1
|
Chen W, Xu W, Ma L, Bi C, Yang M, Yang W. Inflammatory biomarkers and therapeutic potential of milk exosome-mediated CCL7 siRNA in murine intestinal ischemia-reperfusion injury. Front Immunol 2025; 15:1513196. [PMID: 39902039 PMCID: PMC11788141 DOI: 10.3389/fimmu.2024.1513196] [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: 10/18/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
Background Intestinal ischemia-reperfusion injury (IIRI) is a severe clinical condition associated with high morbidity and mortality. Despite advances in understanding the pathophysiology of IIRI, effective diagnostic and therapeutic strategies remain limited. Methods Using transcriptome sequencing in a mouse model of IIRI, we identified potential biomarkers that were significantly upregulated in the IIRI group compared to the sham group. Based on these findings, we developed and evaluated a therapeutic strategy using milk-derived exosomes loaded with siRNA targeting CCL7 (M-Exo/siCCL7). Results Focusing on Ccl7 as a hub gene, we explored the therapeutic efficacy of milk-derived exosomes loaded with siRNA targeting Ccl7 (M-Exo/siCCL7) in the IIRI model. M-Exo/siCCL7 treatment effectively attenuated intestinal inflammation and injury, as evidenced by reduced histological damage, decreased serum markers of intestinal barrier dysfunction, and attenuated systemic inflammation. Conclusion Our findings provide new insights into the molecular mechanisms underlying IIRI, identify potential diagnostic biomarkers, and highlight the promise of exosome-based siRNA delivery as a novel therapeutic approach for IIRI.
Collapse
Affiliation(s)
- WenDong Chen
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical
University, Kunming, Yunnan, China
| | | | | | | | | | | |
Collapse
|
2
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
3
|
Li P, Zhou M, Wang J, Tian J, Zhang L, Wei Y, Yang F, Xu Y, Wang G. Important Role of Mitochondrial Dysfunction in Immune Triggering and Inflammatory Response in Rheumatoid Arthritis. J Inflamm Res 2024; 17:11631-11657. [PMID: 39741752 PMCID: PMC11687318 DOI: 10.2147/jir.s499473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 12/15/2024] [Indexed: 01/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease, primarily characterized by chronic symmetric synovial inflammation and erosive bone destruction.Mitochondria, the primary site of cellular energy production, play a crucial role in energy metabolism and possess homeostatic regulation capabilities. Mitochondrial function influences the differentiation, activation, and survival of both immune and non-immune cells involved in RA pathogenesis. If the organism experiences hypoxia, genetic predisposition, and oxidative stress, it leads to mitochondrial dysfunction, which further affects immune cell energy metabolism, synovial cell proliferation, apoptosis, and inflammatory signaling, causing the onset and progression of RA; and, mitochondrial regulation is becoming increasingly important in the treatment of RA.In this review, we examine the structure and function of mitochondria, analyze the potential causes of mitochondrial dysfunction in RA, and focus on the mechanisms by which mitochondrial dysfunction triggers chronic inflammation and immune disorders in RA. We also explore the effects of mitochondrial dysfunction on RA immune cells and osteoblasts, emphasizing its key role in the immune response and inflammatory processes in RA. Furthermore, we discuss potential biological processes that regulate mitochondrial homeostasis, which are of great importance for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Pingshun Li
- College of Integrative Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Mengru Zhou
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Jia Wang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Jiexiang Tian
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Lihuan Zhang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Yong Wei
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Fang Yang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Yali Xu
- College of Integrative Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Gang Wang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| |
Collapse
|
4
|
Miao Y, Wang M, Sun H, Zhang Y, Zhou W, Yang W, Duan L, Niu L, Li Z, Chen J, Li Y, Fan A, Xie Q, Wei S, Bai H, Wang C, Chen Q, Wang X, Li Y, Liu J, Han Y, Fan D, Hong L. Bifidobacterium longum Metabolite Indole-3-Carboxaldehyde Blocks HDAC3 and Inhibits Macrophage NLRP3 Inflammasome Activation in Intestinal Ischemia/Reperfusion Injury. Inflammation 2024:10.1007/s10753-024-02211-2. [PMID: 39663332 DOI: 10.1007/s10753-024-02211-2] [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: 10/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Indole-3-carboxaldehyde (3-IAld), a tryptophan metabolite derived from gut microbiota, has been reported to protect the intestine against radiation injury. This study aimed to clarify the role of Bifidobacterium longum (B. longum) and its metabolite indole-3-carboxaldehyde (3-IAld) in the pathophysiology of intestinal ischemia/reperfusion (II/R) injury. Superior mesenteric artery occlusion and reperfusion were performed to establish II/R mice, and pathological injury in II/R mice was evaluated. II/R mice showed impaired gut microbiota diversity and reduced abundance of B. longum in the intestines. Transplantation of B. longum mitigated II/R injury by protecting the integrity of the intestinal barrier and reducing inflammatory response. The 3-IAld level increased after transplantation of B. longum, and 3-IAld treatment inhibited the inflammatory response of bone marrow-derived macrophages (BMDM). Histone deacetylase 3 (HDAC3) was a target of 3-IAld, and HDAC3 was translocated to mitochondria to promote mitochondrial fatty acid oxidation (FAO) during macrophage inflammasome formation. HDAC3 overexpression promoted the formation of macrophage inflammasomes in intestinal tissues. Overall, this study confirmed the beneficial effects of B. longum in combating II/R injury through HDAC3-mediated control of mitochondrial FAO and macrophage inflammasome formation via 3-IAld.
Collapse
Affiliation(s)
- Yan Miao
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Mian Wang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Hao Sun
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Yujie Zhang
- Department of Histology and Embryology, School of Basic Medicine, Xi'an Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Wei Zhou
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Wanli Yang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Lili Duan
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Liaoran Niu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Zhenshun Li
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Junfeng Chen
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Yiding Li
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Aqiang Fan
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Qibin Xie
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Siyu Wei
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Han Bai
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Chenyang Wang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Qian Chen
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Xiangjie Wang
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Yunlong Li
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Jinqiang Liu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Daiming Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China
| | - Liu Hong
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air Force Medical University, No. 127, Changle West Road, Xi'an, 710032, Shaanxi, P.R. China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, Shaanxi, P.R. China.
| |
Collapse
|
5
|
Wang X, Zhang H, XinZhang, Liu Y. Abscopal effect: from a rare phenomenon to a new frontier in cancer therapy. Biomark Res 2024; 12:98. [PMID: 39228005 PMCID: PMC11373306 DOI: 10.1186/s40364-024-00628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Radiotherapy (RT) controls local lesions, meantime it has the capability to induce systemic response to inhibit distant, metastatic, non-radiated tumors, which is referred to as the "abscopal effect". It is widely recognized that radiotherapy can stimulate systemic immune response. This provides a compelling theoretical basis for the combination of immune therapy combined with radiotherapy(iRT). Indeed, this phenomenon has also been observed in clinical treatment, bringing significant clinical benefits to patients, and a series of basic studies are underway to amplify this effect. However, the molecular mechanisms of immune response induced by RT, determination of the optimal treatment regimen for iRT, and how to amplify the abscopal effect. In order to amplify and utilize this effect in clinical management, these key issues require to be well addressed; In this review, we comprehensively summarize the growing consensus and emphasize the emerging limitations of enhancing the abscopal effect with radiotherapy or immunotherapy. Finally, we discuss the prospects and barriers to the current clinical translational applications.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Haoyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - XinZhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Yao Z, Liang Y, Pan C, Zeng K, Qu Z. Lonicerin alleviates intestinal myenteric neuron injury induced by hypoxia/reoxygenation treated macrophages by downregulating EZH2. J Biochem Mol Toxicol 2024; 38:e23810. [PMID: 39163614 DOI: 10.1002/jbt.23810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/01/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024]
Abstract
Intestinal ischemia-reperfusion (IR) injury is a common gastrointestinal disease that induces severe intestinal dysfunction. Intestinal myenteric neurons participate in maintaining the intestinal function, which will be severely injured by IR. Macrophages are widely reported to be involved in the pathogenesis of organ IR injury, including intestine, which is activated by NLRP3 signaling. Lonicerin (LCR) is a natural extracted monomer with inhibitory efficacy against the NLRP3 pathway in macrophages. The present study aims to explore the potential protective function of LCR in intestinal IR injury. Myenteric neurons were extracted from mice. RAW 264.7 cells were stimulated by H/R with or without 10 μM and 30 μM LCR. Remarkable increased release of IL-6, MCP-1, and TNF-α were observed in H/R treated RAW 264.7 cells, along with an upregulation of NLRP3, cleaved-caspase-1, IL-1β, and EZH2, which were sharply repressed by LCR. Myenteric neurons were cultured with the supernatant collected from each group. Markedly decreased neuron number and shortened length of neuron axon were observed in the H/R group, which were signally reversed by LCR. RAW 264.7 cells were stimulated by H/R, followed by incubated with 30 μM LCR with or without pcDNA3.1-EZH2. The inhibition of LCR on NLRP3 signaling in H/R treated RAW 264.7 cells was abolished by EZH2 overexpression. Furthermore, the impact of LCR on neuron number and neuron axon length in myenteric neurons in the H/R group was abated by EZH2 overexpression. Collectively, LCR alleviated intestinal myenteric neuron injury induced by H/R treated macrophages via downregulating EZH2.
Collapse
Affiliation(s)
- Zhiguang Yao
- Department of Surgical District 2, Eighth People's Hospital of Dongguan City, Dongguan, China
| | - Yuan Liang
- Department of Pediatrics, Eighth People's Hospital of Dongguan City, Dongguan, China
| | - Chunyan Pan
- Department of Health Management, Eighth People's Hospital of Dongguan City, Dongguan, China
| | - Kun Zeng
- Department of Science and Education, Eighth People's Hospital of Dongguan City, Dongguan, China
| | - Zhibo Qu
- Department of Surgical District 2, Eighth People's Hospital of Dongguan City, Dongguan, China
| |
Collapse
|
7
|
Wang J, Yao N, Chen Y, Li X, Jiang Z. Research progress of cGAS-STING signaling pathway in intestinal diseases. Int Immunopharmacol 2024; 135:112271. [PMID: 38762923 DOI: 10.1016/j.intimp.2024.112271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/05/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signal has drawn much consideration due to its sensitivity to DNA in innate immune mechanisms. Activation of the cGAS-STIN signaling pathway induces the production of interferon and inflammatory cytokines, resulting in immune responses, or inflammatory diseases. The intestinal tract is a vital organ for the body's nutrition absorption, recent studies have had various points of view on the job of cGAS-STING pathway in various intestinal sicknesses. Therefore, understanding its role and mechanism in the intestinal environment can help to develop new strategies for the treatment of intestinal diseases. This article examines the mechanism of the cGAS-STING pathway and its function in inflammatory bowel disease, intestinal cancer, and long-injury ischemia-reperfusion, lists the current medications that target it for the treatment of intestinal diseases, and discusses the impact of intestinal flora on this signaling pathway, to offer a theoretical and scientific foundation for upcoming targeted therapies for intestinal disorders via the cGAS-STING pathway.
Collapse
Affiliation(s)
- Jiamin Wang
- College of Pharmacy, Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanji, Jilin 133002, China
| | - Naiqi Yao
- Department of Pharmacy, Yanbian University Hospital, Yanji, Jilin 133000, China
| | - Yonghu Chen
- College of Pharmacy, Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanji, Jilin 133002, China
| | - Xuezheng Li
- College of Pharmacy, Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanji, Jilin 133002, China; Department of Pharmacy, Yanbian University Hospital, Yanji, Jilin 133000, China
| | - Zhe Jiang
- College of Pharmacy, Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanji, Jilin 133002, China; Department of Pharmacy, Yanbian University Hospital, Yanji, Jilin 133000, China.
| |
Collapse
|
8
|
Sun L, Fang K, Yang Z. Combination therapy with probiotics and anti-PD-L1 antibody synergistically ameliorates sepsis in mouse model. Heliyon 2024; 10:e31747. [PMID: 38828304 PMCID: PMC11140784 DOI: 10.1016/j.heliyon.2024.e31747] [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: 10/29/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The study investigated the protective effects and mechanisms of probiotics in conjunction with an anti-PD-L1 antibody on the immune functions of septic mice. Sixty-four mice were assigned to sepsis groups receiving vehicle, probiotics, and anti-PD-L1 antibody individually or in combination, with healthy mice as controls. Sepsis was induced by cecal ligation and puncture (CLP), followed by intraperitoneal Lipopolysaccharide (LPS) injection. Blood and tissues were collected one day post-injection for detecting inflammation-related cytokines, Treg, PI3K/Akt pathway-related protein expression, and lung tissue pathology. The survival time of the remaining ten mice was recorded over seven days. Compared to healthy mice, septic mice given PBS exhibited significantly different serum levels of IL-6, IL-8, IL-17, IL-10, and IFN-γ (all p < 0.001). Treatment with anti-PD-L1 antibody combined with probiotics significantly increased the 7-day survival rate in septic mice, accompanied by decreased pro-inflammatory cytokines, increased anti-inflammatory cytokines, improved oxidative stress, reduced lung injury, and enhanced Th17/Treg balance. This combined therapy demonstrated superior efficacy compared to antibodies or probiotics alone. Additionally, it facilitated peripheral blood polymorphonuclear neutrophil apoptosis, enhancing protection by blocking PD-L1 function and inhibiting PI3K-dependent AKT phosphorylation. In conclusion, combining probiotics with an anti-PD-L1 antibody enhances protective effects in septic mice by reducing serum inflammatory factors, promoting neutrophil apoptosis, regulating Th17/Treg balance, and inhibiting the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Leiming Sun
- Department of Critical Care Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang Province, China
| | - Kun Fang
- Department of Critical Care Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang Province, China
| | - Zheng Yang
- Department of Critical Care Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang Province, China
| |
Collapse
|
9
|
Hof S, Untiedt H, Hübner A, Marcus C, Kuebart A, Herminghaus A, Vollmer C, Bauer I, Picker O, Truse R. Effects of remote ischemic preconditioning on early markers of intestinal injury in experimental hemorrhage in rats. Sci Rep 2024; 14:12960. [PMID: 38839819 PMCID: PMC11153647 DOI: 10.1038/s41598-024-63293-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
The maintenance of intestinal integrity and barrier function under conditions of restricted oxygen availability is crucial to avoid bacterial translocation and local inflammation. Both lead to secondary diseases after hemorrhagic shock and might increase morbidity and mortality after surviving the initial event. Monitoring of the intestinal integrity especially in the early course of critical illness remains challenging. Since microcirculation and mitochondrial respiration are main components of the terminal stretch of tissue oxygenation, the evaluation of microcirculatory and mitochondrial variables could identify tissues at risk during hypoxic challenges, indicate an increase of intestinal injury, and improve our understanding of regional pathophysiology during acute hemorrhage. Furthermore, improving intestinal microcirculation or mitochondrial respiration, e.g. by remote ischemic preconditioning (RIPC) that was reported to exert a sufficient tissue protection in various tissues and was linked to mediators with vasoactive properties could maintain intestinal integrity. In this study, postcapillary oxygen saturation (µHbO2), microvascular flow index (MFI) and plasmatic D-lactate concentration revealed to be early markers of intestinal injury in a rodent model of experimental hemorrhagic shock. Mitochondrial function was not impaired in this experimental model of acute hemorrhage. Remote ischemic preconditioning (RIPC) failed to improve intestinal microcirculation and intestinal damage during hemorrhagic shock.
Collapse
Affiliation(s)
- Stefan Hof
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Hendrik Untiedt
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Anne Hübner
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Carsten Marcus
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Anne Kuebart
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Anna Herminghaus
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christian Vollmer
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Inge Bauer
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Olaf Picker
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Richard Truse
- Department of Anesthesiology, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
10
|
Liao SS, Zhang LL, Zhang YG, Luo J, Kadier T, Ding K, Chen R, Meng QT. Ghrelin alleviates intestinal ischemia-reperfusion injury by activating the GHSR-1α/Sirt1/FOXO1 pathway. FASEB J 2024; 38:e23681. [PMID: 38814725 DOI: 10.1096/fj.202302155rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Ischemia-reperfusion (IR) injury is primarily characterized by the restoration of blood flow perfusion and oxygen supply to ischemic tissue and organs, but it paradoxically leads to tissue injury aggravation. IR injury is a challenging pathophysiological process that is difficult to avoid clinically and frequently occurs during organ transplantation, surgery, shock resuscitation, and other processes. The major causes of IR injury include increased levels of free radicals, calcium overload, oxidative stress, and excessive inflammatory response. Ghrelin is a newly discovered brain-intestinal peptide with anti-inflammatory and antiapoptotic effects that improve blood supply. The role and mechanism of ghrelin in intestinal ischemia-reperfusion (IIR) injury remain unclear. We hypothesized that ghrelin could attenuate IIR-induced oxidative stress and apoptosis. To investigate this, we established IIR by using a non-invasive arterial clip to clamp the root of the superior mesenteric artery (SMA) in mice. Ghrelin was injected intraperitoneally at a dose of 50 μg/kg 20 min before IIR surgery, and [D-Lys3]-GHRP-6 was injected intraperitoneally at a dose of 12 nmol/kg 20 min before ghrelin injection. We mimicked the IIR process with hypoxia-reoxygenation (HR) in Caco-2 cells, which are similar to intestinal epithelial cells in structure and biochemistry. Our results showed that ghrelin inhibited IIR/HR-induced oxidative stress and apoptosis by activating GHSR-1α. Moreover, it was found that ghrelin activated the GHSR-1α/Sirt1/FOXO1 signaling pathway. We further inhibited Sirt1 and found that Sirt1 was critical for ghrelin-mediated mitigation of IIR/HR injury. Overall, our data suggest that pretreatment with ghrelin reduces oxidative stress and apoptosis to attenuate IIR/HR injury by binding with GHSR-1α to further activate Sirt1.
Collapse
Affiliation(s)
- Shi-Shi Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Le-le Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Guo Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Luo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tulanisa Kadier
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Li W, Li Y, Zhao J, Liao J, Wen W, Chen Y, Cui H. Release of damaged mitochondrial DNA: A novel factor in stimulating inflammatory response. Pathol Res Pract 2024; 258:155330. [PMID: 38733868 DOI: 10.1016/j.prp.2024.155330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/03/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
Mitochondrial DNA (mtDNA) is a circular double-stranded genome that exists independently of the nucleus. In recent years, research on mtDNA has significantly increased, leading to a gradual increase in understanding of its physiological and pathological characteristics. Reactive oxygen species (ROS) and other factors can damage mtDNA. This damaged mtDNA can escape from the mitochondria to the cytoplasm or extracellular space, subsequently activating immune signaling pathways, such as NLR family pyrin domain protein 3 (NLRP3), and triggering inflammatory responses. Numerous studies have demonstrated the involvement of mtDNA damage and leakage in the pathological mechanisms underlying various diseases including infectious diseases, metabolic inflammation, and immune disorders. Consequently, comprehensive investigation of mtDNA can elucidate the pathological mechanisms underlying numerous diseases. The prevention of mtDNA damage and leakage has emerged as a novel approach to disease treatment, and mtDNA has emerged as a promising target for drug development. This article provides a comprehensive review of the mechanisms underlying mtDNA-induced inflammation, its association with various diseases, and the methods used for its detection.
Collapse
Affiliation(s)
- Wenting Li
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Yunnan 650500, China
| | - Yuting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jie Zhao
- Department of TCM Endocrinology, Yunnan Provincial Hospital of Traditional Chinese Medicine, Yunnan 650021, China
| | - Jiabao Liao
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Yunnan 650500, China
| | - Weibo Wen
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Yunnan 650500, China.
| | - Yao Chen
- Department of TCM Encephalopathy, Yunnan Provincial Hospital of Traditional Chinese Medicine, Yunnan 650021, China.
| | - Huantian Cui
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Yunnan 650500, China.
| |
Collapse
|
12
|
Randle RK, Amara VR, Popik W. IFI16 Is Indispensable for Promoting HIF-1α-Mediated APOL1 Expression in Human Podocytes under Hypoxic Conditions. Int J Mol Sci 2024; 25:3324. [PMID: 38542298 PMCID: PMC10970439 DOI: 10.3390/ijms25063324] [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: 01/30/2024] [Revised: 02/28/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Genetic variants in the protein-coding regions of APOL1 are associated with an increased risk and progression of chronic kidney disease (CKD) in African Americans. Hypoxia exacerbates CKD progression by stabilizing HIF-1α, which induces APOL1 transcription in kidney podocytes. However, the contribution of additional mediators to regulating APOL1 expression under hypoxia in podocytes is unknown. Here, we report that a transient accumulation of HIF-1α in hypoxia is sufficient to upregulate APOL1 expression in podocytes through a cGAS/STING/IRF3-independent pathway. Notably, IFI16 ablation impedes hypoxia-driven APOL1 expression despite the nuclear accumulation of HIF-1α. Co-immunoprecipitation assays indicate no direct interaction between IFI16 and HIF-1α. Our studies identify hypoxia response elements (HREs) in the APOL1 gene enhancer/promoter region, showing increased HIF-1α binding to HREs located in the APOL1 gene enhancer. Luciferase reporter assays confirm the role of these HREs in transcriptional activation. Chromatin immunoprecipitation (ChIP)-qPCR assays demonstrate that IFI16 is not recruited to HREs, and IFI16 deletion reduces HIF-1α binding to APOL1 HREs. RT-qPCR analysis indicates that IFI16 selectively affects APOL1 expression, with a negligible impact on other hypoxia-responsive genes in podocytes. These findings highlight the unique contribution of IFI16 to hypoxia-driven APOL1 gene expression and suggest alternative IFI16-dependent mechanisms regulating APOL1 gene expression under hypoxic conditions.
Collapse
Affiliation(s)
- Richaundra K. Randle
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA;
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA;
| | - Venkateswara Rao Amara
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA;
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, Bihar, India
| | - Waldemar Popik
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN 37208, USA;
- Department of Internal Medicine, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
13
|
Huang Z, Bai Y, Chen Y, Chen Y, Jiang Y, Zhou J. Attenuation of intestinal ischemia-reperfusion-injury by anesthetics: a potentially protective effect of anesthetic management in experimental studies. Front Pharmacol 2024; 15:1367170. [PMID: 38444936 PMCID: PMC10912591 DOI: 10.3389/fphar.2024.1367170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
Intestinal ischemia-reperfusion injury (IRI) is a potentially severe clinical syndrome after major surgical procedures. In addition to causing intestinal mucosa injury, intestinal IRI further damages distant organs, causing the severity of the condition in patients. So far, effective therapy for intestinal IRI is still absent, and the survival rate of the patients is low. Previous experimental studies have shown that some anesthetics can alleviate intestinal IRI and protect organs while exerting their pharmacological effects, indicating that reasonable perioperative anesthesia management may provide potential benefits for patients to avoid intestinal IRI. These meaningful findings drive scholars to investigate the mechanism of anesthetics in treating intestinal IRI in-depth to discuss the possible new clinical uses. In the present mini-review, we will introduce the protective effects of different anesthetics in intestinal IRI to help us enrich our knowledge in this area.
Collapse
Affiliation(s)
- Zhan Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
- Department of Anesthesiology, Dazhou Integrated TCM & Western Medicine Hospital, Dazhou Second People’s Hospital, Dazhou, China
| | - Yiping Bai
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Ying Chen
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Ye Chen
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yuan Jiang
- Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| |
Collapse
|
14
|
Chu C, Wang X, Yang C, Chen F, Shi L, Xu W, Wang K, Liu B, Wang C, Sun D, Ding W. Neutrophil extracellular traps drive intestinal microvascular endothelial ferroptosis by impairing Fundc1-dependent mitophagy. Redox Biol 2023; 67:102906. [PMID: 37812880 PMCID: PMC10579540 DOI: 10.1016/j.redox.2023.102906] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/11/2023] Open
Abstract
Microvascular endothelial damage caused by intestinal ischemia‒reperfusion (II/R) is a primary catalyst for microcirculation dysfunction and enterogenous infection. Previous studies have mainly focused on how neutrophil extracellular traps (NETs) and ferroptosis cause intestinal epithelial injury, and little attention has been given to how NETs, mainly from circulatory neutrophils, affect intestinal endothelial cells during II/R. This study aimed to unravel the mechanisms through which NETs cause intestinal microvascular dysfunction. We first detected heightened local NET infiltration around the intestinal microvasculature, accompanied by increased endothelial cell ferroptosis, resulting in microcirculation dysfunction in both human and animal II/R models. However, the administration of the ferroptosis inhibitor ferrostatin-1 or the inhibition of NETs via neutrophil-specific peptidylarginine deiminase 4 (Pad4) deficiency led to positive outcomes, with reduced intestinal endothelial ferroptosis and microvascular function recovery. Moreover, RNA-seq analysis revealed a significant enrichment of mitophagy- and ferroptosis-related signaling pathways in HUVECs incubated with NETs. Mechanistically, elevated NET formation induced Fundc1 phosphorylation at Tyr18 in intestinal endothelial cells, which led to mitophagy inhibition, mitochondrial quality control imbalance, and excessive mitochondrial ROS generation and lipid peroxidation, resulting in endothelial ferroptosis and microvascular dysfunction. Nevertheless, using the mitophagy activator urolithin A or AAV-Fundc1 transfection could reverse this process and ameliorate microvascular damage. We first demonstrate that increased NETosis could result in intestinal microcirculatory dysfunction and conclude that suppressed NET formation can mitigate intestinal endothelial ferroptosis by improving Fundc1-dependent mitophagy. Targeting NETs could be a promising approach for treating II/R-induced intestinal microcirculatory dysfunction.
Collapse
Affiliation(s)
- Chengnan Chu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xinyu Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chao Yang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Fang Chen
- School of Medicine, Southeast University, Nanjing, 210002, Jiangsu Province, China
| | - Lin Shi
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Weiqi Xu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Kai Wang
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Baochen Liu
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chenyang Wang
- Key Laboratory of Intestinal Injury, Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, PR China
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Weiwei Ding
- Division of Trauma and Acute Care Surgery, Department of Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
15
|
Lv J, Zhu X, Xing C, Chen Y, Bian H, Yin H, Gu X, Su L. Stimulator of interferon genes (STING): Key therapeutic targets in ischemia/reperfusion injury. Biomed Pharmacother 2023; 167:115458. [PMID: 37699319 DOI: 10.1016/j.biopha.2023.115458] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
The Stimulator of Interferon Genes (STING) is predominantly expressed in immune cells, including macrophages, natural killer cells, dendritic cells, and T cells, functioning as a pattern recognition receptor. STING activation upon detecting cytosolic DNA released from damaged cells initiates downstream pathways, leading to the production of inflammatory cytokines such as IFNs, IL-6, and TNF-α. Dysregulated STING activation has been implicated in inflammatory and metabolic diseases. Ischemia/reperfusion injury (I/RI) is common in stroke, acute myocardial infarction, organ transplantation, and surgeries for certain end-stage diseases. Recent studies suggest that STING could be a novel therapeutic target for I/RI treatment. In this review, we provide a concise overview of the cGAS-STING signaling pathway's general functions and summarize STING's role in I/RI across various organs, including the heart, liver, kidney, and lung. Moreover, we explore potential therapeutic approaches for I/RI by targeting STING.
Collapse
Affiliation(s)
- Juan Lv
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Xuanxuan Zhu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, China
| | - Chunlei Xing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Yuhong Chen
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Huihui Bian
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Heng Yin
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, China.
| | - Xiaofeng Gu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, China.
| | - Li Su
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
16
|
Khalifa AA, Ali MA, Elsokkary NH, Elblehi SS, El-Mas MM. Mitochondrial modulation of amplified preconditioning influences of remote ischemia plus erythropoietin against skeletal muscle ischemia/reperfusion injury in rats. Life Sci 2023; 329:121979. [PMID: 37516431 DOI: 10.1016/j.lfs.2023.121979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
AIMS Skeletal muscle ischemia and reperfusion (S-I/R) injury is relieved by interventions like remote ischemic preconditioning (RIPC). Here, we tested the hypothesis that simultaneous exposure to a minimal dose of erythropoietin (EPO) boosts the protection conferred by RIPC against S-I/R injury and concomitant mitochondrial oxidative and apoptotic defects. MAIN METHODS S-I/R injury was induced in rats by 3-h right hindlimb ischemia followed by 3-h of reperfusion, whereas RIPC involved 3 brief consecutive I/R cycles of the contralateral hindlimb. KEY FINDINGS S-I/R injury caused (i) rises in serum lactate dehydrogenase and creatine kinase and falls in serum pyruvate, (ii) structural deformities like sarcoplasm vacuolations, segmental necrosis, and inflammatory cells infiltration, and (iii) decreased amplitude and increased duration of electromyography action potentials. These defects were partially ameliorated by RIPC and dose-dependently by EPO (500 or 5000 IU/kg). Further, greater repairs of S-I/R-evoked damages were seen after prior exposure to the combined RIPC/EPO-500 intervention. The latter also caused more effective (i) preservation of mitochondrial number (confocal microscopy assessed Mitotracker red staining) and function (citrate synthase activity), (ii) suppression of mitochondrial DNA damage and indices of oxidative stress and apoptosis (succinate dehydrogenase, myeloperoxidase, cardiolipin, and cytochrome c), (iii) preventing calcium and nitric oxide metabolites (NOx) accumulation and glycogen consumption, and (iv) upregulating EPO receptors (EPO-R) gene expression. SIGNIFICANCE dual RIPC/EPO conditioning exceptionally mends structural, functional, and neuronal deficits caused by I/R injury and interrelated mitochondrial oxidative and apoptotic damage. Clinically, the utilization of relatively low EPO doses could minimize the hormone-related adverse effects.
Collapse
Affiliation(s)
- Asmaa A Khalifa
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| | - Mennatallah A Ali
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| | - Nahed H Elsokkary
- Department of Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, Behera, Egypt.
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Kuwait.
| |
Collapse
|
17
|
Li B, Wang Y, Yuan X, Liu G, Diao Y, Liu J. 6-Shogaol from Dried Ginger Protects against Intestinal Ischemia/Reperfusion by Inhibiting Cell Apoptosis via the BDNF/TrkB/PI3K/AKT Pathway. Mol Nutr Food Res 2023; 67:e2200773. [PMID: 37118920 DOI: 10.1002/mnfr.202200773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/12/2023] [Indexed: 04/30/2023]
Abstract
SCOPE Intestinal ischemia-reperfusion (II/R) injury is a common pathological process with high morbidity and mortality. Effective prevention and treatment therapies for II/R are clinically necessary. 6-Shogaol (6-SG), the main active ingredient in dried ginger, behaviors multiple biological activities, including anti-inflammation, antioxidation, and anti-apoptosis. This study aims to elucidate the protective effects and mechanism of 6-SG against II/R-induced injury. METHODS AND RESULTS Sprague-Dawley rats are pre-treated orally with 6-SG and subjected to II/R injury by clamping superior mesenteric artery for 1 h and reperfusion for 2 h. Caco-2 cells are challenged by hypoxia/reoxygenation to mimic II/R in vitro. 6-SG pre-treatment protects against II/R injury by reducing intestinal morphological damage and intestinal barrier injury via inhibiting cell apoptosis. Network pharmacology and molecular docking analyses reveal that 6-SG has a high affinity with brain-derived neurotrophic factor (BDNF) formed homodimer or heterodimer with NT4 instead of the monomer, and thus the dimer configuration is stabilized, activating BDNF/TrkB/PI3K/AKT signaling pathway and inhibiting II/R-induced cell apoptosis. The outcome is further validated both in vivo and in vitro. CONCLUSION 6-Shogaol protects against II/R injury by inhibiting cell apoptosis through the BDNF/TrkB/PI3K/AKT pathway. This study offers a new understanding of the protection mechanism of 6-SG against II/R-induced injury.
Collapse
Affiliation(s)
- Bin Li
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, 116044, China
| | - Yunxiang Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xin Yuan
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Guanting Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, 116044, China
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, 116044, China
| |
Collapse
|
18
|
Clarysse M, Accarie A, Panisello-Roselló A, Farré R, Canovai E, Monbaliu D, De Hertogh G, Vanuytsel T, Pirenne J, Ceulemans LJ. Intravenous Polyethylene Glycol Alleviates Intestinal Ischemia-Reperfusion Injury in a Rodent Model. Int J Mol Sci 2023; 24:10775. [PMID: 37445954 DOI: 10.3390/ijms241310775] [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: 06/07/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Intestinal ischemia-reperfusion injury (IRI) is a common clinical entity, and its outcome is unpredictable due to the triad of inflammation, increased permeability and bacterial translocation. Polyethylene glycol (PEG) is a polyether compound that is extensively used in pharmacology as an excipient in various products. More recently, this class of products have shown to have potent anti-inflammatory, anti-apoptotic, immunosuppressive and cell-membrane-stabilizing properties. However, its effects on the outcome after intestinal IRI have not yet been investigated. We hypothesized that PEG administration would reduce the effects of intestinal IRI in rodents. In a previously described rat model of severe IRI (45 min of ischemia followed by 60 min of reperfusion), we evaluated the effect of IV PEG administration at different doses (50 and 100 mg/kg) before and after the onset of ischemia. In comparison to control animals, PEG administration stabilized the endothelial glycocalyx, leading to reduced reperfusion edema, bacterial translocation and inflammatory reaction as well as improved 7-day survival. These effects were seen both in a pretreatment and in a treatment setting. The fact that this product is readily available and safe should encourage further clinical investigations in settings of intestinal IRI, organ preservation and transplantation.
Collapse
Affiliation(s)
- Mathias Clarysse
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Alison Accarie
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Arnau Panisello-Roselló
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Cientificas (CSIC)-Institut D'Investigacions Biomèdique August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Ricard Farré
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| | - Emilio Canovai
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Diethard Monbaliu
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Gert De Hertogh
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
- Laboratory of Translational Cell & Tissue Research, KU Leuven, 3000 Leuven, Belgium
| | - Tim Vanuytsel
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Jacques Pirenne
- Department of Abdominal Transplant Surgery & Transplant Coordination, University Hospitals Leuven, 3000 Leuven, Belgium
- Abdominal Transplantation Laboratory, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation Center (LIFT), University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
19
|
Wang H, Wang K, Liu B, Bian X, Tan X, Jiang H. The efficacy of bone marrow mesenchymal stem cells on rat intestinal immune-function injured by ischemia/reperfusion. Heliyon 2023; 9:e15585. [PMID: 37131448 PMCID: PMC10149202 DOI: 10.1016/j.heliyon.2023.e15585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 05/04/2023] Open
Abstract
Background Transplantation of bone marrow mesenchymal stem cells (BMSCs) has a promising therapeutic efficiency for varieties of disorders caused by ischemia or reperfusion impairment. It has been shown that BMSCs can mitigate intestinal ischemia/reperfusion (I/R) injuries, but the underlying mechanism is still unclear. This study aimed at investigating the efficacy of BMSCs on the immune function of intestinal mucosal microenvironment after I/R injuries. Methods Twenty adult Sprague-Dawley rats were randomly assigned to a treatment or a control group. All the rats underwent superior mesenteric artery clamping and unclamping. In the treatment group, BMSCs were implanted into the intestine of ten rats by direct submucosal injection whereas the other ten rats in the control group were injected with the same volume of saline. On the fourth and seventh day after BMSCs transplantation, intestinal samples were examined for the CD4 (CD4-positive T-lymphocytes)/CD8 (CD8-positive T-lymphocytes) ratio of the bowel mucosa via flow cytometry, and for the level of Interleukin-2 (IL-2), Interleukin-4 (IL-4) and Interleukin-6 (IL-6) via ELISA. Paneth cell counts and Secretory Immunoglobulin A (SIgA) level were examined via immunohistochemical (IHC) analysis. Real time PCR (RT-PCR) was used to detect the expression levels of tumor necrosis factor-α (TNF-α) and trypsinogen (Serine 2) (PRSS2) genes. White blood cell (WBC) count was measured by manual counting under the microscope. Results The CD4/CD8 ratio in the treatment group was significantly lower compared with that in the control group. The concentration of IL-2 and IL-6 was lower in the treatment group compared with the control group, while the level of IL-4 is the reverse between the two groups. The number of Paneth cells in intestinal mucosa increased significantly, while the level of SIgA in intestinal mucosa decreased significantly, after BMSCs transplantation. The gene expression levels of TNF-α and PRSS2 in intestinal mucosa of treatment group were significantly lower than those of control group. The WBC count in the treatment group was significantly lower than that in the control group. Conclusion We identified immune-relevant molecular changes that may explain the mechanism of BMSCs transplantation efficacy in alleviating rat intestinal immune-barrier after I/R.
Collapse
Affiliation(s)
- He Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, China
| | - Kun Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, China
| | - Bo Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaoqian Bian
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, China
| | - Xiaojie Tan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, China
| | - Haitao Jiang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, China
- Corresponding author. No. 16 Jiangsu Road, Qingdao, Shandong Province, 266003, China.
| |
Collapse
|
20
|
Liu W, Liu T, Zheng Y, Xia Z. Metabolic Reprogramming and Its Regulatory Mechanism in Sepsis-Mediated Inflammation. J Inflamm Res 2023; 16:1195-1207. [PMID: 36968575 PMCID: PMC10038208 DOI: 10.2147/jir.s403778] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/08/2023] [Indexed: 03/22/2023] Open
Abstract
Sepsis is a systemic inflammatory disease caused by an infection that can lead to multiple organ failure. Sepsis alters energy metabolism, leading to metabolic reprogramming of immune cells, which consequently disrupts innate and adaptive immune responses, triggering hyperinflammation and immunosuppression. This review summarizes metabolic reprogramming and its regulatory mechanism in sepsis-induced hyperinflammation and immunosuppression, highlights the significance and intricacies of immune cell metabolic reprogramming, and emphasizes the pivotal role of mitochondria in metabolic regulation and treatment of sepsis. This review provides an up-to-date overview of the relevant literature to inform future research directions in understanding the regulation of sepsis immunometabolism. Metabolic reprogramming has great promise as a new target for sepsis treatment in the future.
Collapse
Affiliation(s)
- Wenzhang Liu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Tianyi Liu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China
- Correspondence: Yongjun Zheng; Zhaofan Xia, Email ;
| | - Zhaofan Xia
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, People’s Republic of China
- Research Unit of Key Techniques for Treatment of burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
21
|
Protective Effect of Oxygen and Isoflurane in Rodent Model of Intestinal Ischemia-Reperfusion Injury. Int J Mol Sci 2023; 24:ijms24032587. [PMID: 36768910 PMCID: PMC9917127 DOI: 10.3390/ijms24032587] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/22/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Animal research in intestinal ischemia-reperfusion injury (IRI) is mainly performed in rodent models. Previously, intraperitoneal (I.P.) injections with ketamine-xylazine mixtures were used. Nowadays, volatile anesthetics (isoflurane) are more common. However, the impact of the anesthetic method on intestinal IRI has not been investigated. We aim to analyze the different anesthetic methods and their influence on the extent of intestinal IRI in a rat model. Male Sprague-Dawley rats were used to investigate the effect of I.P. anesthesia on 60 min of intestinal ischemia and 60 min of reperfusion in comparison to hyperoxygenation (100% O2) and volatile isoflurane anesthesia. In comparison to I.P. anesthesia with room air (21% O2), supplying 100% O2 improved 7-day survival by cardiovascular stabilization, reducing lactic acidosis and preventing vascular leakage. However, this had no effect on the intestinal epithelial damage, permeability, and inflammatory response observed after intestinal IRI. In contrast to I.P. + 100% O2, isoflurane anesthesia reduced intestinal IRI by preventing ongoing low-flow reperfusion hypotension, limiting intestinal epithelial damage and permeability, and by having anti-inflammatory effects. When translating the aforementioned results of this study to clinical situations, such as intestinal ischemia or transplantation, the potential protective effects of hyperoxygenation and volatile anesthetics require further research.
Collapse
|
22
|
Dong YH, Hu JJ, Deng F, Chen XD, Li C, Liu KX, Zhao BC. Use of dexmedetomidine to alleviate intestinal ischemia-reperfusion injury via intestinal microbiota modulation in mice. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1161. [PMID: 36467356 PMCID: PMC9708495 DOI: 10.21037/atm-22-824] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/08/2022] [Indexed: 11/06/2022]
Abstract
Background Intestinal ischemia-reperfusion (I/R) injury is a serious condition with unacceptable mortality rates. Our previous study revealed a protective effect of dexmedetomidine (DEX) on intestinal I/R injury, but its underlying mechanism remains unclear. Gut microbiota imbalance is associated with the progression of I/R injury. We hypothesized that DEX would attenuate intestinal I/R injury via modulating gut microbiota. Methods An I/R injury model was established in C57BL/6 mice in the presence or absence of DEX preconditioning. Some mice were treated with antibiotics to deplete intestinal bacteria. Fecal microbiota transplantation (FMT) was performed by transplanting the feces of DEX-pretreated mice into a new batch of I/R mice. We analyzed the expression of Bacteroidetes and Firmicutes in feces, survival rate, and inflammatory cytokines. Results DEX reversed I/R-induced bacterial abnormalities by increasing the ratio of Firmicutes to Bacteroidetes [DEX + I/R 3.02±0.36 vs. normal saline (NS) + I/R 0.82±0.15; 95% CI: 0.80-3.60; P<0.05] and was accompanied by increased 72-hour survival (0.40±0.16 vs. 0.10±0.09; P<0.05). The protective effect of DEX did not significantly differ from that of DEX + antibiotics. Furthermore, the bacteria of the DEX-pretreated mice decreased the release of inflammatory factors. Conclusions This study revealed that DEX can alleviate intestinal I/R injury through a microbiota-related mechanism, providing a potential avenue for the management of intestinal I/R injury.
Collapse
Affiliation(s)
- Ye-Hong Dong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Dong Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bing-Cheng Zhao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Dery KJ, Kupiec-Weglinski JW. New insights into ischemia-reperfusion injury signaling pathways in organ transplantation. Curr Opin Organ Transplant 2022; 27:424-433. [PMID: 35857344 DOI: 10.1097/mot.0000000000001005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Ischemia-reperfusion injury (IRI) leading to allograft rejection in solid organ transplant recipients is a devastating event that compromises graft and patient survival. As our clinical knowledge regarding its definition and presentation has significantly improved over the last years, adequate biomarkers translating to important therapeutic intervention remains a challenge. This review will summarize recent findings in this area. RECENT FINDINGS In the past 18 months, our understanding of organ transplantation IRI has improved. IRI involves a positive amplification feedback loop encompassing damaged cells at the graft site, the activity of redox-sensitive damage-associated molecular patterns, and local sequestration of recipient-derived monocytes, lymphocytes and polymorphonuclear leukocytes, like neutrophils, to sustain the immunological cascade and to enhance the destruction of the foreign tissue. Recent studies have identified critical components leading to IRI, including the oxidation state of high mobility group box 1, a classic danger signal, its role in the Toll-like receptor 4-interleukin (IL)-23-IL-17A signaling axis, and the role of neutrophils and CD321, a marker for transmigration of circulating leukocytes into the inflamed tissue. In addition, recent findings imply that the protective functions mediated by autophagy activation counterbalance the detrimental nucleotide-binding domain-like receptor family, pyrin domain containing 3 inflammasome pathway. Finally, clinical studies reveal the posttransplant variables associated with early allograft dysfunction and IRI. SUMMARY The future challenge will be understanding how crosstalk at the molecular and cellular levels integrate prospectively to predict which peri-transplant signals are essential for long-term clinical outcomes.
Collapse
Affiliation(s)
- Kenneth J Dery
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | |
Collapse
|
24
|
Kobritz M, Borjas T, Patel V, Coppa G, Aziz M, Wang P. H151, A SMALL MOLECULE INHIBITOR OF STING AS A NOVEL THERAPEUTIC IN INTESTINAL ISCHEMIA-REPERFUSION INJURY. Shock 2022; 58:241-250. [PMID: 35959789 PMCID: PMC9489661 DOI: 10.1097/shk.0000000000001968] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Intestinal ischemia-reperfusion (I/R) injury is a severe disease associated with high mortality. Stimulator of interferon genes (STING) is an intracellular protein that is activated by cytosolic DNA and is implicated in I/R injury, resulting in transcription of type I interferons (IFN-α and IFN-β) and other proinflammatory molecules. Extracellular cold-inducible RNA-binding protein (eCIRP), a damage-associated molecular pattern, induces STING activation. H151 is a small molecule inhibitor of STING that has not yet been studied as a potential therapeutic. We hypothesize that H151 reduces inflammation, tissue injury, and mortality after intestinal I/R. Methods: In vitro, RAW264.7 cells were pretreated with H151 then stimulated with recombinant murine (rm) CIRP, and IFN-β levels in the culture supernatant were measured at 24 hours after stimulation. In vivo, male C57BL/6 mice were subjected to 60-minute intestinal ischemia via superior mesenteric artery occlusion. At the time of reperfusion, mice were intraperitoneally instilled with H151 (10 mg/kg BW) or 10% Tween-80 in PBS (vehicle). Four hours after reperfusion, the small intestines, lungs, and serum were collected for analysis. Mice were monitored for 24 hours after intestinal I/R to assess survival. Results: In vitro, H151 reduced rmCIRP-induced IFN-β levels in a dose-dependent manner. In vivo, intestinal levels of pIRF3 were increased after intestinal I/R and decreased after H151 treatment. There was an increase in serum levels of tissue injury markers (lactate dehydrogenase, aspartate aminotransferase) and cytokine levels (interleukin 1β, interleukin 6) after intestinal I/R, and these levels were decreased after H151 treatment. Ischemia-reperfusion-induced intestinal and lung injury and inflammation were significantly reduced after H151 treatment, as evaluated by histopathologic assessment, measurement of cell death, chemokine expression, neutrophil infiltration, and myeloperoxidase activity. Finally, H151 improved the survival rate from 41% to 81% after intestinal I/R. Conclusions: H151, a novel STING inhibitor, attenuates the inflammatory response and reduces tissue injury and mortality in a murine model of intestinal I/R. H151 shows promise as a potential therapeutic in the treatment of this disease.
Collapse
Affiliation(s)
- Molly Kobritz
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Timothy Borjas
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Vihas Patel
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Gene Coppa
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Monowar Aziz
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| |
Collapse
|
25
|
Wan Y, Dong P, Zhu X, Lei Y, Shen J, Liu W, Liu K, Zhang X. Bibliometric and visual analysis of intestinal ischemia reperfusion from 2004 to 2022. Front Med (Lausanne) 2022; 9:963104. [PMID: 36052333 PMCID: PMC9426633 DOI: 10.3389/fmed.2022.963104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury is a common tissue-organ damage occurring in surgical practice. This study aims to comprehensively review the collaboration and impact of countries, institutions, authors, subject areas, journals, keywords, and critical literature on intestinal I/R injury from a bibliometric perspective, and to assess the evolution of clustering of knowledge structures and identify hot trends and emerging topics. Methods Articles and reviews related to intestinal I/R were retrieved through subject search from Web of Science Core Collection. Bibliometric analyses were conducted on Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio). Results A total of 1069 articles and reviews were included from 2004 to 2022. The number of articles on intestinal I/R injury gradually plateaued, but the number of citations increased. These publications were mainly from 985 institutions in 46 countries, led by China and the United States. Liu Kx published the most papers, while Chiu Cj had the largest number of co-citations. Analysis of the journals with the most outputs showed that most journals focused on surgical sciences, cell biology, and immunology. Macroscopic sketch and microscopic characterization of the entire knowledge domain were achieved through co-citation analysis. The roles of cell death, exosomes, intestinal flora, and anesthetics in intestinal I/R injury are the current and developing research focuses. The keywords "dexmedetomidine", "proliferation", and "ferroptosis" may also become new trends and focus of future research. Conclusion This study comprehensively reviews the research on intestinal I/R injury using bibliometric and visualization methods, and will help scholars better understand the dynamic evolution of intestinal I/R injury and provide directions for future research.
Collapse
Affiliation(s)
- Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Peng Dong
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Xiaobing Zhu
- Department of Anesthesiology, Hospital of Traditional Chinese Medicine of Zhongshan City, Zhongshan, China
| | - Yuqiong Lei
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Junyi Shen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Weifeng Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Kexuan Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Xiyang Zhang
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Zhu XX, Zhang WW, Wu CH, Wang SS, Smith FG, Jin SW, Zhang PH. The Novel Role of Metabolism-Associated Molecular Patterns in Sepsis. Front Cell Infect Microbiol 2022; 12:915099. [PMID: 35719361 PMCID: PMC9202891 DOI: 10.3389/fcimb.2022.915099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Sepsis, a life-threatening organ dysfunction, is not caused by direct damage of pathogens and their toxins but by the host’s severe immune and metabolic dysfunction caused by the damage when the host confronts infection. Previous views focused on the damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), including metabolic proinflammatory factors in sepsis. Recently, new concepts have been proposed to group free fatty acids (FFAs), glucose, advanced glycation end products (AGEs), cholesterol, mitochondrial DNA (mtDNA), oxidized phospholipids (OxPLs), ceramides, and uric acid into metabolism-associated molecular patterns (MAMPs). The concept of MAMPs will bring new guidance to the research and potential treatments of sepsis. Nowadays, sepsis is regarded as closely related to metabolic disorders, and MAMPs play an important role in the pathogenesis and development of sepsis. According to this view, we have explained MAMPs and their possible roles in the pathogenesis of sepsis. Next, we have further explained the specific functions of different types of MAMPs in the metabolic process and their interactional relationship with sepsis. Finally, the therapeutic prospects of MAMPs in sepsis have been summarized.
Collapse
Affiliation(s)
- Xin-xu Zhu
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Wen-wu Zhang
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Cheng-hua Wu
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Shun-shun Wang
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
| | - Fang Gao Smith
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- Academic Department of Anesthesia, Critical Care, Resuscitation and Pain, Heart of England NHS Foundation Trust, Birmingham, United Kingdom
| | - Sheng-wei Jin
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- *Correspondence: Sheng-wei Jin, ; Pu-hong Zhang,
| | - Pu-hong Zhang
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, China
- *Correspondence: Sheng-wei Jin, ; Pu-hong Zhang,
| |
Collapse
|