) were associated with an increased odds of having endoscopically active UC. CONCLUSIONS Plasma MPO shows promise as a potential blood-based biomarker of IBD activity, especially in UC. The combined use of pMPO and CRP adds diagnostic utility in discriminating between active versus quiescent IBD.
Collapse
Affiliation(s)
- Duo Hou
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Akhilesh Swaminathan
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Gastroenterology, Christchurch Hospital, Christchurch, New Zealand
| | - Grace M Borichevsky
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris M Frampton
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Antony J Kettle
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Richard B Gearry
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Gastroenterology, Christchurch Hospital, Christchurch, New Zealand
| |
Collapse
|
11
|
Choi G, Bessman NJ. Iron at the crossroads of host-microbiome interactions in health and disease. Nat Microbiol 2025:10.1038/s41564-025-02001-y. [PMID: 40399686 DOI: 10.1038/s41564-025-02001-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/31/2025] [Indexed: 05/23/2025]
Abstract
Iron is an essential dietary micronutrient for both humans and microorganisms. Disruption of iron homeostasis is closely linked, as both a cause and an effect, to the development and progression of gut microbiota dysbiosis and multiple diseases. Iron absorption in humans is impacted by diverse environmental factors, including diet, medication and microbiota-derived molecules. Accordingly, treatment outcomes for iron-associated diseases may depend on an individual patient's microbiome. Here we describe various iron acquisition strategies used by the host, commensal microorganisms and pathogens to benefit or outcompete each other in the complex gut environment. We further explore recently discovered microbial species and metabolites modulating host iron absorption, which represent potential effectors of disease and therapeutic targets. Finally, we discuss the need for mechanistic studies on iron-host-microbiome interactions that can affect disease and treatment outcomes, with the ultimate aim of supporting the development of microbiome-based personalized medicine.
Collapse
Affiliation(s)
- Garam Choi
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, USA
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Nicholas J Bessman
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, USA.
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
12
|
Saheb Sharif-Askari N, Mdkhana B, Hafezi S, Elamin OF, Eladham MW, Al-Sheakly BKS, Halwani H, Saheb Sharif-Askari F, Halwani R. Calprotectin inhibition attenuates silica-induced lung fibrosis. Inflammopharmacology 2025:10.1007/s10787-025-01771-5. [PMID: 40381145 DOI: 10.1007/s10787-025-01771-5] [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: 01/03/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Respirable silica exposure adversely affects lung tissue immunopathology, triggering oxidative bursts in macrophages and neutrophils, releasing Damage-associated molecular patterns (DAMPs), including calprotectin proteins, S100A8, and S100A9. Calprotectin constitutes up to 45% of these innate immune cells, and serum levels of these alarmins correlate with inflammation, fibrosis, remodelling, and drug response in chronic diseases, including inflammatory bowel disease, asthma, and cystic fibrosis. The consequence of releasing calprotectin protein could trigger the pro-fibrotic effect of silicosis. This study aimed to investigate the role of calprotectin (S100A8/S100A9) as a pro-inflammatory and pro-fibrotic mediator in silica-induced lung fibrosis and evaluated the therapeutic potential of the calprotectin inhibitor, paquinimod. Using a mouse model of silicosis, silica exposure significantly elevated calprotectin expression, lung inflammation, and fibrosis, as evidenced by increased levels of epithelial-to-mesenchymal transition (EMT) markers, collagen deposition, and matrix metalloproteinases (MMPs). In vitro, stimulation of human bronchial fibroblasts with S100A8/S100A9 upregulated fibrotic markers (COL1A1 and α-SMA), which were reduced by inhibitors of TLR4 and RAGE receptors, as well as by paquinimod. Treatment with paquinimod effectively reduced these pathological changes, normalized calprotectin levels, decreased fibrosis scores, and attenuated NF-κB activation. These findings highlighted calprotectin's pivotal role in silica-induced lung fibrosis and inflammation, suggesting that its inhibition could be a promising therapeutic approach for silicosis and other fibro-inflammatory lung diseases. Further research is warranted to explore the precise mechanisms linking calprotectin to lung fibrosis and its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Bushra Mdkhana
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Shirin Hafezi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ola Faisal Elamin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mariam Wed Eladham
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Hala Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
13
|
Gade IL, Bodilsen J, Mariager T, Hertz S, Duerlund LS, Holm CK, Madsen PH, Bennike TB, Honoré B. Exhaled breath protein composition in patients hospitalised during the first wave of COVID-19. J Breath Res 2025; 19:036008. [PMID: 40341493 DOI: 10.1088/1752-7163/add617] [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: 11/28/2024] [Accepted: 05/08/2025] [Indexed: 05/10/2025]
Abstract
Coronavirus 2019 (COVID-19) leads to substantial morbidity and excess mortality all over the world which may be aggravated by the propensity of Severe Acute Respiratory Syndrome Coronavirus 2 to mutate. Mechanisms for development of severe COVID-19 are poorly understood. The air we exhale contains endogenous proteins and represents a highly accessible yet unexploited biological sample that can be collected without use of invasive procedures. We collected exhaled breath condensate samples from 28 patients hospitalised due to COVID-19 at admission and discharge using RTubes™. Bottom-up proteomic analysis of tandem mass-tag-labelled single exhaled breath samples was performed in 25 exhaled breath samples collected at admission and 13 samples collected at discharge using discovery-based nano-liquid chromatography-tandem mass spectrometry. In total, 232 proteins were identified in the exhaled breath samples after stringent data filtering. Most of the exhaled proteins were related to the immune systems function and regulation. The levels of four proteins, KRT77, DCD, CASP14 and SERPINB12 decreased from admission to discharge as patients generally recovered from the infection. These proteins are expressed in lung epithelium or macrophages and are associated with the regulation of inflammation drivers in COVID-19. In particular, the alarmins S100A8 and S100A9 accounted for 8% of the exhaled breath proteins. In conclusion, our study demonstrates that analysis of the exhaled breath protein composition can give insights into mechanisms related to inflammation and response to infections, and hereby also of severe COVID-19.Clinical Trial No: NCT04598620.
Collapse
Affiliation(s)
- Inger Lise Gade
- Department of Hematology and Clinical Cancer Research Center, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Jacob Bodilsen
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
- Department of Infectious Diseases, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Theis Mariager
- Department of Infectious Diseases, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Sandra Hertz
- Department of Infectious Diseases, Aalborg University Hospital, 9000 Aalborg, Denmark
| | | | | | - Poul Henning Madsen
- Department of Clinical Biochemistry, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Tue Bjerg Bennike
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Bent Honoré
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
14
|
Rodrigues C, Gomes ATPC, Leal J, Pereira P, Lopes PC, Mendes K, Correia MJ, Veiga N, Rosa N, Soares C, Ministro P. Oral health in inflammatory bowel disease: the overlooked impact and the potential role of salivary calprotectin. BMC Oral Health 2025; 25:729. [PMID: 40375227 DOI: 10.1186/s12903-025-06064-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/28/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Inflammatory Bowel Disease (IBD), a chronic condition characterized by gastrointestinal inflammation, is influenced by genetic and environmental factors. Emerging evidence suggests a "mouth-gut axis," with the oral cavity reflecting extra-intestinal manifestations of IBD. This study evaluated the oral health status of IBD patients and the potential of salivary calprotectin (SCP) as a biomarker for assessing IBD activity and oral health. METHODS Oral health was assessed in 100 IBD patients [60 with Crohn's disease (CD) and 40 with ulcerative colitis (UC)] and 14 controls. Evaluations included the Decayed, Missing, and Filled Teeth (DMFT) Score, Periodontal Diagnosis and the need for dental or prosthetic treatment. Saliva and stool samples were collected to measure SCP and faecal calprotectin (FCP) levels using the Elia Calprotectin 2 Test. IBD activity was evaluated with FCP, the Harvey-Bradshaw Index for CD, and the Partial Mayo Score for UC. RESULTS The DMFT index mean was comparable between IBD patients (mean 7.99, SD 7.73) and controls (mean 10.00, SD 6.49). However, periodontal disease was significantly more prevalent in IBD patients (57% in CD, 70% in UC) than in controls (29%), with severe cases (stages III/IV) more frequent in IBD. Additionally, 89% of IBD patients required dental treatment, and 39% needed prosthetic rehabilitation. SCP levels showed no significant correlation with disease activity or oral health status, while FCP correlated with C-reactive protein and erythrocyte sedimentation rate. CONCLUSIONS This study underscores the need for improved oral health management in IBD patients and suggests that SCP may not be a reliable biomarker for monitoring IBD or periodontal disease. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Cláudio Rodrigues
- Gastroenterology Department, Viseu Dão-Lafões Health Unit, Viseu, Portugal.
| | - Ana T P C Gomes
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal.
| | - Joana Leal
- Clinical Pathology Department, Viseu Dão-Lafões Health Unit, Viseu, 3504-509, Portugal
| | - Pedro Pereira
- Faculty of Dental Medicine, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Pedro C Lopes
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Karina Mendes
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Maria J Correia
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Nélio Veiga
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Nuno Rosa
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Caroline Soares
- Gastroenterology Department, Viseu Dão-Lafões Health Unit, Viseu, Portugal
| | - Paula Ministro
- Gastroenterology Department, Viseu Dão-Lafões Health Unit, Viseu, Portugal
| |
Collapse
|
15
|
Xie S, Wu M, Shang Y, Tuo W, Wang J, Cai Q, Yuan C, Yao C, Xiang Y. Development and validation of an early diagnosis model for severe mycoplasma pneumonia in children based on interpretable machine learning. Respir Res 2025; 26:182. [PMID: 40361124 PMCID: PMC12070602 DOI: 10.1186/s12931-025-03262-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Pneumonia is a major threat to the health of children, especially those under the age of five. Mycoplasma pneumoniae infection is a core cause of pediatric pneumonia, and the incidence of severe mycoplasma pneumoniae pneumonia (SMPP) has increased in recent years. Therefore, there is an urgent need to establish an early warning model for SMPP to improve the prognosis of pediatric pneumonia. METHODS The study comprised 597 SMPP patients aged between 1 month and 18 years. Clinical data were selected through Lasso regression analysis, followed by the application of eight machine learning algorithms to develop early warning model. The accuracy of the model was assessed using validation and prospective cohort. To facilitate clinical assessment, the study simplified the indicators and constructed visualized simplified model. The clinical applicability of the model was evaluated by DCA and CIC curve. RESULTS After variable selection, eight machine learning models were developed using age, sex and 21 serum indicators identified as predictive factors for SMPP. A Light Gradient Boosting Machine (LightGBM) model demonstrated strong performance, achieving AUC of 0.92 for prospective validation. The SHAP analysis was utilized to screen advantageous variables, which contains of serum S100A8/A9, tracheal computed tomography (CT), retinol-binding protein(RBP), platelet larger cell ratio(P-LCR) and CD4+CD25+Treg cell counts, for constructing a simplified model (SCRPT) to improve clinical applicability. The SCRPT diagnostic model exhibited favorable diagnostic efficacy (AUC > 0.8). Additionally, the study found that S100A8/A9 outperformed clinical inflammatory markers can also differentiate the severity of MPP. CONCLUSIONS The SCRPT model consisting of five dominant variables (S100A8/A9, CT, RBP, PLCR and Treg cell) screened based on eight machine learning is expected to be a tool for early diagnosis of SMPP. S100A8/A9 can also be used as a biomarker for validity differentiation of SMPP when medical conditions are limited.
Collapse
Affiliation(s)
- Si Xie
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Mo Wu
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Yu Shang
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Wenbin Tuo
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Jun Wang
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Qinzhen Cai
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China.
| | - Cong Yao
- Health Care Department, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430016, China.
| | - Yun Xiang
- Department of Laboratory Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430016, China.
| |
Collapse
|
16
|
Zhao J, Zhou M, Yang C, Liu YW, Yang T, Sun B, Li B, Zheng J, Dai S, Yang Z, Wang X. S100A9 as a potential novel target for experimental autoimmune cystitis and interstitial cystitis/bladder pain syndrome. Biomark Res 2025; 13:72. [PMID: 40346703 PMCID: PMC12065242 DOI: 10.1186/s40364-025-00763-5] [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: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic inflammatory disease of the bladder for which no effective therapy is currently available. Understanding the pathogenesis of IC/BPS and identifying effective intervention targets are of great clinical importance for its effective treatment. Our work focuses on elucidating the key targets and underlying mechanisms of IC/BPS. METHODS We established an experimental autoimmune cystitis (EAC) mouse model and generated gene knockout mice to elucidate key mediators triggering chronic inflammatory damage in IC/BPS through using single-cell RNA sequencing, proteomic sequencing, and molecular biology experiments. RESULTS Our study revealed that the infiltration and activation of macrophages, T cells, and mast cells exacerbated inflammatory bladder damage in both IC/BPS and EAC mice. Notably, cell-cell communication among bladder immune cells was significantly enhanced in EAC mice. Macrophages, as the main cell types altered in EAC mice, received and transmitted the most intensity signalling. Mechanistically, macrophages synthesized and secreted S100A9, which in turn facilitated macrophage polarization and promoted the production of pro-inflammatory cytokines. S100A9 emerged as an important pro-inflammatory and pathogenic molecule in IC/BPS and EAC. Further analysis demonstrated that S100A9 activation enhanced the inflammatory response and exacerbated bladder tissue damage in IC/BPS patients and EAC mice via TLR4/NF-κB and TLR4/p38 signalling pathways. Importantly, inhibition of S100A9 with paquinimod, as well as genetic knockout of S100A9, significantly attenuated the pathological process. CONCLUSIONS S100A9 is an important pro-inflammatory and pathogenic molecule in IC/BPS and EAC. Targeting S100A9-initiated signalling pathways may offer a novel therapeutic strategy for IC/BPS.
Collapse
Affiliation(s)
- Jiang Zhao
- Department of Urology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, PR China.
- Department of Urology, Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, PR China.
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Mi Zhou
- Department of Biochemistry and Molecular Biology, Army Military Medical University, Chongqing, 400038, PR China
- Department of Central Laboratory, Qianjiang Hospital, Chongqing University, Chongqing, RP, 409000, China
| | - Chengfei Yang
- Department of Thoracic Surgery, Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, PR China
| | - Yang-Wuyue Liu
- Department of Biochemistry and Molecular Biology, Army Military Medical University, Chongqing, 400038, PR China
| | - Teng Yang
- Department of Biochemistry and Molecular Biology, Army Military Medical University, Chongqing, 400038, PR China
| | - Bishao Sun
- Department of Urology, Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, PR China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ji Zheng
- Department of Urology, Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, PR China
| | - Shuangshuang Dai
- Department of Biochemistry and Molecular Biology, Army Military Medical University, Chongqing, 400038, PR China.
| | - Zhenxing Yang
- Department of Blood Transfusion, Irradiation Biology Laboratory, Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, PR China.
| | - Xiangwei Wang
- Department of Urology, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, PR China.
| |
Collapse
|
17
|
Schwerdtfeger LA, Montini F, Chitnis T, Cox LM, Weiner HL. Faecal mucoprotein MUC2 is decreased in multiple sclerosis and is associated with mucin degrading bacteria. EBioMedicine 2025; 116:105721. [PMID: 40344717 DOI: 10.1016/j.ebiom.2025.105721] [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: 12/30/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The gut microbiome is altered in MS and may contribute to disease by disrupting the intestinal barrier. The colonic mucus barrier, which is primarily composed of mucin protein 2 (MUC2), plays a crucial role in providing a barrier between colonic epithelial cells and the microbiome. Disruption of intestinal epithelial and mucus barriers has been reported in inflammatory bowel disease (IBD) and Parkinson's disease (PD) but has not been studied in the context of the microbiome in multiple sclerosis (MS). METHODS We investigated the epithelial tight junction protein zonulin occludins 1 (ZO-1), mucus protein MUC2, inflammatory stool markers (calprotectin), and gut microbiota composition in a cohort of subjects with relapsing and progressive MS. FINDINGS MUC2 was decreased in stool of subjects with both relapsing and progressive MS. ZO-1 was elevated in the serum of subjects with progressive MS but was not altered in the stool. Inflammatory markers typically elevated in IBD and PD, including calprotectin, were not altered in MS stool, suggesting disease specificity of altered gut physiology in MS. Microbiota with known mucus degrading capacity were elevated in the stool of subjects with MS and negatively correlated with mucus protein levels. INTERPRETATION Taken together, these findings suggest reduced gut barrier function in MS which is linked to increased mucin degrading bacteria. FUNDING This work was supported by grants from the National MS Society, the NIH/NINDS, the Nancy Davis Race to Erase MS Young Investigator Award, the Water Cove Charitable Foundation, and the Clara E. and John H. Ware Jr. FOUNDATION
Collapse
Affiliation(s)
- Luke A Schwerdtfeger
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Federico Montini
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Tanuja Chitnis
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Vermeire S, Nitcheu J, Gineste P, Flatres A, Santo J, Scherrer D, Peyrin-Biroulet L, Dulai PS, Danese S, Dubinsky M, Tilg H, Siegmund B, Hisamatsu T, Shan K, Rabbat CJ, Sands BE. Obefazimod in patients with moderate-to-severely active ulcerative colitis: efficacy and safety analysis from the 96-week open-label maintenance phase 2b study. J Crohns Colitis 2025; 19:jjaf074. [PMID: 40417999 PMCID: PMC12124117 DOI: 10.1093/ecco-jcc/jjaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND AND AIMS Obefazimod is an oral small molecule that selectively enhances the expression of a single micro-RNA (miRNA), miR-124. Obefazimod has demonstrated safety and efficacy in patients with moderate-to-severely active ulcerative colitis (UC) in a phase 2b induction trial. This analysis presents the 2-year outcome data of the open-label maintenance (OLM) study. METHODS Patients received placebo or obefazimod 25, 50, or 100 mg once-daily (od) during the induction trial and, irrespective of their clinical response, could enter the 96-week OLM study with obefazimod 50 mg od. Safety was monitored through monthly visits in the first year and quarterly visits in the second year. Efficacy was assessed at weeks 48 and 96 using nonresponder imputation (NRI) for missing data. RESULTS Of 222 eligible patients, 217 were enrolled and 164 (75.6%) completed week 96 of the OLM study. Clinical response was achieved at weeks 48 and 96 in 177 (81.6%) and 158 (72.8%) patients and clinical remission in 119 (54.8%) and 114 (52.5%) of patients. A total of 133 (61.3%) and 128 (59.0%) patients showed endoscopic improvement, and 72 (33.2%) and 78 (35.9%) endoscopic remission. In total, 148/217 patients (68.2%) reported at least 1 treatment-emergent adverse event (TEAE). The most frequent TEAEs were COVID-19 (14.3%), headache (11.5%), UC (7.8%), and nasopharyngitis (6.9%). No new safety risks emerged over 96 weeks. CONCLUSIONS The 96-week OLM study supports the long-term efficacy and favorable safety profile of obefazimod 50 mg od. A phase 3 program with obefazimod in patients with moderate-to-severe UC is ongoing. TRIAL REGISTRATION NAME/NUMBER A phase 2b, open-label, efficacy and safety study of ABX464 as maintenance therapy in patients with moderate-to-severe UC. NCT04023396.
Collapse
Affiliation(s)
- Severine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | | | | | | | | | | | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, INFINY Institute, INSERM NGERE, CHRU Nancy, Vandœuvre-lès-Nancy, France
| | - Parambir S Dulai
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marla Dubinsky
- Pediatric GI and Nutrition, Mount Sinai Kravis Children’s Hospital, New York, NY, United States
| | - Herbert Tilg
- Department of Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Britta Siegmund
- Department of Gastroenterology, Infectiology and Rheumatology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tadakazu Hisamatsu
- Department of Gastroenterology and Hepatology, Kyorin University Hospital, Tokyo, Japan
| | | | | | - Bruce E Sands
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
19
|
Orlandi KN, Harms MJ. Zebrafish do not have a calprotectin ortholog. PLoS One 2025; 20:e0322649. [PMID: 40315184 PMCID: PMC12047837 DOI: 10.1371/journal.pone.0322649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 03/25/2025] [Indexed: 05/04/2025] Open
Abstract
The protein heterodimer calprotectin and its component proteins, S100A8 and S100A9, play important antibacterial and pro-inflammatory roles in the mammalian innate immune response. Gaining mechanistic insights into the regulation and biological function of calprotectin will help facilitate patient diagnostics and therapy for inflammation and further our understanding of the host-microbe interface. Recent literature has identified zebrafish s100a10b as zebrafish calprotectin based on sequence similarity, genomic context, and transcriptional upregulation during the immune response to bacterial infections. The field would benefit from expanding the breadth of calprotectin studies into a zebrafish innate immunity model. Here, we carefully evaluated the possibility that zebrafish possess a calprotectin ortholog or a paralog that convergently evolved similar function. Using careful bioinformatics approaches, we found that zebrafish do not have an ortholog of either mammalian S100A8 or S100A9. To look for paralogs with convergent function, we identified four zebrafish s100 proteins-including s100a10b-that are expressed in immune cells and upregulated during the immune response. We recombinantly expressed and purified these proteins and measured their antimicrobial activity. None of the zebrafish proteins exhibited activity comparable to mammalian calprotectin. We also generated structural models of all homodimers and heterodimers of all annotated zebrafish s100 genes. None of these complexes were predicted to have an antimicrobial transition metal binding site equivalent to calprotectin. Finally, we measured the ability of our four purified zebrafish s100 proteins to activate inflammation via Toll-like receptor 4, a key feature of human S100A9; none of the proteins activated the receptor. Our work demonstrates conclusively that zebrafish have no ortholog of calprotectin and suggests that similar proteins have not convergently evolved analogous functions.
Collapse
Affiliation(s)
- Kona N. Orlandi
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
- Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Michael J. Harms
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
- Department of Chemistry and Biochemistry, University of Oregon, Eugene, Oregon, United States of America
| |
Collapse
|
20
|
Gu JM, Zhao M, Zhu J, Tao HW, Shao XP, Qin LQ, Ge YY, Chen GC. Dietary inflammatory potential, genetic predisposition, and incidence of Crohn's disease and ulcerative colitis. Nutr Metab (Lond) 2025; 22:35. [PMID: 40312362 PMCID: PMC12044715 DOI: 10.1186/s12986-025-00934-z] [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: 12/09/2024] [Accepted: 04/23/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Evidence for a potential link between dietary inflammatory potential and inflammatory bowel disease is limited, and the moderating role of genetic susceptibility remains to be assessed. OBJECTIVE To evaluate energy-adjusted dietary inflammatory index (E-DII) for the associations with incident Crohn's disease (CD) and ulcerative colitis (UC) and the role of genetic susceptibility. METHODS A total of 205,706 UK Biobank participants who were aged 39-72 years and had no known CD or UC at baseline (2006-2010) were included. The E-DII score was calculated based on energy-adjusted average intakes of 33 food or nutrient items derived from up to five 24-hour dietary recalls. Multivariable Cox regression models were used estimate hazard ratios (HRs) with 95% confidence interval (CI) for incident CD and UC. RESULTS During a median 12.3 years of follow-up, 382 incident CD and 798 incident UC cases were ascertained. A higher E-DII score was not associated with risk of CD (HR Q4 VS. Q1 = 1.28, 95% CI: 0.94-1.74; P-trend = 0.09) or UC (HR Q4 VS. Q1 = 1.10, 95% CI: 0.90-1.36; P-trend = 0.17). There was an interaction between the E-DII and the polygenic risk score (PRS) for CD on incident CD (P-interaction = 0.023), with an association only among participants with a high PRS (HR Q4 VS. Q1 = 1.64, 95% CI: 1.03-2.61) (P-interaction = 0.023). As compared with the participants with a low PRS for CD and a low E-DII score, participants with high levels of both scores had a particularly higher risk of CD (HR = 3.12; 95% CI: 1.74-5.60). CONCLUSIONS The association of dietary inflammatory potential with incident CD appears to be amplified by high genetic susceptibility to CD.
Collapse
Affiliation(s)
- Ji-Mei Gu
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Miao Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Jie Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Hao-Wei Tao
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Xiao-Ping Shao
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China
| | - Yang-Yang Ge
- Department of Radiation Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, 30 Tongyang North Road, Nantong, 226361, China.
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, The Fourth Affiliated Hospital, Suzhou Medical College of Soochow University, 199 Ren'ai Road, Suzhou, 215123, China.
| |
Collapse
|
21
|
Whiley PJ, Dixit OVA, Das Gupta M, Patel H, Zhao G, Connor SJ, Summers KM, Hume DA, Pavli P, O'Brien CL. Transcriptomic signatures of host immune responses in aphthous ulcers, the earliest lesions of Crohn's disease, suggest that bacterial uptake, rather than global dysbiosis, is the initiating factor. Immunol Cell Biol 2025; 103:473-484. [PMID: 40386941 DOI: 10.1111/imcb.70031] [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: 10/01/2024] [Revised: 03/08/2025] [Accepted: 04/05/2025] [Indexed: 05/20/2025]
Abstract
Crohn's disease is a chronic, transmural inflammatory disease of the human gut. Changes in the fecal microbial composition and dysbiosis are consistent features in studies of Crohn's disease patients, but whether dysbiosis is a cause or consequence of inflammation remains unresolved. Genetic susceptibility plays a role in the development of Crohn's disease and has been linked to genes involved in recognition of intestinal bacteria by the mononuclear phagocyte system. The earliest visible lesions in Crohn's disease are aphthous ulcers, overlying Peyer's patches and lymphoid follicles. To identify mechanisms underlying the earliest stages of disease we compared gene expression in aphthous ulcers, Peyer's patches, inflamed and endoscopically normal mucosa from patients and controls using total RNA-seq. The resulting data were subjected to network analysis to identify coregulated gene expression signatures of cell types and processes. These results were compared to single-cell RNA-seq analysis of intestinal macrophages in normal and diseased mucosa. The analysis of aphthous ulcers revealed signatures of epithelial stress and antimicrobial defense, plasma cell activation and immunoglobulin production, monocyte recruitment, inflammatory gene expression and induction of interferon-γ. These signatures were not present in the normal appearing mucosa adjacent to aphthous ulcers, which were similar to healthy control mucosa. Given the role of Peyer's patches and lymphoid follicles in sampling the luminal contents, these findings suggest the initial lesion in Crohn's disease arises from the uptake of bacteria and the activation of multiple host defense pathways rather than the breakdown of epithelial barrier integrity and widespread bacterial translocation.
Collapse
Affiliation(s)
- Phillip J Whiley
- School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia
- Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, ACT, Australia
| | - Ojas V A Dixit
- Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, ACT, Australia
| | - Mukta Das Gupta
- School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia
- Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, ACT, Australia
| | - Hardip Patel
- John Curtin School of Medical Research, College of Health and Medicine, Australian National University, Canberra, ACT, Australia
| | - Guoyan Zhao
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan J Connor
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW, Australia
- Department of Gastroenterology, Liverpool Hospital, Sydney, NSW, Australia
- Ingham Institute, Sydney, NSW, Australia
| | - Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Paul Pavli
- School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia
- Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, ACT, Australia
| | - Claire L O'Brien
- Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| |
Collapse
|
22
|
Hochnadel I, Hoenicke L, Petriv N, Suo H, Groebe L, Olijnik C, Bondarenko N, Alfonso JC, Jarek M, Shi R, Jeron A, Timrott K, Hirsch T, Jedicke N, Bruder D, Klawonn F, Lichtinghagen R, Geffers R, Lenzen H, Manns MP, Yevsa T. In vivo RNAi screen and validation reveals Ngp, Hba-a1, and S100a8 as novel inhibitory targets on T lymphocytes in liver cancer. Front Immunol 2025; 16:1549229. [PMID: 40352930 PMCID: PMC12061932 DOI: 10.3389/fimmu.2025.1549229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/21/2025] [Indexed: 05/14/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) represents the third deadliest cancer worldwide with limited treatment options. Immune checkpoint inhibitors (ICIs) have revolutionized HCC therapy, but immune suppression within the tumor microenvironment remains a major challenge. Therefore, in this study, we aimed to define novel ICI molecules arising on T cells during aggressive HCC development. Methods Using autochthonous HCC models, we performed microarray analyses followed by in vivo RNA interference screen and identified several new ICI molecules on CD4 and CD8 T lymphocytes in HCC-bearing mice. Short hairpin RNA (shRNA)-mediated knockdown of the ICI molecules was performed to validate their functional role in T cell activity and survival of HCC-bearing mice. Finally, we searched for the presence of the defined ICI molecules in HCC patients. Results We identified neutrophilic granule protein (Ngp), hemoglobin subunit alpha-1 (Hba-a1), and S100 calcium-binding protein a8 (S100a8) as novel inhibitory molecules of T cells in HCC. The specific shRNA-based knockdown of these inhibitory targets was safe, led to a downregulation of classical ICI molecules (PD-1, PD-L1, 4-1BBL, CD160), and kept liver parameters under control in murine HCC. Besides, we detected upregulation of S100A8 and S100A9 in blood and liver tissues in HCC patients, supporting their clinical relevance. Conclusion The obtained results pave the way for the use of the newly defined ICI molecules Ngp, Hba-a1, and S100a8 as novel immunotherapeutic targets in further preclinical and clinical studies in HCC patients.
Collapse
Affiliation(s)
- Inga Hochnadel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lisa Hoenicke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Nataliia Petriv
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lothar Groebe
- Experimental Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Chantal Olijnik
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Nina Bondarenko
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Department of Pathological Anatomy, Forensic Medicine and Pathological Physiology, Dnipro State Medical University, Dnipro, Ukraine
| | - Juan C. Alfonso
- Department of Systems Immunology, Technical University Braunschweig and HZI, Braunschweig, Germany
| | | | - Ruibing Shi
- Biostatistics Research Group, HZI, Braunschweig, Germany
| | - Andreas Jeron
- Immune Regulation Group, HZI, Braunschweig, Germany
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Kai Timrott
- Department of General, Visceral and Transplant Surgery, MHH, Hannover, Germany
| | | | - Nils Jedicke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Dunja Bruder
- Immune Regulation Group, HZI, Braunschweig, Germany
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Frank Klawonn
- Biostatistics Research Group, HZI, Braunschweig, Germany
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Center, Technical University of Munich, Munich, Germany
- Department of Computer Science, Ostfalia University, Wolfenbüttel, Germany
| | | | | | - Henrike Lenzen
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Department of Gastroenterology, Hepatology, Interventional Endoscopy and Diabetology, Academic Teaching Hospital Braunschweig, Braunschweig, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
23
|
Huss M, Elger T, Kunst C, Loibl J, Krautbauer S, Liebisch G, Kandulski A, Müller M, Tews HC, Buechler C. Fecal Arachidonic Acid: A Potential Biomarker for Inflammatory Bowel Disease Severity. Int J Mol Sci 2025; 26:4034. [PMID: 40362272 PMCID: PMC12071911 DOI: 10.3390/ijms26094034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Arachidonic acid levels are elevated in the colonic mucosa of patients with inflammatory bowel disease (IBD). Fecal metabolites are emerging as valuable diagnostic tools for IBD. This study aimed to investigate associations between 31 fecal fatty acids, including arachidonic acid, to identify potential correlations with disease severity. Among the 31 fatty acids analyzed in feces, dihomo-γ-linolenic acid, arachidonic acid, and adrenic acid were significantly increased in patients with IBD compared to controls. In contrast, levels of linoleic acid and γ-linolenic acid, the precursors of arachidonic acid, were similar between both groups. No significant differences in fatty acid levels were observed between patients with Crohn's disease and ulcerative colitis. Arachidonic acid and adrenic acid levels positively correlated with fecal calprotectin, a clinically established marker of IBD severity, but showed no association with stool consistency or the Gastrointestinal Symptom Rating Scale. This suggests that these fatty acids are linked to disease severity rather than disease-related symptoms. Current IBD-specific medications had no significant impact on the fecal levels of any of the 31 fatty acids. In summary, this study demonstrates elevated fecal levels of dihomo-γ-linolenic acid, arachidonic acid, and adrenic acid in IBD patients. Normal levels of precursor fatty acids suggest that impaired downstream metabolism may contribute to the accumulation of these n-6 polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Muriel Huss
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Tanja Elger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Claudia Kunst
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Johanna Loibl
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany; (S.K.); (G.L.)
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany; (S.K.); (G.L.)
| | - Arne Kandulski
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Martina Müller
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Hauke Christian Tews
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| | - Christa Buechler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (M.H.); (C.K.); (J.L.); (A.K.); (M.M.); (H.C.T.)
| |
Collapse
|
24
|
Wan R, Fang S, Zhang X, Zhou W, Bi X, Yuan L, Lv Q, Song Y, Tang W, Shi Y, Li T. S100A9 as a promising therapeutic target for diabetic foot ulcers. Chin Med J (Engl) 2025; 138:973-981. [PMID: 40143429 PMCID: PMC12037093 DOI: 10.1097/cm9.0000000000003543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Diabetic foot is a complex condition with high incidence, recurrence, mortality, and disability rates. Current treatments for diabetic foot ulcers are often insufficient. This study was conducted to identify potential therapeutic targets for diabetic foot. METHODS Datasets related to diabetic foot and diabetic skin were retrieved from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using R software. Enrichment analysis was conducted to screen for critical gene functions and pathways. A protein interaction network was constructed to identify node genes corresponding to key proteins. The DEGs and node genes were overlapped to pinpoint target genes. Plasma and chronic ulcer samples from diabetic and non-diabetic individuals were collected. Western blotting, immunohistochemistry, and enzyme-linked immunosorbent assays were performed to verify the S100 calcium binding protein A9 (S100A9), inflammatory cytokine, and related pathway protein levels. Hematoxylin and eosin staining was used to measure epidermal layer thickness. RESULTS In total, 283 common DEGs and 42 node genes in diabetic foot ulcers were identified. Forty-three genes were differentially expressed in the skin of diabetic and non-diabetic individuals. The overlapping of the most significant DEGs and node genes led to the identification of S100A9 as a target gene. The S100A9 level was significantly higher in diabetic than in non-diabetic plasma (178.40 ± 44.65 ng/mL vs. 40.84 ± 18.86 ng/mL) and in chronic ulcers, and the wound healing time correlated positively with the plasma S100A9 level. The levels of inflammatory cytokines (tumor necrosis factor-α, interleukin [IL]-1, and IL-6) and related pathway proteins (phospho-extracellular signal regulated kinase [ERK], phospho-p38, phospho-p65, and p-protein kinase B [Akt]) were also elevated. The epidermal layer was notably thinner in chronic diabetic ulcers than in non-diabetic skin (24.17 ± 25.60 μm vs. 412.00 ± 181.60 μm). CONCLUSIONS S100A9 was significantly upregulated in diabetic foot and was associated with prolonged wound healing. S100A9 may impair diabetic wound healing by disrupting local inflammatory responses and skin re-epithelialization.
Collapse
Affiliation(s)
- Renhui Wan
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Shuo Fang
- Department of Plastics, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xingxing Zhang
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Weiyi Zhou
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xiaoyan Bi
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Le Yuan
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Qian Lv
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yan Song
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Wei Tang
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yongquan Shi
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Tuo Li
- Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
25
|
Fang HM. Intricacy of Crohn's disease: Incongruity between diagnostic modalities and histopathologic assessment. World J Gastrointest Endosc 2025; 17:103979. [PMID: 40291133 PMCID: PMC12019121 DOI: 10.4253/wjge.v17.i4.103979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 04/14/2025] Open
Abstract
Crohn's disease (CD) is a chronic and recurrent inflammatory condition. Histologic healing is associated with better outcomes in CD, while less is known regarding the assessment of histological condition. Recently, a study has examined the discordance between endoscopic and histopathologic assessment in ileal CD, revealing a poor correlation between endoscopic and histologic evaluations in assessing mucosal inflammation and disease activity. However, the involvement of CD can span the entire gastrointestinal tract, as well as numerous clinical manifestations and extraintestinal complications, and the patchy nature of transmural inflammation is a well-established characteristic of this disease. The diagnosis of CD relies on a comprehensive evaluation that includes clinical, biochemical, stool, endoscopic, cross-sectional imaging, and histological investigations due to the incomplete understanding of its etiology and pathogenesis. Upon diagnosis, complimentary investigations should focus on markers of disease activity. Since transmural inflammation can only be assessed in resections, therefore, we primarily focused on the evaluation value of clinical aspects, histological scoring systems, particular in vivo imaging evaluation such as computed tomography enterography, magnetic resonance elastography, scintigraphy, sonographically measurement, endoscopic ultrasonography, and advanced endoscopic imaging techniques.
Collapse
Affiliation(s)
- Hai-Ming Fang
- Department of Gastroenterology, Pingshan Hospital of Southern Medical University, Pingshan District People’s Hospital of Shenzhen, Shenzhen 518118, Guangdong Province, China
| |
Collapse
|
26
|
Issa IA, Issa T. Assessing endoscopic remission in small bowel Crohn's disease: Are markers enough? World J Gastrointest Endosc 2025; 17:106083. [PMID: 40291128 PMCID: PMC12019123 DOI: 10.4253/wjge.v17.i4.106083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/23/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Mucosal healing in Crohn's disease (CD) has been established as a crucial target of treatment, leading to long term remission and decrease in complication rates. Endoscopy still serves as the gold standard for assessment, particularly in the small bowel where balloon or capsule enteroscopy is frequently needed. However, these modalities are often unavailable, expensive, and invasive, posing risks to patients. Consequently, the identification of accessible and reliable biomarkers, especially in small intestinal CD, remains a challenge. The study by Ohno et al, published in this issue, further illuminates this field. It confirms the potential role of fecal biomarker leucine-rich α2 glycoprotein (LRG) and validates findings from previous smaller trials. Comparing to other markers LRG showed a much higher predictive value for mucosal healing of the small bowel, making it a useful option for small intestinal CD follow up. In this editorial, we explore the optimal marker of inflammation or mucosal healing in CD, particularly in the small bowel. We provide an overview of available conventional biomarkers and introduce several novel biomarkers, including an update on emerging technologies and innovations.
Collapse
Affiliation(s)
- Iyad A Issa
- Department of Gastroenterology and Hepatology, Harley Street Medical Center, Abu Dhabi 41475, United Arab Emirates
| | - Taly Issa
- Medical School, University of Nicosia, Nicosia 24005, Lefkosía, Cyprus
| |
Collapse
|
27
|
Sthity RA, Islam MZ, Sagar MEK, Gazi MA, Ferdous J, Kabir MM, Mahfuz M, Ahmed T, Mostafa I. Association of Escherichia coli pathotypes with fecal markers of enteropathy and nutritional status among underweight adults in Bangladesh. Front Cell Infect Microbiol 2025; 15:1553688. [PMID: 40276386 PMCID: PMC12018316 DOI: 10.3389/fcimb.2025.1553688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Environmental enteric dysfunction (EED), a subclinical intestinal disorder, is characterized by chronic fecal-oral exposure to entero-pathogens and could be diagnosed by measuring non-invasive biomarkers. Escherichia coli is the one of the key bacterial enteric pathogens that drives EED, but there is a lack of information on the E. coli pathotypes in relation to the biomarkers of EED in malnourished adults. Here, we intended to measure the possible association of these pathotypes with EED biomarkers and nutritional status of adults residing in a slum in Bangladesh. Method Fecal samples were collected from 524 malnourished adults (BMI ≤18.5 kg/m2) living in a slum-setting in Dhaka from March 2016 to September 2019 and analyzed by TaqMan Array Card assays to evaluate the presence of E. coli pathotypes and other entero-pathogens. The multivariable linear regression model was used to assess the association. Results In these malnourished adults, the most prevalent pathotype of E. coli was EAEC (61.7%) and the least prevalent was STEC (6.7%). The prevalence of atypical EPEC, ETEC and Shigella/EIEC were 52%, 48.9% and 45.1% respectively. The infection with atypical EPEC had significant positive association with levels of Myeloperoxidase (b = 0.38; 95% CI = 0.11, 0.65; p-value = 0.006). Similarly, a significantly higher concentration of alpha-1-antitrypsin (b = 0.13; 95% CI = 0.03, 0.22; p-value = 0.011) was found in the STEC-infected adults. However, no notable association was found between the E. coli pathotypes and nutritional status of these adult participants. Moreover, Plesiomonas infected adults were more likely to be infected with EAEC (p-value = 0.017), ETEC (p-value <0.001) and STEC (pvalue = 0.002). Significant coinfection was also detected among the pathotypes and other entero-pathogens such as Giardia, Ascaris, Campylobacter, Salmonella, Enterocytozoon bieneusi, and Adenovirus. Discussion The study results imply that there is an influence of particular E. coli pathotypes (EPEC and STEC) on intestinal inflammation and gut permeability of the malnourished Bangladeshi adults, but no association with nutritional status is found. Potential pathogenicity of the E. coli pathotypes is also observed when co-infection with other pathogens exists in these adults.
Collapse
Affiliation(s)
- Rahvia Alam Sthity
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Zahidul Islam
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Ehsanul Kabir Sagar
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Jafrin Ferdous
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mamun Kabir
- Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Office of the Executive Director, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | - Ishita Mostafa
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
28
|
Kalkan AE, BinMowyna MN, Raposo A, Ahmad MF, Ahmed F, Otayf AY, Carrascosa C, Saraiva A, Karav S. Beyond the Gut: Unveiling Butyrate's Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review. Nutrients 2025; 17:1305. [PMID: 40284169 PMCID: PMC12029953 DOI: 10.3390/nu17081305] [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: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within this narrative review, the vital physiological functions of SCFAs were examined, with emphasis on butyrate's role as an energy source for colonocytes and its ability to enhance the gut barrier while exhibiting anti-inflammatory effects. Knowledge of butyrate synthesis, primarily generated by Firmicutes bacteria, can be influenced by diets with specifically high contents of resistant starches and fiber. Butyrate can inhibit histone deacetylase, modulate gene expression, influence immune functionality, and regulate tight junction integrity, supporting the idea of its role in gut barrier preservation. Butyrate possesses systemic anti-inflammatory properties, particularly, its capacity to reduce pro-inflammatory cytokines and maintain immune homeostasis, highlighting its therapeutic potential in managing dysbiosis and inflammatory diseases. Although butyrate absorption into circulation is typically minimal, its broader health implications are substantial, especially regarding obesity and type 2 diabetes through its influence on metabolic regulation and inflammation. Furthermore, this narrative review thoroughly examines butyrate's growing recognition as a modulator of neurological health via its interaction with the gut-brain axis. Additionally, butyrate's neuroprotective effects are mediated through activation of specific G-protein-coupled receptors, such as FFAR3 and GPR109a, and inhibition of histone deacetylases (HDACs). Research indicates that butyrate can alleviate neurological disorders, including Alzheimer's, Parkinson's, autism spectrum disorder, and Huntington's disease, by reducing neuroinflammation, enhancing neurotransmitter modulation, and improving histone acetylation. This focus will help unlock its full therapeutic potential for metabolic and neurological health, rather than exclusively on its well-known benefits for gut health, as these are often interconnected.
Collapse
Affiliation(s)
- Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia;
| | - Abdullah Y. Otayf
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain;
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
29
|
Luo R, Liang Z, Chen H, Bao D, Lin X. Potential diagnostic markers and therapeutic targets for DM2 and periodontitis based on bioinformatics analysis. PLoS One 2025; 20:e0320061. [PMID: 40173189 PMCID: PMC11964240 DOI: 10.1371/journal.pone.0320061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/11/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Diabetes Mellitus type 2 (DM2) is thought to have a bidirectional relationship with Periodontitis (PD). However, the complex molecular interactions between DM2 and PD remain unclear. This study aimed to explore the shared genes and common signatures of DM2 and PD via bioinformatic analysis. METHODS Firstly, using bioinformatic methods to investigate common genes. The series matrix files of GSE6751 for DM and GSE15932 for PD were downloaded from the Gene Expression Omnibus (GEO) database. The data was normalized using the R package, and the limma package was utilized to identify the Differentially Expressed Genes (DEGs). Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of DEGs were performed using the "clusterProfiler" package in the R software. The protein-protein network was constructed to analyze the potential relationship among the proteins. CytoHubba, a plugin for the Cytoscape software, was used to identify the hub genes. The validation datasets selected for DM2 and PD were GSE10334 and GSE7014, respectively. Receiver Operating Characteristic (ROC) curve analysis was performed to obtain the area under the ROC curve. Lipopolysaccharide (LPS) + high glucose-induced DM-related PD was simulated to verify the three hub genes through quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) and Western blot (WB). RESULTS In total, 44 common DEGs were identified. ITGAM, H2BC21, S100A9 was identified as he hub genes of DM2 and PD, with all of them were up-regulated. In addition, the area under the curve of all three hub genes was more than 0.65. In-vitro experiments revealed that the relative expression of S100A9 was increased after the treatment with LPS + high glucose. Besides, TLR4 and p-NF-κB levels were also improved in model group. CONCLUSION S100A9 was identified as the hub gene of DM2 and PD. S100A9 could trigger TLR4 signaling way to promote disease development, which can be the potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Rong Luo
- Department of Pharmacy, Taizhou First People’s Hospital, Taizhou, China
| | - Zhenye Liang
- Department of Pharmacy, Taizhou First People’s Hospital, Taizhou, China
| | - Huijun Chen
- Department of Pharmacy, Taizhou First People’s Hospital, Taizhou, China
| | - Dandan Bao
- Department of Pharmacy, Taizhou First People’s Hospital, Taizhou, China
| | - Xinlu Lin
- Department of Pharmacy, Taizhou First People’s Hospital, Taizhou, China
| |
Collapse
|
30
|
Fitria AL, Dinana Z, Maharani AT, Fumizuki AS, Mumtazya FN, Atmaka DR, Yamani LN, Juniastuti, Lusida MI, Soetjipto, Puspikawati SI. Level of fecal neopterin and calprotectin in asymptomatic and symptomatic norovirus-infected children with malnutrition in Indonesia. J Infect Public Health 2025; 18:102687. [PMID: 39933424 DOI: 10.1016/j.jiph.2025.102687] [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: 10/25/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Norovirus infection is the second most frequent viral agent causing gastroenteritis worldwide. Asymptomatic norovirus infection is prevalent in children and is associated with growth failure. Evaluating the intestinal inflammation of asymptomatic norovirus infection is pivotal to overcoming malnutrition, especially in resource-limited settings. The current study aimed to compare the level of fecal neopterin and calprotectin, as the well-established biomarker for intestinal inflammation, in malnourished children with symptomatic and asymptomatic norovirus infections. METHODS In total 78 stool samples from children under five years old were analyzed in this comparative study. The 67 of the participants were malnourished children, with 24 had asymptomatic (ANV) and 18 had symptomatic norovirus infection (SNV), while 25 had no infections (Mal). We assigned 11 healthy children without any infection as control (Healthy). We assessed the fecal neopterin and calprotectin from the stools samples collected during viral diarrhea surveillance in Surabaya, East Java, Indonesia, using commercially ELISA kit from BT Lab (Zhejiang, China) for human neopterin and calprotectin. The statistical differences were tested using Kruskal-Wallis nonparametric test. RESULTS The level of fecal neopterin (mean ± SD, 15.46 ± 1.75 nmol/L and 15.12 ± 2.41 nmol/L; p-value: 0.744) and calprotectin (mean ± SD, 184.35 ± 35.09 ng/ml and 177.95 ± 49.55 ng/ml; p-value: 0.457) between ANV and SNV was comparable, also between Mal and Healthy groups. Meanwhile, the level of both biomarkers was significantly different in norovirus positive group and norovirus negative group (p-value < 0.000). CONCLUSIONS Norovirus infection, regardless of the presence of symptoms, triggered intestinal inflammation, represented by the increased of fecal neopterin and calprotectin level.
Collapse
Affiliation(s)
- Anisa Lailatul Fitria
- Department of Nutrition, Faculty of Public Health, Universitas Airlangga, Indonesia; Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia
| | - Zayyin Dinana
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia
| | - Aussie Tahta Maharani
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia
| | - Alinda Syifa Fumizuki
- Undergraduate Program of Nutrition, Faculty of Public Health, Universitas Airlangga, Indonesia
| | - Faiz Nabila Mumtazya
- Undergraduate Program of Nutrition, Faculty of Public Health, Universitas Airlangga, Indonesia
| | | | - Laura Navika Yamani
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Epidemiology, Faculty of Public Health, Universitas Airlangga, Indonesia
| | - Juniastuti
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Microbiology, Faculty of Medicine, Universitas Airlangga, Indonesia
| | - Maria Inge Lusida
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Microbiology, Faculty of Medicine, Universitas Airlangga, Indonesia
| | - Soetjipto
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Biochemistry, Faculty of Medicine, Universitas Airlangga, Indonesia.
| | | |
Collapse
|
31
|
Jin S, Zhang M, Gu D, Zhu X, Wang Z, Shi X, Hao Y, Xu S. Prediction of postoperative recurrence of perianal fistulizing Crohn's disease by fecal calprotectin combined with serum miRNA6086. J Int Med Res 2025; 53:3000605251328245. [PMID: 40215410 PMCID: PMC12033616 DOI: 10.1177/03000605251328245] [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: 01/12/2025] [Accepted: 02/28/2025] [Indexed: 04/29/2025] Open
Abstract
ObjectivesThis study aimed to identify risk factors for postoperative recurrence in patients with perianal fistulizing Crohn's disease and assess the predictive value of fecal calprotectin and serum miRNA6086.MethodsFrom 105 patients with perianal fistulizing Crohn's disease, blood and fecal samples as well as clinical data were collected. Analysis of blood tests, C-reactive protein, miRNA6086, and fecal calprotectin revealed postoperative recurrence risk factors. Receiver operating characteristic curve analysis assessed the predictive accuracy of miRNA6086 and fecal calprotectin for perianal fistulizing Crohn's disease recurrence and determined their optimal cutoff values, sensitivity, and specificity.ResultsOf the 105 patients with perianal fistulizing Crohn's disease, 33 (31.4%) experienced recurrence. Anal fistula type, preoperative miRNA6086, and fecal calprotectin levels were identified as independent risk factors for postoperative recurrence. Receiver operating characteristic curve analysis revealed that miRNA6086 had a cutoff value of 0.3195, sensitivity of 65.28%, specificity of 66.67%, and area under curve value of 0.6589 (95% confidence interval, 0.5503-0.7674). Fecal calprotectin had a cutoff value of 0.6073, sensitivity of 81.94%, specificity of 78.79%, and area under curve value of 0.8224 (95% confidence interval, 0.5503-0.7674). Combined miRNA6086 and fecal calprotectin detection had a cutoff value of 0.7121, sensitivity of 83.33%, specificity of 87.88%, and area under curve value of 0.9146 (95% confidence interval, 0.8547-0.9744).ConclusionAnal fistula type, preoperative miRNA6086, and fecal calprotectin levels are independent risk factors for perianal fistulizing Crohn's disease recurrence. Combined detection of miRNA6086 and fecal calprotectin levels enhances predictive accuracy for postoperative recurrence.
Collapse
Affiliation(s)
- Shengnan Jin
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Maozhen Zhang
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongdong Gu
- Department of Blood transfusion, Yancheng No. 1 People’s Hospital, Yancheng, China
| | - Xingping Zhu
- Basic Education College, Yancheng kindergarten Teachers College, Yancheng, China
| | - Zuolei Wang
- School of Mathematics and Statistics, Yancheng Teachers University, Yancheng, China
| | - Xuerong Shi
- School of Mathematics and Statistics, Yancheng Teachers University, Yancheng, China
| | - Yanping Hao
- Department of Gastroenterology, Yancheng No. 1 People’s Hospital, Yancheng, China
| | - Su Xu
- Department of Anorectal Surgery, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, China
| |
Collapse
|
32
|
Khan H, Girdharry NR, Massin SZ, Abu-Raisi M, Saposnik G, Mamdani M, Qadura M. Current Prognostic Biomarkers for Peripheral Arterial Disease: A Comprehensive Systematic Review of the Literature. Metabolites 2025; 15:224. [PMID: 40278353 PMCID: PMC12029480 DOI: 10.3390/metabo15040224] [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: 02/22/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Peripheral arterial disease (PAD) is a chronic atherosclerotic disease characterized by atheromatous plaque buildup within arteries of the lower limbs. It can lead to claudication, skin ulcerations, and, in severe cases, chronic limb-threatening ischemia, requiring amputation. There are several plasma protein biomarkers that have been suggested as prognostic markers for adverse events, including major adverse cardiovascular and limb events. However, the clinical benefit and ability to clinically adapt these biomarkers remains uncertain due to inconsistent findings possibly related to heterogenous study designs and differences in methodology. Objectives: This review aims to evaluate the current literature on the prognostic value of plasma protein biomarkers for PAD, their predictive ability for PAD-related adverse outcomes, and their potential roles in guiding PAD management. Methods: To address these challenges, we conducted a systematic review of MEDLINE, Embase, and Cochrane CENTRAL libraries of the current literature (2010-2024). Results: We found 55 studies that evaluated the prognostic value of 44 distinct plasma proteins across various pathophysiological processes. These included markers of immunity and inflammation, markers of metabolism, cardiac biomarkers, markers of kidney function, growth factors and hormones, markers of coagulation and platelet function, extracellular matrix and tissue remodeling proteins, and transport proteins. This review summarizes the existing evidence for prognostic protein plasma biomarkers for PAD and their association with adverse events related to PAD. Conclusions: With this review, we hope to provide a comprehensive list of the prognostic markers and their value as prognostic biomarkers to guide clinical decision making in these patients.
Collapse
Affiliation(s)
- Hamzah Khan
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.)
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1T8, Canada
| | | | - Sophia Z. Massin
- Toronto General Hospital, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Mohamed Abu-Raisi
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.)
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1T8, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Muhammad Mamdani
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.)
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1T8, Canada
- Vascular Surgery, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Abu Dhabi PO Box 112412, United Arab Emirates
| |
Collapse
|
33
|
Hansen CB, Møller MEE, Pérez-Alós L, Israelsen SB, Drici L, Ottenheijm ME, Nielsen AB, Wewer Albrechtsen NJ, Benfield T, Garred P. Differences in biomarker levels and proteomic survival prediction across two COVID-19 cohorts with distinct treatments. iScience 2025; 28:112046. [PMID: 40124495 PMCID: PMC11927729 DOI: 10.1016/j.isci.2025.112046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/07/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Prognostic biomarkers have been widely studied in COVID-19, but their levels may be influenced by treatment strategies. This study examined plasma biomarkers and proteomic survival prediction in two unvaccinated hospitalized COVID-19 cohorts receiving different treatments. In a derivation cohort (n = 126) from early 2020, we performed plasma proteomic profiling and evaluated innate and complement system immune markers. A proteomic model based on differentially expressed proteins predicted 30-day mortality with an area under the curve (AUC) of 0.81. The model was tested in a validation cohort (n = 80) from late 2020, where patients received remdesivir and dexamethasone, and performed with an AUC of 0.75. Biomarker levels varied considerably between cohorts, sometimes in opposite directions, highlighting the impact of treatment regimens on biomarker expression. These findings underscore the need to account for treatment effects when developing prognostic models, as treatment differences may limit their generalizability across populations.
Collapse
Affiliation(s)
- Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | | | - Laura Pérez-Alós
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Simone Bastrup Israelsen
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
| | - Lylia Drici
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
| | - Maud Eline Ottenheijm
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
| | - Annelaura Bach Nielsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
| | - Nicolai J. Wewer Albrechtsen
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Xu H, Zhu J, Lin X, Chen C, Tao J. A Comprehensive Review of Traditional Chinese Medicine in the Management of Ulcerative Colitis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:435-473. [PMID: 40066486 DOI: 10.1142/s0192415x2550017x] [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: 04/08/2025]
Abstract
Ulcerative colitis (UC) is a chronic, nonspecific inflammatory disorder characterized by symptoms such as abdominal pain, diarrhea, hematochezia, and urgency during defecation. While the primary site of involvement is the colon, UC can extend to encompass the entire rectum and colon. The causes and development mechanisms of UC are still not well understood; nonetheless, it is currently held that factors including environmental influences, genetic predispositions, intestinal mucosal integrity, gut microbiota composition, and immune dysregulation contribute to its development. Dysregulated immune responses are pivotal in the pathophysiology of UC, and these aberrant responses are considered key contributors to the disease onset. In patients with UC, immune cells become hyperactive and erroneously target normal intestinal tissue, resulting in inflammatory cascades and damage to the intestinal mucosa. The therapeutic strategies currently employed for UC include immunosuppressive agents such as aminosalicylates and corticosteroids. However, these treatments often prove costly and carry significant adverse effects - imposing a considerable burden on patients. Traditional Chinese Medicine (TCM) has attracted worldwide attention because of its multi-target approach, minimal side effects, cost-effectiveness, and favorable efficacy profiles. In this review, the ways in which TCM modulates inflammatory responses in the treatment of ulcerative colitis have been outlined. Research into TCM modalities for modulating inflammatory pathways in the treatment of UC, which has yielded promising advancements, including individual herbs, herbal formulations, and their derivatives, has been summarized. TCM has been utilized to treat UC and the immune system plays a key role in regulating intestinal homeostasis. It is imperative to facilitate large-scale evidence-based medical research and promote the clinical application of TCM in the management of UC.
Collapse
Affiliation(s)
- Huate Xu
- School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
- School of Pharmacy, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
| | - Jinhui Zhu
- School of Pharmacy, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
| | - Xiangyun Lin
- School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
| | - Chao Chen
- School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
| | - Jinhua Tao
- School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
- School of Pharmacy, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, P. R. China
| |
Collapse
|
35
|
Xu J, Gao Y, Huang X, Li J, Sun T, Wang X, Zhao Y, Wang T. S100A9 in sepsis: A biomarker for inflammation and a mediator of organ damage. Biochem Biophys Res Commun 2025; 752:151484. [PMID: 39955951 DOI: 10.1016/j.bbrc.2025.151484] [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: 12/18/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Sepsis is the body's response to infection, which can result in multiple organ failure. The immune imbalance in patients with sepsis leads to high mortality. Recent research has greatly advanced our understanding of sepsis pathophysiology, especially in the regulation of inflammatory pathways and immune suppression. S100A9, an alarmin, plays a critical role in modulating the immune response during sepsis and is associated with the potential for multiple organ dysfunction. In the early stage of sepsis, S100A9 can represent the occurrence of inflammation, while in the late stage of sepsis, S100A9 is related to immune suppression. This review summarizes the latest developments in S100A9 research, including its biological functions, role in immune responses, effects on organ damage across different systems during sepsis, and potential clinical applications. It provides insights into the interactions between S100A9 and the immune response and explores S100A9's involvement in sepsis-associated organ injuries. Additionally, this review outlines a framework for future applications of targeted S100A9 interventions and therapeutic strategies to reduce organ injury in sepsis.
Collapse
Affiliation(s)
- Jinlian Xu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yuru Gao
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Xiao Huang
- Department of Medical Technology, Binzhou Polytechnic, Binzhou, Shandong, 256603, China
| | - Jie Li
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Ting Sun
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Xiaozhi Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yi Zhao
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Tao Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China.
| |
Collapse
|
36
|
Mattern S, Hollfoth V, Bag E, Ali A, Riemenschneider P, Jarboui MA, Boldt K, Sulyok M, Dickemann A, Luibrand J, Fusco S, Franz-Wachtel M, Singer K, Goeppert B, Schilling O, Malek N, Fend F, Macek B, Ueffing M, Singer S. An AI-assisted morphoproteomic approach is a supportive tool in esophagitis-related precision medicine. EMBO Mol Med 2025; 17:441-468. [PMID: 39901020 PMCID: PMC11903792 DOI: 10.1038/s44321-025-00194-7] [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: 11/03/2023] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 02/05/2025] Open
Abstract
Esophagitis is a frequent, but at the molecular level poorly characterized condition with diverse underlying etiologies and treatments. Correct diagnosis can be challenging due to partially overlapping histological features. By proteomic profiling of routine diagnostic FFPE biopsy specimens (n = 55) representing controls, Reflux- (GERD), Eosinophilic-(EoE), Crohn's-(CD), Herpes simplex (HSV) and Candida (CA)-esophagitis by LC-MS/MS (DIA), we identified distinct signatures and functional networks (e.g. mitochondrial translation (EoE), immunoproteasome, complement and coagulations system (CD), ribosomal biogenesis (GERD)), and pathogen-specific proteins for HSV and CA. Moreover, combining these signatures with histological parameters in a machine learning model achieved high diagnostic accuracy (100% training set, 93.8% test set), and supported diagnostic decisions in borderline/challenging cases. Applied to a young patient representing a use case, the external GERD diagnosis could be revised to CD and ICAM1 was identified as highly abundant therapeutic target. This resulted in CyclosporinA as a personalized treatment recommendation by the local multidisciplinary molecular inflammation board. Our integrated AI-assisted morphoproteomic approach allows deeper insights in disease-specific molecular alterations and represents a promising tool in esophagitis-related precision medicine.
Collapse
Affiliation(s)
- Sven Mattern
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
- Center for Personalized Medicine (ZPM), Tübingen, Germany
| | - Vanessa Hollfoth
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Eyyub Bag
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Arslan Ali
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | | | - Mohamed A Jarboui
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Mihaly Sulyok
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Anabel Dickemann
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Julia Luibrand
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Stefano Fusco
- Center for Personalized Medicine (ZPM), Tübingen, Germany
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | | | - Kerstin Singer
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Benjamin Goeppert
- Institute of Pathology and Neuropathology, Hospital RKH Kliniken Ludwigsburg, Ludwigsburg, Germany
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Oliver Schilling
- Institute of Pathology, University Medical Center Freiburg, Faculty of Medicine - University of Freiburg, Freiburg, Germany
- Center for Personalized Medicine (ZPM), Freiburg, Germany
| | - Nisar Malek
- Center for Personalized Medicine (ZPM), Tübingen, Germany
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Falko Fend
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Marius Ueffing
- Core Facility for Medical Proteomics, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Stephan Singer
- Department of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany.
- Center for Personalized Medicine (ZPM), Tübingen, Germany.
| |
Collapse
|
37
|
Pang J, Huang J, Yu J, Li B, Wei S, Cen W, Xuan Y, Yang J, Yu Y, Mo J, Lu J, Zheng X, Zhang J. Immune dysregulation in COVID-19 induced ARDS in kidney transplant recipients revealed by single-cell RNA sequencing. Sci Rep 2025; 15:6895. [PMID: 40011702 PMCID: PMC11865448 DOI: 10.1038/s41598-025-91439-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Since the emergence of COVID-19 at the end of 2019, the disease has led to widespread acute respiratory distress syndrome (ARDS), particularly among kidney transplant recipients (KTRs), who are at increased risk due to long-term immunosuppressive therapy. This study aims to explore the differences in immune responses between kidney transplant recipients and non-kidney transplant recipients in COVID-19-induced ARDS to identify potential therapeutic targets for improving outcomes. Single-cell RNA sequencing was performed on 108,320 cells derived from peripheral blood samples to construct a global single-cell map of COVID-19 induced ARDS in kidney transplant recipients(ARDSKT), COVID-19 induced ARDS in non transplant recipients(ARDSNKT), and healthy controls. Subsequently, using cellular clustering analysis, we obtained single-cell maps of different cell types. We employed enrichment analysis to determine the pathways involved in different subpopulations and focused on the role of key immune cells such as monocytes, megakaryocytes, B cells, and CD8+ T cells in the pathogenesis of ARDS. Significant immune differences were observed between ARDSKT and ARDSNKT. In ARDSKT, the S100A9+ MK subpopulation, which activates the NF-κB signaling pathway, was elevated, promoting inflammation. In contrast, the S100A12+ monocyte subpopulation that activates the chemokine signaling pathway was more abundant in ARDSNKT, reflecting a stronger inflammatory response, while its abundance was reduced in ARDSKT due to immunosuppression. The CXCR4+ B subpopulation, crucial for adaptive immunity, was significantly reduced in ARDSKT. Additionally, the XAF1+ Teff subpopulation, associated with apoptosis, was more abundant in ARDSKT, potentially impairing immune recovery. This study highlights the immune differences between ARDSKT and ARDSNKT, revealing the impact of immunosuppression on immune dysregulation. These findings suggest that targeting specific immune pathways can improve therapeutic strategies for ARDSKT.
Collapse
Affiliation(s)
- Jielong Pang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
- Guangxi Health Commission key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China
| | - Jingyu Huang
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Jianing Yu
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Binbin Li
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Shanshan Wei
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Weiluan Cen
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Yixuan Xuan
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Junzhi Yang
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Yongbing Yu
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Jingjia Mo
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Junyu Lu
- Guangxi Health Commission key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China.
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
- The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, 530007, Guangxi, China.
| | - Xiaowen Zheng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
- Guangxi Health Commission key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China.
- The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, 530007, Guangxi, China.
| | - Jianfeng Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
- Guangxi Health Commission key Laboratory of Emergency and Critical Medicine, Nanning, 530007, China.
- Department of General Practice, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
- The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, 530007, Guangxi, China.
| |
Collapse
|
38
|
Du X, Yu W, Chen F, Jin X, Xue L, Zhang Y, Wu Q, Tong H. HDAC inhibitors and IBD: Charting new approaches in disease management. Int Immunopharmacol 2025; 148:114193. [PMID: 39892171 DOI: 10.1016/j.intimp.2025.114193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/14/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Inflammatory bowel disease (IBD) represents a group of chronic inflammatory disorders of the gastrointestinal tract. Despite substantial advances in our understanding of IBD pathogenesis, the currently available therapeutic options remain limited in their efficacy and often come with significant side effects. Therefore, there is an urgent need to explore novel approaches for the management of IBD. One promising avenue of investigation revolves around the use of histone deacetylase (HDAC) inhibitors, which have garnered considerable attention for their potential in modulating gene expression and curbing inflammatory responses. This review emphasizes the pressing need for innovative drugs in the treatment of IBD, and drawing from a wealth of preclinical studies and clinical trials, we underscore the multifaceted roles and the therapeutic effects of HDAC inhibitors in IBD models and patients. This review aims to contribute significantly to the understanding of HDAC inhibitors' importance and prospects in the management of IBD, ultimately paving the way for improved therapeutic strategies in this challenging clinical landscape.
Collapse
Affiliation(s)
- Xueting Du
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China
| | - Weilai Yu
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Fangyu Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China
| | - Xiaosheng Jin
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Liwei Xue
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Ya Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China; Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Qifang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China.
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, China.
| |
Collapse
|
39
|
Tsenkova M, Brauer M, Pozdeev VI, Kasakin M, Busi SB, Schmoetten M, Cheung D, Meyers M, Rodriguez F, Gaigneaux A, Koncina E, Gilson C, Schlicker L, Herebian D, Schmitz M, de Nies L, Mayatepek E, Haan S, de Beaufort C, Cramer T, Meiser J, Linster CL, Wilmes P, Letellier E. Ketogenic diet suppresses colorectal cancer through the gut microbiome long chain fatty acid stearate. Nat Commun 2025; 16:1792. [PMID: 39979287 PMCID: PMC11842570 DOI: 10.1038/s41467-025-56678-0] [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: 03/14/2024] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
Colorectal cancer (CRC) patients have been shown to possess an altered gut microbiome. Diet is a well-established modulator of the microbiome, and thus, dietary interventions might have a beneficial effect on CRC. An attenuating effect of the ketogenic diet (KD) on CRC cell growth has been previously observed, however the role of the gut microbiome in driving this effect remains unknown. Here, we describe a reduced colonic tumor burden upon KD consumption in a CRC mouse model with a humanized microbiome. Importantly, we demonstrate a causal relationship through microbiome transplantation into germ-free mice, whereby alterations in the gut microbiota were maintained in the absence of continued selective pressure from the KD. Specifically, we identify a shift toward bacterial species that produce stearic acid in ketogenic conditions, whereas consumers were depleted, resulting in elevated levels of free stearate in the gut lumen. This microbial product demonstrates tumor-suppressing properties by inducing apoptosis in cancer cells and decreasing colonic Th17 immune cell populations. Taken together, the beneficial effects of the KD are mediated through alterations in the gut microbiome, including, among others, increased stearic acid production, which in turn significantly reduces intestinal tumor growth.
Collapse
Grants
- OT2 OD030544 NIH HHS
- U2C DK119886 NIDDK NIH HHS
- This work was supported by the Luxembourg National Research Fund (FNR) (grant nos. CORE/C16/BM/11282028 (E.L.), PoC/18/12554295 (E.L.), AFR 17103240 (C.G.), PRIDE17/11823097 (M.T., M.K., L.d.N.) and CORE/15/BM/10404093 (P.W.)), by the Luxembourg National Research Fund and the Fondation Cancer Luxembourg (grant no. CORE/C20/BM/14591557 (E.L.)), AFR 17103240 as well as by the Fondation du Pélican de Mie and Pierre Hippert-Faber under the aegis of the Fondation de Luxembourg (‘Pelican Grant’; M.T. and M.M.), a FNRS-Télévie grant to M.M., no. 7.4565.21-40007364), an Internal Research Project at the University of Luxembourg (MiDiCa—integrated analysis of the effects of microbiome-diet interactions on human colorectal adenocarcinoma enterocytes; E.L., P.W. and S.H.), the Fondation Cancer and the Fondation Kriibskrank Kanner Luxembourg (V.I.P), the Action LIONS Vaincre le Cancer Luxembourg and a European Research Council grant under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 863664 to P.W.). This project was also supported by the Doctoral School in Science and Engineering (M.T., M.K., M.M. and L.d.N.) and the Department of Life Sciences and Medicine at the University of Luxembourg. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.
Collapse
Affiliation(s)
- Mina Tsenkova
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Madita Brauer
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Institute for Advanced Studies, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Vitaly Igorevich Pozdeev
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marat Kasakin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Susheel Bhanu Busi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- UK Centre for Ecology and Hydrology, Wallingford, United Kingdom
| | - Maryse Schmoetten
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dean Cheung
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marianne Meyers
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Fabien Rodriguez
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anthoula Gaigneaux
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Eric Koncina
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric Gilson
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lisa Schlicker
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Martine Schmitz
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Laura de Nies
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Serge Haan
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Carine de Beaufort
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Pediatric Clinic, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Thorsten Cramer
- Department of General, Visceral, Children and Transplantation Surgery, RWTH University Hospital Aachen, Aachen, Germany
| | - Johannes Meiser
- Department of Cancer Research (DOCR), Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Paul Wilmes
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
40
|
Valdés-Mas R, Leshem A, Zheng D, Cohen Y, Kern L, Zmora N, He Y, Katina C, Eliyahu-Miller S, Yosef-Hevroni T, Richman L, Raykhel B, Allswang S, Better R, Shmueli M, Saftien A, Cullin N, Slamovitz F, Ciocan D, Ouyang KS, Mor U, Dori-Bachash M, Molina S, Levin Y, Atarashi K, Jona G, Puschhof J, Harmelin A, Stettner N, Chen M, Suez J, Honda K, Lieb W, Bang C, Kori M, Maharshak N, Merbl Y, Shibolet O, Halpern Z, Shouval DS, Shamir R, Franke A, Abdeen SK, Shapiro H, Savidor A, Elinav E. Metagenome-informed metaproteomics of the human gut microbiome, host, and dietary exposome uncovers signatures of health and inflammatory bowel disease. Cell 2025; 188:1062-1083.e36. [PMID: 39837331 DOI: 10.1016/j.cell.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Host-microbiome-dietary interactions play crucial roles in regulating human health, yet their direct functional assessment remains challenging. We adopted metagenome-informed metaproteomics (MIM), in mice and humans, to non-invasively explore species-level microbiome-host interactions during commensal and pathogen colonization, nutritional modification, and antibiotic-induced perturbation. Simultaneously, fecal MIM accurately characterized the nutritional exposure landscape in multiple clinical and dietary contexts. Implementation of MIM in murine auto-inflammation and in human inflammatory bowel disease (IBD) characterized a "compositional dysbiosis" and a concomitant species-specific "functional dysbiosis" driven by suppressed commensal responses to inflammatory host signals. Microbiome transfers unraveled early-onset kinetics of these host-commensal cross-responsive patterns, while predictive analyses identified candidate fecal host-microbiome IBD biomarker protein pairs outperforming S100A8/S100A9 (calprotectin). Importantly, a simultaneous fecal nutritional MIM assessment enabled the determination of IBD-related consumption patterns, dietary treatment compliance, and small intestinal digestive aberrations. Collectively, a parallelized dietary-bacterial-host MIM assessment functionally uncovers trans-kingdom interactomes shaping gastrointestinal ecology while offering personalized diagnostic and therapeutic insights into microbiome-associated disease.
Collapse
Affiliation(s)
- Rafael Valdés-Mas
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Avner Leshem
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Danping Zheng
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yotam Cohen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lara Kern
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Zmora
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Yiming He
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Corine Katina
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | | | - Tal Yosef-Hevroni
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Richman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Barbara Raykhel
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Allswang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Reut Better
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Shmueli
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Nyssa Cullin
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Fernando Slamovitz
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dragos Ciocan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Uria Mor
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Mally Dori-Bachash
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shahar Molina
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Ghil Jona
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jens Puschhof
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Stettner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jotham Suez
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank Popgen, University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Michal Kori
- Pediatric Gastroenterology Unit, Kaplan Medical Center, Rehovot, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitsan Maharshak
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yifat Merbl
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Shibolet
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Zamir Halpern
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Dror S Shouval
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Raanan Shamir
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Suhaib K Abdeen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Shapiro
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany.
| |
Collapse
|
41
|
Yang J, Shang N, Li Z, Xu J, Zhou X, Zhou H, Luo W, Xu P, Zhou Y, Sheng X, Zhu Z, Zhang M, Ma X, Tan M, Wu H. Oral Lactoferrin-Responsive Formulation Anchoring around Inflammatory Bowel Region for IBD Therapy. Adv Healthc Mater 2025; 14:e2402731. [PMID: 39722174 DOI: 10.1002/adhm.202402731] [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: 10/11/2024] [Indexed: 12/28/2024]
Abstract
Oral formulation is the ideal treatment method for inflammatory bowel disease (IBD) therapy, but the mucosal damage and diarrhea symptoms impede the drug retention around the inflammatory region, severely limiting IBD therapeutic efficacy. To address this, an oral astaxanthin (Ast) precise delivery formulation is developed with the selective Ast anchoring around the inflammatory region by the novel lactoferrin (LF)-responsive flocculation. This formulation also heightens the apparent solubility of Ast with the minimized edible safety risks for the edible raw materials. For in vivo IBD therapy, the precise delivery formulation exhibits remarkable outcomes, including a significant increase in colon length and a 100% survival rate. Furthermore, it is verified that the mechanism of treatment is primarily attributed to the improved immunoregulation, epithelial repair, and gut microbiota remodeling after the LF-responsive flocculation. This effective inflammatory-responsive delivery design is instructive and valuable to develop more precise delivery systems for IBD therapy.
Collapse
Affiliation(s)
- Jinfan Yang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ning Shang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhengqing Li
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ji Xu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xin Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hui Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wen Luo
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Peng Xu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yucheng Zhou
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xueru Sheng
- Liaoning Key Lab of Lignocellulose Chemistry and BioMaterials, College of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, Liaoning, 116034, China
| | - Hao Wu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
42
|
McMahan RH, Boe D, Giesy LE, Najarro KM, Khair S, Walrath T, Frank DN, Kovacs EJ. Advanced Age Worsens Respiratory Function and Pulmonary Inflammation After Burn Injury and This Correlates With Changes in the Fecal Microbiome in Mice. J Burn Care Res 2025; 46:53-60. [PMID: 38837704 DOI: 10.1093/jbcr/irae101] [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: 04/10/2024] [Indexed: 06/07/2024]
Abstract
Cutaneous burn injury in the elderly is associated with poor clinical outcomes and increased pulmonary-related complications. We and others have shown that burn injury triggers a cascade of inflammatory mediators which increase gut permeability and dysbiosis of the fecal microbiota and this is more dramatic in the aged. Since crosstalk between intestinal microbes and the lung, termed the "gut-lung axis," impacts immunity and homeostasis in the airway, we hypothesized that the increased intestinal dysbiosis in age and burn injury may contribute to excessive pulmonary inflammation and poor prognosis after injury. To explore this hypothesis, we used a clinically relevant murine model of burn injury in which young and aged mice are subjected to a 12% TBSA dorsal scald burn or sham injury. About 24 h after injury, lung function was assessed and lungs and feces were collected for analysis of inflammatory mediators and fecal microbial species. The results show that, when compared to younger mice, burn injury in aged mice triggers a decline in respiratory function and exacerbates pulmonary inflammation. In addition to heightened levels of the neutrophil recruiting chemokine CXCL1, aged mice displayed a profound increase in the pro-inflammatory protein, calprotectin, in the lung after burn injury. Comparison of the fecal microbiome and inflammatory markers in the lung revealed unique, age-dependent, correlation patterns between individual taxa and pulmonary inflammation. Taken together, these findings suggest that the postburn dysbiosis of the gut flora in aged mice may contribute to the changes in pulmonary inflammatory profiles.
Collapse
Affiliation(s)
- Rachel H McMahan
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA
| | - Devin Boe
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lauren E Giesy
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kevin M Najarro
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA
| | - Shanawaj Khair
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Travis Walrath
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel N Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elizabeth J Kovacs
- Alcohol Research Program, Burn Research Program, Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
- Veterans Health Administration, Eastern Colorado Health Care System, Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO 80045, USA
| |
Collapse
|
43
|
Awan A, Bartlett A, Blakeley-Ruiz JA, Richie T, Ziegler A, Kleiner M. Source of dietary protein alters the abundance of proteases, intestinal epithelial and immune proteins both directly and via interactions with the gut microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632171. [PMID: 39829768 PMCID: PMC11741435 DOI: 10.1101/2025.01.09.632171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Dietary protein has been shown to impact long-term health outcomes differentially depending on its amount and source. It has been suggested that interactions of the gut microbiota with dietary proteins mediate some of the effects of dietary protein on health outcomes. However, it remains unclear what specific host responses drive the health effects of dietary proteins from different plant and animal sources. Additionally, which specific host responses are mediated by interactions of dietary protein source with the gut microbiota and which host responses are caused by dietary proteins directly is not well understood. We used metaproteomics to quantify dietary, host, and microbial proteins in fecal samples of conventional and germ-free mice fed purified dietary protein from six different plant and animal sources, including casein, egg-white, soy, brown rice, pea, and yeast. We characterized differences in the host fecal proteome across the six dietary protein sources as well as between the conventional and germ-free mice for each source to determine how the host responds to the different dietary protein sources and the role of the gut microbiota in mediating these responses. We found that both the source of dietary protein and the presence or absence of the gut microbiota drive the host response to dietary protein source in the fecal host proteome. Host proteins pertaining to immune response, digestion, and barrier function were differentially abundant in different protein sources with and without the gut microbiota. These changes in the host response correlated with changes in microbial composition and differences in protein digestibility. Our results show how dietary protein sources, through their interactions with the gut microbiota, impact several aspects of host physiology.
Collapse
Affiliation(s)
- Ayesha Awan
- Department of Plant and Microbial Biology, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
| | - Alexandria Bartlett
- Department of Plant and Microbial Biology, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - J. Alfredo Blakeley-Ruiz
- Department of Plant and Microbial Biology, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
| | - Tanner Richie
- Department of Plant and Microbial Biology, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
| | - Amanda Ziegler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
44
|
Yunusbayev B, Bogdanova A, Nadyrchenko N, Danilov L, Bogdanov V, Sergeev G, Altinbaev R, Bilalov F, Yunusbaeva M. Gut dysbiosis narrative in psoriasis: matched-pair approach identifies only subtle shifts correlated with elevated fecal calprotectin. Microbiol Spectr 2025; 13:e0138224. [PMID: 39656003 PMCID: PMC11705824 DOI: 10.1128/spectrum.01382-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/06/2024] [Indexed: 01/11/2025] Open
Abstract
Many studies have reported gut microbiome alterations in psoriasis patients, suggesting dysbiosis. While evidence for dysbiosis and its link to pathogenesis remains inconclusive, murine models of psoriasis suggest that gut microbiome alterations develop in response to psoriasis-like inflammation. Hence, the dominant narrative about gut microbiome alterations' impact on disease should be evaluated critically with more data and a well-powered approach. In this case-control study, we used deep sequencing of fecal samples from 53 psoriasis patients and 47 healthy donors to reconstruct the strain/species-level content of the gut microbiome. Unlike previous studies, we first identified matched pairs for each patient with healthy donors to adjust for microbiome variability and increase power. We found no evidence for depleted gut community diversity and apparent divergence in structure between patients and healthy individuals. However, our matched-pair approach identified a subtle but systematic increase in select bacteria among patients, e.g., Megasphaera elsdenii and Eubacterium CAG 180. We next showed that these enriched species were correlated with elevated biomarkers of intestinal and systemic inflammation and liver function. Functionally, one of the top species, Megasphaera elsdenii, is a potent lactate utilizer in the context of intestinal lactic acidosis and inflammation. While our findings hardly support overt dysbiosis in the large intestine, the observed microbial changes correlate with moderately elevated calprotectin, albeit at levels not enough to diagnose ongoing inflammation. Hence, the sources of elevated inflammatory markers in patients' intestines remain unclear and warrant further investigation to clarify their cause-and-effect relationship with the disease. IMPORTANCE With sufficient taxonomic resolution and sample size, this study critically evaluates new and published data on the gut microbiome in psoriasis patients. It shows that observed taxonomic changes in patients are modest and do not meet strict criteria for gut dysbiosis, at least in the large intestine. Instead, observed taxonomic changes in psoriasis patients can be explained by the microbial response to possible low-grade inflammation with unknown localization in the intestine and unclear impact on the host. The authors point out that published endoscopic data point to the small intestine as the site of gut inflammation. Therefore, further research focused on the small intestine would be informative to clarify the hypothetical gut-psoriasis link.
Collapse
Affiliation(s)
- Bayazit Yunusbayev
- Institute of Translational Biomedicine, Saint-Petersburg State University, Saint-Petersburg, Russia
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Saint-Petersburg, Russia
| | - Anna Bogdanova
- SCAMT institute, ITMO University, Saint Petersburg, Russia
| | | | - Lavrentii Danilov
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Saint-Petersburg, Russia
| | - Viktor Bogdanov
- Multiomics Laboratory, Moscow Institute of Physics and Technology, Moscow, Russia
- Medical Genetics Laboratory, Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | | | - Radick Altinbaev
- Institute of Higher Nervous Activity and Neurophysiology of RAS, Moscow, Russia
| | - Fanil Bilalov
- Department of Laboratory Medicine, Bashkir State Medical University, Ufa, Russia
- Republic Medical Genetic Centre, Ufa, Russia
| | - Milyausha Yunusbaeva
- Institute of Translational Biomedicine, Saint-Petersburg State University, Saint-Petersburg, Russia
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Saint-Petersburg, Russia
| |
Collapse
|
45
|
Swaminathan A, Borichevsky GM, Frampton CM, Day AS, Hampton MB, Kettle AJ, Gearry RB. Comparison of Fecal Calprotectin and Myeloperoxidase in Predicting Outcomes in Inflammatory Bowel Disease. Inflamm Bowel Dis 2025; 31:28-36. [PMID: 38417068 PMCID: PMC11700882 DOI: 10.1093/ibd/izae032] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Indexed: 03/01/2024]
Abstract
BACKGROUND Biomarkers have been proposed as surrogate treatment targets for the management of inflammatory bowel disease (IBD); however, their relationship with IBD-related complications remains unclear. This study investigated the utility of neutrophil biomarkers fecal calprotectin (fCal) and fecal myeloperoxidase (fMPO) in predicting a complicated IBD course. METHODS Participants with IBD were followed for 24 months to assess for a complicated IBD course (incident corticosteroid use, medication escalation for clinical disease relapse, IBD-related hospitalizations/surgeries). Clinically active IBD was defined as Harvey-Bradshaw index >4 for Crohn's disease (CD) and simple clinical colitis activity index >5 for ulcerative colitis (UC). Area under the receiver-operating-characteristics curves (AUROC) and multivariable logistic regression assessed the performance of baseline symptom indices, fCal, and fMPO in predicting a complicated disease IBD course at 24 months. RESULTS One hundred and seventy-one participants were included (CD, n = 99; female, n = 90; median disease duration 13 years [interquartile range, 5-22]). Baseline fCal (250 μg/g; AUROC = 0.77; 95% confidence interval [CI], 0.69-0.84) and fMPO (12 μg/g; AUROC = 0.77; 95% CI, 0.70-0.84) predicted a complicated IBD course. Fecal calprotectin (adjusted OR = 7.85; 95% CI, 3.38-18.26) and fMPO (adjusted OR = 4.43; 95% CI, 2.03-9.64) were associated with this end point after adjustment for other baseline variables including clinical disease activity. C-reactive protein (CRP) was inferior to fecal biomarkers and clinical symptoms (pdifference < .05) at predicting a complicated IBD course. A combination of baseline CRP, fCal/fMPO, and clinical symptoms provided the greatest precision at identifying a complicated IBD course. CONCLUSIONS Fecal biomarkers are independent predictors of IBD-related outcomes and are useful adjuncts to routine clinical care.
Collapse
Affiliation(s)
- A Swaminathan
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Gastroenterology, Christchurch Hospital, New Zealand
| | - G M Borichevsky
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - C M Frampton
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - A S Day
- Department of Paediatrics, University of Otago Christchurch, Christchurch, New Zealand
| | - M B Hampton
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - A J Kettle
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - R B Gearry
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Gastroenterology, Christchurch Hospital, New Zealand
| |
Collapse
|
46
|
Armas-Ingavélez MP, Galárraga-Pérez EA. Utility of fecal calprotectin in the diagnosis of inflammatory bowel diseases. SALUD, CIENCIA Y TECNOLOGÍA 2025; 5:1125. [DOI: 10.56294/saludcyt20251125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Introduction: Inflammatory bowel disease involves two main disorders: Crohn's disease and ulcerative colitis. Colonoscopy with biopsy is considered the gold standard for the diagnosis of IBD, although these are invasive and costly techniques. In recent years, fecal calprotectin has gained relevance as a non-invasive biomarker with significant clinical utility. The effectiveness of fecal calprotectin has been demonstrated in distinguishing between IBD and irritable bowel syndrome, predicting endoscopic and histological activity, as well as disease recurrence.Objective: Review the current literature on the clinical utility of fecal calprotectin in the diagnosis and management of inflammatory bowel diseases.Methods: Systematic review based on the PRISMA method, of studies obtained through searches in Scopus, PubMed, Virtual Health Library, Web of Science, Latindex, and Google Scholar. Primary and secondary studies published in the last five years in English and Spanish were included.Results: Nineteen studies were analyzed, describing high sensitivity and specificity in distinguishing IBD from IBS, helping to reduce the rate of unnecessary colonoscopies in patients with non-specific gastrointestinal symptoms. Additionally, fecal calprotectin was found to significantly correlate with endoscopic and histological activity.Conclusions: Fecal calprotectin is a reliable biomarker of mucosal inflammation, capable of identifying patients with a higher likelihood of having IBD, allowing for better management of colonoscopy resources and reducing associated costs.
Collapse
|
47
|
Lawrence ALE, Berger RP, Hill DR, Huang S, Yadagiri VK, Bons B, Fields C, Knight JS, Wobus CE, Spence JR, Young VB, Abuaita BH, O'Riordan MX. Neutrophil prime unique transcriptional responses in intestinal organoids during infection with nontyphoidal Salmonella enterica serovars. mSphere 2024; 9:e0069324. [PMID: 39565098 DOI: 10.1128/msphere.00693-24] [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: 08/22/2024] [Accepted: 09/16/2024] [Indexed: 11/21/2024] Open
Abstract
Nontyphoidal strains of Salmonella enterica are a major cause of foodborne illnesses, and infection with these bacteria results in inflammatory gastroenteritis. Polymorphonuclear leukocytes (PMNs), also known as neutrophils, are a dominant immune cell type found at the site of infection in Salmonella-infected individuals, but how they regulate infection outcome is not well understood. Here, we used a co-culture model of primary human PMNs and human intestinal organoids to probe the role of PMNs during infection with two of the most prevalent Salmonella serovars: Salmonella enterica serovar Enteritidis and Typhimurium. Using a transcriptomics approach, we identified a dominant role for PMNs in mounting differential immune responses including production of pro-inflammatory cytokines, chemokines, and antimicrobial peptides. We also identified specific gene sets that were induced by PMNs in response to Enteritidis or Typhimurium infection. By comparing host responses to these serovars, we uncovered differential regulation of host metabolic pathways particularly induction of cholesterol biosynthetic pathways during Typhimurium infection and suppression of RNA metabolism during Enteritidis infection. Together, these findings provide insight into the role of human PMNs in modulating different host responses to pathogens that cause similar disease in humans.IMPORTANCENontyphoidal serovars of Salmonella enterica are known to induce robust recruitment of polymorphonuclear leukocytes (PMNs) in the gut during early stages of infection, but the specific role of PMNs in regulating infection outcome of different serovars is poorly understood. Due to differences in human infection progression compared to small animal models, characterizing the role of PMNs during infection has been challenging. Here, we used a co-culture model of human intestinal organoids with human primary PMNs to study the role of PMNs during infection of human intestinal epithelium. Using a transcriptomics approach, we define PMN-dependent reprogramming of the host response to Salmonella, establishing a clear role in amplifying pro-inflammatory gene expression. Additionally, the host response driven by PMNs differed between two similar nontyphoidal Salmonella serovars. These findings highlight the importance of building more physiological infection models to replicate human infection conditions to study host responses specific to individual pathogens.
Collapse
Affiliation(s)
- Anna-Lisa E Lawrence
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ryan P Berger
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David R Hill
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sha Huang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Veda K Yadagiri
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brooke Bons
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Courtney Fields
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jason S Knight
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Vincent B Young
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Basel H Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Napoli M, Immler R, Rohwedder I, Lupperger V, Pfabe J, Gonzalez Pisfil M, Yevtushenko A, Vogl T, Roth J, Salvermoser M, Dietzel S, Slak Rupnik M, Marr C, Walzog B, Sperandio M, Pruenster M. Cytosolic S100A8/A9 promotes Ca 2+ supply at LFA-1 adhesion clusters during neutrophil recruitment. eLife 2024; 13:RP96810. [PMID: 39699020 DOI: 10.7554/elife.96810] [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] [Indexed: 12/20/2024] Open
Abstract
S100A8/A9 is an endogenous alarmin secreted by myeloid cells during many acute and chronic inflammatory disorders. Despite increasing evidence of the proinflammatory effects of extracellular S100A8/A9, little is known about its intracellular function. Here, we show that cytosolic S100A8/A9 is indispensable for neutrophil post-arrest modifications during outside-in signaling under flow conditions in vitro and neutrophil recruitment in vivo, independent of its extracellular functions. Mechanistically, genetic deletion of S100A9 in mice caused dysregulated Ca2+ signatures in activated neutrophils resulting in reduced Ca2+ availability at the formed LFA-1/F-actin clusters with defective β2 integrin outside-in signaling during post-arrest modifications. Consequently, we observed impaired cytoskeletal rearrangement, cell polarization, and spreading, as well as cell protrusion formation in S100a9-/- compared to wildtype (WT) neutrophils, making S100a9-/- cells more susceptible to detach under flow, thereby preventing efficient neutrophil recruitment and extravasation into inflamed tissue.
Collapse
Affiliation(s)
- Matteo Napoli
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Roland Immler
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Ina Rohwedder
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Valerio Lupperger
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Johannes Pfabe
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Mariano Gonzalez Pisfil
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Anna Yevtushenko
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Melanie Salvermoser
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Steffen Dietzel
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Marjan Slak Rupnik
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Barbara Walzog
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Markus Sperandio
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| | - Monika Pruenster
- Walter Brendel Center of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University, Planegg-Martinsried, München, Germany
| |
Collapse
|
49
|
Lesiak A, Paprocka P, Wnorowska U, Mańkowska A, Król G, Głuszek K, Piktel E, Spałek J, Okła S, Fiedoruk K, Durnaś B, Bucki R. Significance of host antimicrobial peptides in the pathogenesis and treatment of acne vulgaris. Front Immunol 2024; 15:1502242. [PMID: 39744637 PMCID: PMC11688235 DOI: 10.3389/fimmu.2024.1502242] [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: 09/26/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Acne vulgaris (AV) is a chronic inflammatory condition of the pilosebaceous units characterized by multiple immunologic, metabolic, hormonal, genetic, psycho-emotional dysfunctions, and skin microbiota dysbiosis. The latter is manifested by a decreased population (phylotypes, i.e., genetically distinct bacterial subgroups that play different roles in skin health and disease) diversity of the predominant skin bacterial commensal - Cutinbacterium acnes. Like in other dysbiotic disorders, an elevated expression of endogenous antimicrobial peptides (AMPs) is a hallmark of AV. AMPs, such as human β-defensins, cathelicidin LL-37, dermcidin, or RNase-7, due to their antibacterial and immunomodulatory properties, function as the first line of defense and coordinate the host-microbiota interactions. Therefore, AMPs are potential candidates for pharmaceutical prophylaxis or treating this condition. This study outlines the current knowledge regarding the importance of AMPs in AV pathomechanism in light of recent transcriptomic studies. In particular, their role in improving the tight junctions (TJs) skin barrier by activating the fundamental cellular proteins, such as PI3K, GSK-3, aPKC, and Rac1, is discussed. We hypothesized that the increased expression of AMPs and their patterns in AV act as a compensatory mechanism to protect the skin with an impaired permeability barrier. Therefore, AMPs could be key determinants in regulating AV development and progression, linking acne-associated immune responses and metabolic factors, like insulin/IGF-1 and PI3K/Akt/mTOR/FoxO1 signaling pathways or glucotoxicity. Research and development of anti-acne AMPs are also addressed.
Collapse
Affiliation(s)
- Agata Lesiak
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Paulina Paprocka
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Angelika Mańkowska
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Grzegorz Król
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Katarzyna Głuszek
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Białystok, Poland
| | - Jakub Spałek
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Otolaryngology, Holy-Cross Oncology Center of Kielce, Head and Neck Surgery, Kielce, Poland
| | - Sławomir Okła
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Otolaryngology, Holy-Cross Oncology Center of Kielce, Head and Neck Surgery, Kielce, Poland
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Bonita Durnaś
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Clinical Microbiology, Holy-Cross Oncology Center of Kielce, Kielce, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
50
|
Xu Z, Zhu J, Ma Z, Zhen D, Gao Z. Combined Bulk and Single-Cell Transcriptomic Analysis to Reveal the Potential Influences of Intestinal Inflammatory Disease on Multiple Sclerosis. Inflammation 2024:10.1007/s10753-024-02195-z. [PMID: 39680254 DOI: 10.1007/s10753-024-02195-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
Multiple sclerosis (MS) and inflammatory bowel disease (IBD) are both autoimmune disorders caused by dysregulated immune responses. Still, there is a growing awareness of the comorbidity between MS and IBD. However, the shared pathophysiological mechanisms between these two diseases are still lacking. RNA sequencing datasets (GSE126124, GSE9686, GSE36807, GSE21942) were analyzed to identify the shared differential expressed genes (DEGs) for IBD and experimental allergic encephalomyelitis (EAE). Other datasets (GSE17048, GSE75214, and GSE16879) were downloaded for further verification and analysis. Shared pathways and regulatory networks were explored based on these DEGs. The single-cell transcriptome of central nervous system (CNS) immune cells sequenced from EAE brains and the public datasets of IBD (PRJCA003980) were analyzed for the immune characteristics of the shared DEGs. Mass cytometry by time-of-flight (CyTOF) of peripheral blood mononuclear cells (PBMCs) was performed for the systematic immune response in the EAE model. Machine learning algorithms were also used to identify the diagnostic biomarkers of MS. We identified 74 common DEGs from the selected RNA sequencing datasets, and single-cell RNA data of the intestinal tissues of IBD patients showed that 56 of 74 DEGs were highly enriched in IL1B+ macrophages. These 56 DEGs, defined as inflammation-related DEGs (IRGs), were also highly expressed in pro-inflammatory macrophages of EAE mice and MS patients. The abundance of systematic CD14+ monocytes was validated by CyTOF data. These IRGs were highly enriched in immune response, NOD-like receptor signaling pathway, IL-18 signaling pathway, and other related pathways. In addition, 'AddModuleScore_UCell' analysis further validated that these IRGs (such as IL1B, S100A8, and other inflammatory factors) are highly expressed mainly in pro-inflammatory macrophages, which play an essential role in pro-inflammatory activation in IBD and multiple sclerosis, such as IL-17 signaling pathway, NF-kappa B signaling pathway, and TNF signaling pathway. Finally, suppressors of cytokine signaling 3(SOCS3) and formyl peptide receptor 2(FPR2) were identified as potential biomarkers by machine learning. Two genes were highly expressed in pro-inflammatory macrophages of IBD and MS disease compared to control, and other datasets and experiments further revealed that SOCS3 and FPR2 were highly expressed in IBD and EAE samples. These shared IRGs, which encode inflammatory cytokines, exhibit high expression levels in inflammatory macrophages in IBD and may play a significant role in the inflammatory cytokine storm in MS patients. Two potential biomarkers, SOCS3 and FPR2, were screened out with great diagnostic value for MS and IBD.
Collapse
Affiliation(s)
- Zhu Xu
- Neurological Department, Affiliated Hospital of Guizhou Medical University, Guizhou, China.
- Guizhou Medical University, Guizhou, China.
| | - Junyu Zhu
- Neurological Department, Affiliated Hospital of Guizhou Medical University, Guizhou, China
- Guizhou Medical University, Guizhou, China
| | - Zhuo Ma
- Neurological Department, Affiliated Hospital of Guizhou Medical University, Guizhou, China
- Guizhou Medical University, Guizhou, China
| | - Dan Zhen
- Neurological Department, Affiliated Hospital of Guizhou Medical University, Guizhou, China
- Guizhou Medical University, Guizhou, China
| | - Zindan Gao
- Neurological Department, Affiliated Hospital of Guizhou Medical University, Guizhou, China
- Guizhou Medical University, Guizhou, China
| |
Collapse
|