1
|
Zhang X, Li R, Chen MY, Ye WQ, Liang JZ, Yang WJ, Yang F, Ji HM. Investigating the potential mechanism of Pioglitazone in Sepsis-Related brain injury through transcriptomics. Gene 2024; 931:148892. [PMID: 39187138 DOI: 10.1016/j.gene.2024.148892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
Sepsis-related brain injury (SRBI) refers to brain dysfunction and structural damage caused by sepsis, which is characterized by inflammation, oxidative stress, and destruction of the blood-brain barrier. Pioglitazone is a PPAR-γ agonist in which PPAR-γ acts as an inflammatory modulator, determining the relationship between PPAR-γ and SRBI and inflammatory state is critical for the disease. This study aimed to construct a drug-target-disease network for SRBI and Pioglitazone based on network pharmacology, and to investigate the therapeutic effect and potential mechanism of Pioglitazone in SRBI induced by lipopolysaccharide (LPS) in rats through transcriptomics. To establish a rat Model of SRBI by intraperitoneal injection of LPS (10 mg/kg): SD rats were divided into Control, Model (LPS), Pioglitazone, (LPS + Pioglitazone) and GW9662 group (LPS+GW9662). The effects and potential mechanisms of Pioglitazone in the treatment of SRBI were studied using biochemical indexes, pathological changes and transcriptome-sequencing (RNA-seq). RNA-seq results showed 620 DEGs between the Model and the Pioglitazone groups. Enrichment analysis involved multiple inflammatory response processes and chemokine receptor binding functions. TLR4 and CXCL10 in the Toll signaling pathway may play an important role in SRBI as important targets. Pioglitazone may ameliorate SRBI through the PPAR-γ/TLR4/CXCL10 pathway.
Collapse
Affiliation(s)
- Xuan Zhang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030001, China.
| | - Rui Li
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Ming-Yuan Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Wen-Qian Ye
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Jing-Zhen Liang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Wen-Jing Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Fan Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Hong-Ming Ji
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
2
|
Yang JY. miR-574-5p in epigenetic regulation and Toll-like receptor signaling. Cell Commun Signal 2024; 22:567. [PMID: 39593070 PMCID: PMC11600836 DOI: 10.1186/s12964-024-01934-x] [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: 09/12/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
miR-574-5p is an unusual microRNA (miRNA) that is often upregulated or downregulated following exposure to irradiation or toxic chemicals; bacterial, parasitic or viral infection; and a variety of other disease conditions. Canonically, miR-574-5p epigenetically regulates the expression of many messenger RNAs (mRNAs) through miRNA-mediated posttranscriptional regulation, thereby affecting cellular physiology or pathophysiology and contributing to the pathogenesis or progression of a variety of diseases. However, recent studies have established that in addition to serving as a fine-tuning repressor of gene expression, miR-574-5p also stimulates gene expression as an endogenous ligand for Toll-like receptor-8/7 (TLR8/7). Indeed, the binding of miR-574-5p to TLR8/7 triggers the TLR signaling pathway, leading to the induction of interferons, inflammatory cytokines and autoimmune signaling. These findings suggest that miR-574-5p is not only an important epigenetic regulator of gene expression, but also an important regulator of immune and inflammatory responses. Abnormal miR-574-5p-TLR8/7 signaling has been shown to be tightly associated with inflammation-related cancers and a number of autoimmune disorders. miR-574-5p can serve as a potential biomarker for many diseases. Most importantly, miR-574-5p is a promising therapeutic target for the treatment or prevention of human disorders, especially infectious diseases, cancers and autoimmune diseases.
Collapse
Affiliation(s)
- James Y Yang
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China.
- Wuhu Hospital of East China Normal University, Wuhu, 241000, Anhui, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China.
| |
Collapse
|
3
|
Yang Z, Ji S, Liu L, Liu S, Wang B, Ma Y, Cao X. Promotion of TLR7-MyD88-dependent inflammation and autoimmunity in mice through stem-loop changes in Lnc-Atg16l1. Nat Commun 2024; 15:10224. [PMID: 39587108 PMCID: PMC11589596 DOI: 10.1038/s41467-024-54674-4] [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: 02/29/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
Uncontrolled TLR signaling can cause inflammatory immunopathology and trigger autoimmune diseases. For example, TLR7 promotes pathogenesis of systemic lupus erythematosus. However, whether RNA structural changes affect nucleic acids-sensing TLRs signaling and impact disease progression is unclear. Here by iCLIP-seq we identify a TLR7-binding long non-coding RNA, Lnc-Atg16l1, and find that it promotes TLR7 and other MyD88-dependent TLRs signaling in various types of immune cells. Depletion of Lnc-Atg16l1 attenuates development of TLR7-linked autoimmune phenotypes in the mouse SLE model. Mechanistically, we find that Lnc-Atg16l1 binds to TLR7 at bases near U84 and MyD88 at bases around A129. The analysis of Lnc-Atg16l1 in situ structures show that it strengthens the interaction between TIR domain of TLR7 and MyD88 through specific stem-loop structure changes as a molecular scaffold after TLR7 activation to promote TLR7 downstream signaling. Therefore, we discover a mechanism for host RNA regulation of innate signaling and autoimmune disease through its structural changes. These findings provide insights into the pro-inflammatory function of self RNA in a structure-dependent manner and suggest a potential target for TLR-related autoimmune disorders.
Collapse
Affiliation(s)
- Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuchen Ji
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lun Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuo Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bingjing Wang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanwu Ma
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuetao Cao
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
- Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China.
| |
Collapse
|
4
|
Park C, Lei Z, Li Y, Ren B, He J, Huang H, Chen F, Li H, Brunner K, Zhu J, Jay SM, Williams B, Chao W, Wu J, Zou L. Extracellular vesicles in sepsis plasma mediate neuronal inflammation in the brain through miRNAs and innate immune signaling. J Neuroinflammation 2024; 21:252. [PMID: 39375720 PMCID: PMC11460013 DOI: 10.1186/s12974-024-03250-0] [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: 07/10/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Neuroinflammation reportedly plays a critical role in the pathogenesis of sepsis-associated encephalopathy (SAE). We previously reported that circulating plasma extracellular vesicles (EVs) from septic mice are proinflammatory. In the current study, we tested the role of sepsis plasma EVs in neuroinflammation. METHODS To track EVs in cells and tissues, HEK293T cell-derived EVs were labeled with the fluorescent dye PKH26. Cecal ligation and puncture (CLP) was conducted to model polymicrobial sepsis in mice. Plasma EVs were isolated by ultracentrifugation and their role in promoting neuronal inflammation was tested following intracerebroventricular (ICV) injection. miRNA inhibitors (anti-miR-146a, -122, -34a, and -145a) were applied to determine the effects of EV cargo miRNAs in the brain. A cytokine array was performed to profile microglia-released protein mediators. TLR7- or MyD88-knockout (KO) mice were utilized to determine the underlying mechanism of EVs-mediated neuroinflammation. RESULTS We observed the uptake of fluorescent PKH26-EVs inside the cell bodies of both microglia and neurons. Sepsis plasma EVs led to a dose-dependent cytokine release in cultured microglia, which was partially attenuated by miRNA inhibitors against the target miRNAs and in TLR7-KO cells. When administered via the ICV, sepsis plasma EVs resulted in a marked increase in the accumulation of innate immune cells, including monocyte and neutrophil and cytokine gene expression, in the brain. Although sepsis plasma EVs had no direct effect on cytokine production or neuronal injury in vitro, the conditioned media (CM) of microglia treated with sepsis plasma EVs induced neuronal cell death as evidenced by increased caspase-3 cleavage and Annexin-V staining. Cytokine arrays and bioinformatics analysis of the microglial CM revealed multiple cytokines/chemokines and other factors functionally linked to leukocyte chemotaxis and migration, TLR signaling, and neuronal death. Moreover, sepsis plasma EV-induced brain inflammation in vivo was significantly dependent on MyD88. CONCLUSIONS Circulating plasma EVs in septic mice cause a microglial proinflammatory response in vitro and a brain innate immune response in vivo, some of which are in part mediated by TLR7 in vitro and MyD88 signaling in vivo. These findings highlight the importance of circulating EVs in brain inflammation during sepsis.
Collapse
Affiliation(s)
- Chanhee Park
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Zhuofan Lei
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yun Li
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Boyang Ren
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Junyun He
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Huang Huang
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Fengqian Chen
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hui Li
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kavitha Brunner
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jing Zhu
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20740, USA
| | - Brittney Williams
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wei Chao
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Junfang Wu
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Center to Advanced Chronic Pain Research, University of Maryland, Baltimore, MD, 21201, USA.
| | - Lin Zou
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
5
|
Xiao P, Wen Y, Du G, Luo E, Su Z, Liao Z, Ding H, Li W. Clusterin attenuates blood-brain barrier damage and cognitive impairment by inhibiting astrocyte aging in mice with sepsis-associated encephalopathy. Neuroreport 2024; 35:857-867. [PMID: 38973492 DOI: 10.1097/wnr.0000000000002075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Sepsis-associated encephalopathy (SAE) is a severe complication of sepsis, however, its exact mechanism remains unknown. This study aimed to evaluate whether clusterin is essential to the development of SAE during the aging process of astrocytes. In the study, septic mice were established with cecal ligation and puncture (CLP) and lipopolysaccharides were applied to astrocytes in vitro. Evan's blue dye was used in vivo to show blood-brain barrier (BBB) permeability. A morris water maze test was conducted to assess cognitive functions of the mice. Clusterin-knockout mice were used to examine the effect of clusterin on sepsis. The astrocytes were transfected with lentivirus expressing clusterin cDNA for clusterin overexpression or pYr-LV-clusterin small hairpin RNA for clusterin knockdown in vitro . The expression of clusterin, p-p53, p21, GDNF, and iNOS was detected. he CLP mice exhibited a higher clusterin expression in hippocampus tissue, aging astrocytes, lower GDNF expression and higher iNOS expression, accompanied with BBB damage and cognitive deficiency. Following clusterin knockout, this pathological process was further enhanced. In vitro , following lipopolysaccharides treatment, astrocytes exhibited increased clusterin, p-p53, p21, iNOS and decreased GDNF. Following clusterin knockdown, the cells exhibited a further increase in p-p53, p21, and iNOS and decrease in GDNF. Clusterin overexpression, however, helped inhibit astrocytes aging and neuroinflammation evidenced by decreased p-p53, p21, iNOS and increased GDNF. The present study has revealed that clusterin may exert its neuroprotective effect by preventing aging in astrocytes, suppressing the secretion of iNOS and promoting GNDF release.
Collapse
Affiliation(s)
- Ping Xiao
- Surgical Department of Pulmonary Oncology
| | - Yin Wen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou
| | - Guoqiang Du
- Department of Emergency Medicine, Luoding People's Hospital, Yunfu
| | - Ensi Luo
- Department of Endocrinology, Binhaiwan Central Hospital of Dongguan, Dongguan Hospital Affiliated to Medical College of Jinan University, Dongguan
| | - ZhiWei Su
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou
| | - Zhong Liao
- Department of Emergency Medicine, Longnan First People's Hospital, Longnan
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Weifeng Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Chen O, Jiang C, Berta T, Gray B, Furutani K, Sullenger BA, Ji RR. MicroRNA let-7b enhances spinal cord nociceptive synaptic transmission and induces acute and persistent pain through neuronal and microglial signaling. Pain 2024; 165:1824-1839. [PMID: 38452223 PMCID: PMC11257826 DOI: 10.1097/j.pain.0000000000003206] [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: 08/14/2023] [Accepted: 01/02/2024] [Indexed: 03/09/2024]
Abstract
ABSTRACT Secreted microRNAs (miRNAs) have been detected in various body fluids including the cerebrospinal fluid, yet their direct role in regulating synaptic transmission remains uncertain. We found that intrathecal injection of low dose of let-7b (1 μg) induced short-term (<24 hours) mechanical allodynia and heat hyperalgesia, a response that is compromised in Tlr7-/- or Trpa1-/- mice. Ex vivo and in vivo calcium imaging in GCaMP6-report mice revealed increased calcium signal in spinal cord afferent terminals and doral root ganglion/dorsal root ganglia neurons following spinal perfusion and intraplantar injection of let-7b. Patch-clamp recordings also demonstrated enhanced excitatory synaptic transmission (miniature excitatory postsynaptic currents [EPSCs]) in spinal nociceptive neurons following let-7b perfusion or optogenetic activation of axonal terminals. The elevation in spinal calcium signaling and EPSCs was dependent on the presence of toll-like receptor-7 (TLR7) and transient receptor potential ion channel subtype A1 (TRPA1). In addition, endogenous let-7b is enriched in spinal cord synaptosome, and peripheral inflammation increased let-7b in doral root ganglion/dorsal root ganglia neurons, spinal cord tissue, and the cerebrospinal fluid. Notably, let-7b antagomir inhibited inflammatory pain and inflammation-induced synaptic plasticity (EPSC increase), suggesting an endogenous role of let-7b in regulating pain and synaptic transmission. Furthermore, intrathecal injection of let-7b, at a higher dose (10 μg), induced persistent mechanical allodynia for >2 weeks, which was abolished in Tlr7-/- mice. The high dose of let-7b also induced microgliosis in the spinal cord. Of interest, intrathecal minocycline only inhibited let-7b-induced mechanical allodynia in male but not female mice. Our findings indicate that the secreted microRNA let-7b has the capacity to provoke pain through both neuronal and glial signaling, thereby establishing miRNA as an emerging neuromodulator.
Collapse
Affiliation(s)
- Ouyang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Temugin Berta
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, OH 45267, USA
| | - Bethany Gray
- Department of Surgery, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Kenta Furutani
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Bruce A. Sullenger
- Department of Surgery, Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, NC 27710, USA
| |
Collapse
|
7
|
Maalouly G, Martin CMA, Baz Y, Saliba Y, Baramili AM, Fares N. Antioxidant and Anti-Apoptotic Neuroprotective Effects of Cinnamon in Imiquimod-Induced Lupus. Antioxidants (Basel) 2024; 13:880. [PMID: 39061948 PMCID: PMC11274315 DOI: 10.3390/antiox13070880] [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: 05/02/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Despite accumulating evidence correlating oxidative stress with lupus disease activity, the brain redox pathways are still poorly investigated. Cinnamomum cassia, a widely used spice with powerful antioxidant properties, could be a novel therapeutic candidate in lupus. METHODS C57BL/6J female mice were divided into five groups: sham, sham-cinnamon, lupus, lupus-cinnamon starting from induction, and lupus-cinnamon starting two weeks before induction. Lupus was induced by skin application on the right ear with 1.25 mg of 5% imiquimod cream three times per week for six weeks. Cinnamomum cassia was given orally, five days per week, at 200 mg/kg. RESULTS Concomitant to TLR7-MYD88 pathway activation, the p-NRF2/NRF2 and p-FOXO3/FOXO3 ratios were increased in the hippocampus and alleviated by cinnamon treatment. BCL-2 positivity was enhanced in hippocampal neurons and reversed only by preventive cinnamon administration. In vitro, exposure of hippocampal cells to the plasma of different groups induced a surge in oxidative stress. This was associated with an increased t-BID/BID ratio. Cinnamon treatment, particularly in the preventive arm, normalized these modifications. CONCLUSIONS Our study shows a neuroprotective effect of cinnamon by rescuing brain redox and apoptosis homeostasis in lupus, paving the way for its use as a natural therapeutic compound in the clinical management of lupus.
Collapse
Affiliation(s)
| | | | | | | | | | - Nassim Fares
- Laboratory of Research in Physiology and Pathophysiology, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University of Beirut, Beirut 1104 2020, Lebanon; (G.M.); (C.-M.-A.M.); (Y.B.); (Y.S.); (A.-M.B.)
| |
Collapse
|
8
|
Chen YC, Chen JH, Tsai CF, Wu CY, Chang CN, Wu CT, Yeh WL. Protective effects of paeonol against cognitive impairment in lung diseases. J Pharmacol Sci 2024; 155:101-112. [PMID: 38797534 DOI: 10.1016/j.jphs.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
Pulmonary inflammation may lead to neuroinflammation resulting in neurological dysfunction, and it is associated with a variety of acute and chronic lung diseases. Paeonol is a herbal phenolic compound with anti-inflammatory and anti-oxidative properties. The aim of this study is to understand the beneficial effects of paeonol on cognitive impairment, pulmonary inflammation and its underlying mechanisms. Pulmonary inflammation-associated cognitive deficit was observed in TNFα-stimulated mice, and paeonol mitigated the cognitive impairment by reducing the expressions of interleukin (IL)-1β, IL-6, and NOD-like receptor family pyrin domain-containing 3 (NLRP3) in hippocampus. Moreover, elevated plasma miR-34c-5p in lung-inflamed mice was also reduced by paeonol. Pulmonary inflammation induced by intratracheal instillation of TNFα in mice resulted in immune cells infiltration in bronchoalveolar lavage fluid, pulmonary edema, and acute fibrosis, and these inflammatory responses were alleviated by paeonol orally. In MH-S alveolar macrophages, tumor necrosis factor (TNF) α- and phorbol myristate acetate (PMA)-induced inflammasome activation was ameliorated by paeonol. In addition, the expressions of antioxidants were elevated by paeonol, and reactive oxygen species production was reduced. In this study, paeonol demonstrates protective effects against cognitive deficits and pulmonary inflammation by exerting anti-inflammatory and anti-oxidative properties, suggesting a powerful benefit as a potential therapeutic agent.
Collapse
Affiliation(s)
- Yen-Chang Chen
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 88, Sec. 1, Fengxing Road, Taichung, 427213, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, No.500 Lioufeng Road, Taichung, 413305, Taiwan
| | - Chen-Yun Wu
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Chen-Ni Chang
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, No. 2, Yude Road, Taichung, 404332, Taiwan
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan; Department of Biochemistry, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan.
| |
Collapse
|
9
|
Allard RL, Mayfield J, Barchiesi R, Salem NA, Mayfield RD. Toll-like receptor 7: A novel neuroimmune target to reduce excessive alcohol consumption. Neurobiol Stress 2024; 31:100639. [PMID: 38765062 PMCID: PMC11101708 DOI: 10.1016/j.ynstr.2024.100639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Toll-like receptors (TLRs) are a family of innate immune receptors that recognize molecular patterns in foreign pathogens and intrinsic danger/damage signals from cells. TLR7 is a nucleic acid sensing endosomal TLR that is activated by single-stranded RNAs from microbes or by small noncoding RNAs that act as endogenous ligands. TLR7 signals through the MyD88 adaptor protein and activates the transcription factor interferon regulatory factor 7 (IRF7). TLR7 is found throughout the brain and is highly expressed in microglia, the main immune cells of the brain that have also been implicated in alcohol drinking in mice. Upregulation of TLR7 mRNA and protein has been identified in postmortem hippocampus and cortex from AUD subjects that correlated positively with lifetime consumption of alcohol. Similarly, Tlr7 and downstream signaling genes were upregulated in rat hippocampal and cortical slice cultures after chronic alcohol exposure and in these regions after chronic binge-like alcohol treatment in mice. In addition, repeated administration of the synthetic TLR7 agonists imiquimod (R837) or resiquimod (R848) increased voluntary alcohol drinking in different rodent models and produced sustained upregulation of IRF7 in the brain. These findings suggest that chronic TLR7 activation may drive excessive alcohol drinking. In the brain, this could occur through increased levels of endogenous TLR7 activators, like microRNAs and Y RNAs. This review explores chronic TLR7 activation as a pathway of dysregulated neuroimmune signaling in AUD and the endogenous small RNA ligands in the brain that could perpetuate innate immune responses and escalate alcohol drinking.
Collapse
Affiliation(s)
- Ruth L. Allard
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jody Mayfield
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Riccardo Barchiesi
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nihal A. Salem
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol and Addiction Research and The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
10
|
Pawar K, Kawamura T, Kirino Y. The tRNA Val half: A strong endogenous Toll-like receptor 7 ligand with a 5'-terminal universal sequence signature. Proc Natl Acad Sci U S A 2024; 121:e2319569121. [PMID: 38683985 PMCID: PMC11087793 DOI: 10.1073/pnas.2319569121] [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: 11/07/2023] [Accepted: 03/24/2024] [Indexed: 05/02/2024] Open
Abstract
Toll-like receptors (TLRs) are crucial components of the innate immune system. Endosomal TLR7 recognizes single-stranded RNAs, yet its endogenous ssRNA ligands are not fully understood. We previously showed that extracellular (ex-) 5'-half molecules of tRNAHisGUG (the 5'-tRNAHisGUG half) in extracellular vesicles (EVs) of human macrophages activate TLR7 when delivered into endosomes of recipient macrophages. Here, we fully explored immunostimulatory ex-5'-tRNA half molecules and identified the 5'-tRNAValCAC/AAC half, the most abundant tRNA-derived RNA in macrophage EVs, as another 5'-tRNA half molecule with strong TLR7 activation capacity. Levels of the ex-5'-tRNAValCAC/AAC half were highly up-regulated in macrophage EVs upon exposure to lipopolysaccharide and in the plasma of patients infected with Mycobacterium tuberculosis. The 5'-tRNAValCAC/AAC half-mediated activation of TLR7 effectively eradicated bacteria infected in macrophages. Mutation analyses of the 5'-tRNAValCAC/AAC half identified the terminal GUUU sequence as a determinant for TLR7 activation. We confirmed that GUUU is the optimal ratio of guanosine and uridine for TLR7 activation; microRNAs or other RNAs with the terminal GUUU motif can indeed stimulate TLR7, establishing the motif as a universal signature for TLR7 activation. These results advance our understanding of endogenous ssRNA ligands of TLR7 and offer insights into diverse TLR7-involved pathologies and their therapeutic strategies.
Collapse
Affiliation(s)
- Kamlesh Pawar
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
- Department of Life Sciences, School of Natural Science, Shiv Nadar Institution of Eminence Deemed to be University, Delhi National Capital Region, Greater Noida201314, India
| | - Takuya Kawamura
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| | - Yohei Kirino
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| |
Collapse
|
11
|
Huang Y, Xu R, Liu Q, Zhang X, Mao Y, Yang Y, Gu X, Liu Y, Ma Z. Glucose competition between endothelial cells in the blood-spinal cord barrier and infiltrating regulatory T cells is linked to sleep restriction-induced hyperalgesia. BMC Med 2024; 22:189. [PMID: 38715017 PMCID: PMC11077863 DOI: 10.1186/s12916-024-03413-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Sleep loss is a common public health problem that causes hyperalgesia, especially that after surgery, which reduces the quality of life seriously. METHODS The 48-h sleep restriction (SR) mouse model was created using restriction chambers. In vivo imaging, transmission electron microscopy (TEM), immunofluorescence staining and Western blot were performed to detect the status of the blood-spinal cord barrier (BSCB). Paw withdrawal mechanical threshold (PWMT) was measured to track mouse pain behavior. The role of infiltrating regulatory T cells (Tregs) and endothelial cells (ECs) in mouse glycolysis and BSCB damage were analyzed using flow cytometry, Western blot, CCK-8 assay, colorimetric method and lactate administration. RESULTS The 48-h SR made mice in sleep disruption status and caused an acute damage to the BSCB, resulting in hyperalgesia and neuroinflammation in the spinal cord. In SR mice, the levels of glycolysis and glycolysis enzymes of ECs in the BSCB were found significantly decreased [CON group vs. SR group: CD31+Glut1+ cells: p < 0.001], which could cause dysfunction of ECs and this was confirmed in vitro. Increased numbers of infiltrating T cells [p < 0.0001] and Treg population [p < 0.05] were detected in the mouse spinal cord after 48-h SR. In the co-cultured system of ECs and Tregs in vitro, the competition of Tregs for glucose resulted in the glycolysis disorder of ECs [Glut1: p < 0.01, ENO1: p < 0.05, LDHα: p < 0.05; complete tubular structures formed: p < 0.0001; CCK8 assay: p < 0.001 on 24h, p < 0.0001 on 48h; glycolysis level: p < 0.0001]. An administration of sodium lactate partially rescued the function of ECs and relieved SR-induced hyperalgesia. Furthermore, the mTOR signaling pathway was excessively activated in ECs after SR in vivo and those under the inhibition of glycolysis or co-cultured with Tregs in vitro. CONCLUSIONS Affected by glycolysis disorders of ECs due to glucose competition with infiltrating Tregs through regulating the mTOR signaling pathway, hyperalgesia induced by 48-h SR is attributed to neuroinflammation and damages to the barriers, which can be relieved by lactate supplementation.
Collapse
Affiliation(s)
- Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Qi Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Xiao Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Yanting Mao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Yan Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China.
| | - Yue Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital Medical School, Nanjing University, No. 321 of Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
12
|
Dorrity TJ, Shin H, Gertie JA, Chung H. The Sixth Sense: Self-nucleic acid sensing in the brain. Adv Immunol 2024; 161:53-83. [PMID: 38763702 PMCID: PMC11186578 DOI: 10.1016/bs.ai.2024.03.001] [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: 05/21/2024]
Abstract
Our innate immune system uses pattern recognition receptors (PRRs) as a first line of defense to detect microbial ligands and initiate an immune response. Viral nucleic acids are key ligands for the activation of many PRRs and the induction of downstream inflammatory and antiviral effects. Initially it was thought that endogenous (self) nucleic acids rarely activated these PRRs, however emerging evidence indicates that endogenous nucleic acids are able to activate host PRRs in homeostasis and disease. In fact, many regulatory mechanisms are in place to finely control and regulate sensing of self-nucleic acids by PRRs. Sensing of self-nucleic acids is particularly important in the brain, as perturbations to nucleic acid sensing commonly leads to neuropathology. This review will highlight the role of nucleic acid sensors in the brain, both in disease and homeostasis. We also indicate the source of endogenous stimulatory nucleic acids where known and summarize future directions for the study of this growing field.
Collapse
Key Words
- Brain
- DNA sensing PRRs: cGAS, AIM2, TLR9
- Neurodegeneration: Aicardi-Goutieres syndrome (AGS), Alzheimer's disease, Amyotrophic lateral sclerosis, Stroke, Traumatic brain injury
- Neurodevelopment
- Neuroinflammation
- Nuecleic acid immunity
- Pattern recognition receptors (PRRs)
- RNA sensing PRRs: MDA5, RIG-I, PKR, TLR3, TLR7/8
Collapse
Affiliation(s)
- Tyler J Dorrity
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, United States
| | - Heegwon Shin
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, United States
| | - Jake A Gertie
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, United States; Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States; Medical Scientist Training Program, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Hachung Chung
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, United States.
| |
Collapse
|
13
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [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: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Williams B, Zou L, Pittet JF, Chao W. Sepsis-Induced Coagulopathy: A Comprehensive Narrative Review of Pathophysiology, Clinical Presentation, Diagnosis, and Management Strategies. Anesth Analg 2024; 138:696-711. [PMID: 38324297 PMCID: PMC10916756 DOI: 10.1213/ane.0000000000006888] [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] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Physiological hemostasis is a balance between pro- and anticoagulant pathways, and in sepsis, this equilibrium is disturbed, resulting in systemic thrombin generation, impaired anticoagulant activity, and suppression of fibrinolysis, a condition termed sepsis-induced coagulopathy (SIC). SIC is a common complication, being present in 24% of patients with sepsis and 66% of patients with septic shock, and is often associated with poor clinical outcomes and high mortality. 1 , 2 Recent preclinical and clinical studies have generated new insights into the molecular pathogenesis of SIC. In this article, we analyze the complex pathophysiology of SIC with a focus on the role of procoagulant innate immune signaling in hemostatic activation--tissue factor production, thrombin generation, endotheliopathy, and impaired antithrombotic functions. We also review clinical presentations of SIC, the diagnostic scoring system and laboratory tests, the current standard of care, and clinical trials evaluating the efficacies of anticoagulant therapies.
Collapse
Affiliation(s)
- Brittney Williams
- From the Division of Cardiothoracic Anesthesia, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Jean-Francois Pittet
- Division of Critical Care, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
15
|
Gumas J, Kawamura T, Shigematsu M, Kirino Y. Immunostimulatory short non-coding RNAs in the circulation of patients with tuberculosis infection. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102156. [PMID: 38481936 PMCID: PMC10933579 DOI: 10.1016/j.omtn.2024.102156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/14/2024] [Indexed: 03/20/2024]
Abstract
Mycobacterium tuberculosis (Mtb) infection is among the world's deadliest infectious diseases. Developing effective treatments and biomarkers for tuberculosis requires a deeper understanding of its pathobiology and host responses. Here, we report a comprehensive characterization of circulating short non-coding RNAs (sncRNAs) in plasma samples from Mtb-infected patients. We achieved this by pre-treating plasma RNAs with T4 polynucleotide kinase to convert all RNA ends to those compatible with sncRNA sequencing. We discovered a global and drastic upregulation of plasma sncRNAs in Mtb-infected patients, with tRNA-derived sncRNAs representing the most dramatically elevated class. Most of these tRNA-derived sncRNAs originated from a limited subset of tRNAs, specifically from three tRNA isoacceptors, and exhibited skewed patterns to 5'-derived fragments, such as 5' halves, 5' tRNA fragments (tRFs), and internal tRFs (i-tRFs) from the 5' regions. Further, Mtb-infected patients displayed markedly upregulated and distinct profiles of both rRNA- and mRNA-derived sncRNAs. Some of these sncRNAs, which are abundant and specific to Mtb-infected patients, robustly activated human macrophages via Toll-like receptor 7 and induced cytokine production. This drastic accumulation of circulating, immunostimulatory sncRNAs in the plasma of Mtb-infected patients offers insights into the sncRNA-driven aspects of host immune response against infectious diseases and suggests a pool of potential therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Justin Gumas
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Takuya Kawamura
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Megumi Shigematsu
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yohei Kirino
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
16
|
Qin N, Miao Y, Xie L, Ma X, Xie P. Sepsis-associated encephalopathy: Autophagy and miRNAs regulate microglial activation. Physiol Rep 2024; 12:e15964. [PMID: 38439741 PMCID: PMC10912956 DOI: 10.14814/phy2.15964] [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: 11/14/2023] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Sepsis-associated encephalopathy (SAE) describes diffuse or multifocal cerebral dysfunction caused by the systemic inflammatory response to sepsis. SAE is a common neurological complication in patients in the middle and late stages of sepsis in the intensive care unit. Microglia, resident macrophages of the central nervous system, phagocytose small numbers of neuronal cells and apoptotic cells, among other cells, to maintain the dynamic balance of the brain's internal environment. The neuroinflammatory response induced by activated microglia plays a central role in the pathogenesis of various central nervous system diseases. In this paper, we systematically describe the functions and phenotypes of microglia, summarize how microglia mediate neuroinflammation and contribute to the occurrence and development of SAE, and discuss recent progress in autophagy- and microRNA-mediated regulation of microglial activation to provide a theoretical basis for the prevention and treatment of SAE and identify related therapeutic targets.
Collapse
Affiliation(s)
- Nannan Qin
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Yanmei Miao
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Leiyu Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Xinglong Ma
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Peng Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| |
Collapse
|
17
|
Zhang J, Chen S, Hu X, Huang L, Loh P, Yuan X, Liu Z, Lian J, Geng L, Chen Z, Guo Y, Chen B. The role of the peripheral system dysfunction in the pathogenesis of sepsis-associated encephalopathy. Front Microbiol 2024; 15:1337994. [PMID: 38298892 PMCID: PMC10828041 DOI: 10.3389/fmicb.2024.1337994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Sepsis is a condition that greatly impacts the brain, leading to neurological dysfunction and heightened mortality rates, making it one of the primary organs affected. Injury to the central nervous system can be attributed to dysfunction of various organs throughout the entire body and imbalances within the peripheral immune system. Furthermore, central nervous system injury can create a vicious circle with infection-induced peripheral immune disorders. We collate the pathogenesis of septic encephalopathy, which involves microglial activation, programmed cell death, mitochondrial dysfunction, endoplasmic reticulum stress, neurotransmitter imbalance, and blood-brain barrier disruption. We also spotlight the effects of intestinal flora and its metabolites, enterocyte-derived exosomes, cholinergic anti-inflammatory pathway, peripheral T cells and their cytokines on septic encephalopathy.
Collapse
Affiliation(s)
- Jingyu Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuangli Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiyou Hu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lihong Huang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - PeiYong Loh
- School of International Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinru Yuan
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinyu Lian
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lianqi Geng
- Binhai New Area Hospital of TCM, Fourth Teaching Hospital of Tianjin University of TCM, Tianjin, China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture and Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bo Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Binhai New Area Hospital of TCM, Fourth Teaching Hospital of Tianjin University of TCM, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture and Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
18
|
Liu H, Tan S, Han S, Liu X, Li Z, Wang N, Wu Z, Ma J, Shi K, Wang W, Sha Z. Effects of miR-722 on gene expression and alternative splicing in the liver of half-smooth tongue sole after infection with Vibrio anguillarum. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109275. [PMID: 38081443 DOI: 10.1016/j.fsi.2023.109275] [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: 10/12/2023] [Revised: 11/22/2023] [Accepted: 12/04/2023] [Indexed: 12/31/2023]
Abstract
MicroRNAs play crucial roles in various biological processes, including but not limited to differentiation, development, disease, and immunity. However, their immunoregulatory roles in half-smooth tongue sole are lacking. Our previous studies indicated that miR-722 could target C5aR1 to modulate the complement pathway to alleviate inflammatory response and even affect the mortality after the bacterial infection with Vibrio anguillarum. Driven by the purpose of revealing the underlying mechanisms, in this study, we investigated the effects of miR-722 on the gene expression and alternative splicing (AS) in the liver of half-smooth tongue sole after Vibrio anguillarum infection, with the approach of miR-722 overexpression/silencing and subsequent RNA-seq. Among the different comparisons, the I group (miR-722 inhibitor and V. anguillarum) versus blank control (PBS) exhibited the highest number of differentially expressed genes (DEGs), suggesting that the immune response was overactivated after inhibiting the miR-722. In addition, enrichment analyses were performed to reveal the functions of DEGs and differential AS (DAS) genes, reflecting the enrichment of RNA splicing and immune-related pathways including NF-κB and T cell receptor signaling pathway. Comparing the M group (miR-722 mimic and V. anguillarum) with the negative control (random sequence and V. anguillarum), two immune-related genes, cd48 and mapk8, were differentially expressed, of which mapk8 was also differentially spliced, indicating their importance in the immune response. Furthermore, representative gene analysis was performed, suggesting their corresponding functional changes due to AS. To verify the RNA-seq data, quantitative real-time PCR was employed with twenty pairs of primers for DEGs and DAS events. Overall, our results demonstrated that miR-722 could mediate the transcriptome-wide changes of gene expression and AS in half-smooth tongue sole, and provided insights into the regulatory role of miR-722 in immune responses, laying the foundation for further functional analyses and practical applications in aquaculture.
Collapse
Affiliation(s)
- Hongning Liu
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Suxu Tan
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Sen Han
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Xinbao Liu
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Zhujun Li
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Ningning Wang
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China; College of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Zhendong Wu
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Jie Ma
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Kunpeng Shi
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Wenwen Wang
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China
| | - Zhenxia Sha
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao, 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
19
|
Wang J, Tian F, Cao L, Du R, Tong J, Ding X, Yuan Y, Wang C. Macrophage polarization in spinal cord injury repair and the possible role of microRNAs: A review. Heliyon 2023; 9:e22914. [PMID: 38125535 PMCID: PMC10731087 DOI: 10.1016/j.heliyon.2023.e22914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The prevention, treatment, and rehabilitation of spinal cord injury (SCI) have always posed significant medical challenges. After mechanical injury, disturbances in microcirculation, edema formation, and the generation of free radicals lead to additional damage, impeding effective repair processes and potentially exacerbating further dysfunction. In this context, inflammatory responses, especially the activation of macrophages, play a pivotal role. Different phenotypes of macrophages have distinct effects on inflammation. Activation of classical macrophage cells (M1) promotes inflammation, while activation of alternative macrophage cells (M2) inhibits inflammation. The polarization of macrophages is crucial for disease healing. A non-coding RNA, known as microRNA (miRNA), governs the polarization of macrophages, thereby reducing inflammation following SCI and facilitating functional recovery. This study elucidates the inflammatory response to SCI, focusing on the infiltration of immune cells, specifically macrophages. It examines their phenotype and provides an explanation of their polarization mechanisms. Finally, this paper introduces several well-known miRNAs that contribute to macrophage polarization following SCI, including miR-155, miR-130a, and miR-27 for M1 polarization, as well as miR-22, miR-146a, miR-21, miR-124, miR-223, miR-93, miR-132, and miR-34a for M2 polarization. The emphasis is placed on their potential therapeutic role in SCI by modulating macrophage polarization, as well as the present developments and obstacles of miRNA clinical therapy.
Collapse
Affiliation(s)
- Jiawei Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Feng Tian
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Lili Cao
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Ruochen Du
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Jiahui Tong
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Xueting Ding
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Yitong Yuan
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Chunfang Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| |
Collapse
|
20
|
Liu H, Tan S, Chen Y, Chen X, Liu X, Li Z, Wang N, Han S, Wu Z, Ma J, Shi K, Wang W, Sha Z. Regulatory mechanism of miR-722 on C5aR1 and its functions against bacterial inflammation in half-smooth tongue sole (Cynoglossus semilaevis). Int J Biol Macromol 2023; 252:126445. [PMID: 37611685 DOI: 10.1016/j.ijbiomac.2023.126445] [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: 02/17/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/25/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs involved in various biological processes, including immunity. Previously, we investigated the miRNAs of half-smooth tongue sole (Cynoglossus semilaevis) and found that miR-722 (designated Cse-miR-722) was significantly differentially expressed after infection with Vibrio anguillarum, reflecting its importance in immune response. Our preliminary bioinformatic analysis suggested that Cse-miR-722 could target C5aR1 (designated CsC5aR1), which was known to play crucial roles in complement activation and inflammatory response, as a receptor of C5a. However, the underlying mechanisms of their interactions and specific functions in inflammatory and immune response are still enigmas. In this study, we successfully cloned the precursor sequence of Cse-miR-722 (94 bp) and the full length of CsC5aR1 (1541 bp, protein molecular weight 39 kDa). The target gene of Cse-miR-722 was verified as CsC5aR1 by a dual luciferase reporter assay, and Cse-miR-722 was confirmed to regulate CsC5aR1 at the protein level using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The expression of CsC5aR1 and Cse-miR-722 in liver cells and four immune tissues of half-smooth tongue sole changed significantly after LPS stimulation and infection with V. anguillarum. To explore the functional role of Cse-miR-722 in half-smooth tongue sole, we performed both in vitro and in vivo experiments. Cse-miR-722 was observed to affect phagocytosis and respiratory burst activity of macrophages by regulating CsC5aR1 in half-smooth tongue sole. Furthermore, we found that Cse-miR-722 regulated the expression of CsC5aR1, CsC5a, and the inflammatory factors CsIL1-β, CsIL6, CsIL8, and CsTNF-α both in vitro and in vivo. In addition, Cse-miR-722 reduced mortality and pathological damage. This study clarified the regulatory mechanism of Cse-miR-722 on CsC5aR1 and provided insight into the regulatory roles of Cse-miR-722 in immune responses, laying a theoretical foundation for the feasibility of using miR-722 to prevent and control bacterial diseases in teleost.
Collapse
Affiliation(s)
- Hongning Liu
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Suxu Tan
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Yadong Chen
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Xuejie Chen
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Xinbao Liu
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Zhujun Li
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Ningning Wang
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China; College of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Sen Han
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Zhendong Wu
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Jie Ma
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Kunpeng Shi
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Wenwen Wang
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Zhenxia Sha
- Institute of Aquatic Biotechnology, College of Life Sciences, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
21
|
Suen AO, Chen F, Wang S, Li Z, Zhu J, Yang Y, Conn O, Lopez K, Cui P, Wechsler L, Cross A, Fiskum G, Kozar R, Hu P, Miller C, Zou L, Williams B, Chao W. Extracellular RNA Sensing Mediates Inflammation and Organ Injury in a Murine Model of Polytrauma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1990-2000. [PMID: 37133342 PMCID: PMC10235856 DOI: 10.4049/jimmunol.2300103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/06/2023] [Indexed: 05/04/2023]
Abstract
Severe traumatic injury leads to marked systemic inflammation and multiorgan injury. Endogenous drivers such as extracellular nucleic acid may play a role in mediating innate immune response and the downstream pathogenesis. Here, we explored the role of plasma extracellular RNA (exRNA) and its sensing mechanism in inflammation and organ injury in a murine model of polytrauma. We found that severe polytrauma-bone fracture, muscle crush injury, and bowel ischemia-induced a marked increase in plasma exRNA, systemic inflammation, and multiorgan injury in mice. Plasma RNA profiling with RNA sequencing in mice and humans revealed a dominant presence of miRNAs and marked differential expression of numerous miRNAs after severe trauma. Plasma exRNA isolated from trauma mice induced a dose-dependent cytokine production in macrophages, which was almost abolished in TLR7-deficient cells but unchanged in TLR3-deficient cells. Moreover, RNase or specific miRNA inhibitors against the selected proinflammatory miRNAs (i.e., miR-7a-5p, miR-142, let-7j, miR-802, and miR-146a-5p) abolished or attenuated trauma plasma exRNA-induced cytokine production, respectively. Bioinformatic analyses of a group of miRNAs based on cytokine readouts revealed that high uridine abundance (>40%) is a reliable predictor in miRNA mimic-induced cytokine and complement production. Finally, compared with the wild-type, TLR7-knockout mice had attenuated plasma cytokine storm and reduced lung and hepatic injury after polytrauma. These data suggest that endogenous plasma exRNA of severely injured mice and ex-miRNAs with high uridine abundance prove to be highly proinflammatory. TLR7 sensing of plasma exRNA and ex-miRNAs activates innate immune responses and plays a role in inflammation and organ injury after trauma.
Collapse
Affiliation(s)
- Andrew O. Suen
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
- Department of Anesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Fengqian Chen
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Sheng Wang
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Zhu
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Yang Yang
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Olivia Conn
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Kerri Lopez
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Ping Cui
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Laurence Wechsler
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Alan Cross
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Gary Fiskum
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Rosemary Kozar
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Peter Hu
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Catriona Miller
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
- Enroute Care Division, Department of Aeromedical Research, U.S. Air Force School of Aerospace Medicine, Wright Patterson Air Force Base, Dayton, OH
| | - Lin Zou
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Brittney Williams
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| | - Wei Chao
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
22
|
Lajqi T, Köstlin-Gille N, Bauer R, Zarogiannis SG, Lajqi E, Ajeti V, Dietz S, Kranig SA, Rühle J, Demaj A, Hebel J, Bartosova M, Frommhold D, Hudalla H, Gille C. Training vs. Tolerance: The Yin/Yang of the Innate Immune System. Biomedicines 2023; 11:766. [PMID: 36979747 PMCID: PMC10045728 DOI: 10.3390/biomedicines11030766] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
For almost nearly a century, memory functions have been attributed only to acquired immune cells. Lately, this paradigm has been challenged by an increasing number of studies revealing that innate immune cells are capable of exhibiting memory-like features resulting in increased responsiveness to subsequent challenges, a process known as trained immunity (known also as innate memory). In contrast, the refractory state of endotoxin tolerance has been defined as an immunosuppressive state of myeloid cells portrayed by a significant reduction in the inflammatory capacity. Both training as well tolerance as adaptive features are reported to be accompanied by epigenetic and metabolic alterations occurring in cells. While training conveys proper protection against secondary infections, the induction of endotoxin tolerance promotes repairing mechanisms in the cells. Consequently, the inappropriate induction of these adaptive cues may trigger maladaptive effects, promoting an increased susceptibility to secondary infections-tolerance, or contribute to the progression of the inflammatory disorder-trained immunity. This review aims at the discussion of these opposing manners of innate immune and non-immune cells, describing the molecular, metabolic and epigenetic mechanisms involved and interpreting the clinical implications in various inflammatory pathologies.
Collapse
Affiliation(s)
- Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Jena University Hospital, D-07745 Jena, Germany
| | - Sotirios G. Zarogiannis
- Department of Physiology, School of Health Sciences, Faculty of Medicine, University of Thessaly, GR-41500 Larissa, Greece
| | - Esra Lajqi
- Department of Radiation Oncology, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Valdrina Ajeti
- Department of Pharmacy, Alma Mater Europaea—Campus College Rezonanca, XK-10000 Pristina, Kosovo
| | - Stefanie Dietz
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Simon A. Kranig
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| | - Jessica Rühle
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Ardian Demaj
- Faculty of Medical Sciences, University of Tetovo, MK-1200 Tetova, North Macedonia
| | - Janine Hebel
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine Heidelberg, University of Heidelberg, D-69120 Heidelberg, Germany
| | - David Frommhold
- Klinik für Kinderheilkunde und Jugendmedizin, D-87700 Memmingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| |
Collapse
|
23
|
Dobkin J, Wu L, Mangalmurti NS. The ultimate tradeoff: how red cell adaptations to malaria alter the host response during critical illness. Am J Physiol Lung Cell Mol Physiol 2023; 324:L169-L178. [PMID: 36594846 PMCID: PMC9902222 DOI: 10.1152/ajplung.00127.2022] [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: 04/18/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
The human immune system evolved in response to pathogens. Among these pathogens, malaria has proven to be one of the deadliest and has exerted the most potent selective pressures on its target cell, the red blood cell. Red blood cells have recently gained recognition for their immunomodulatory properties, yet how red cell adaptations contribute to the host response during critical illness remains understudied. This review will discuss how adaptations that may have been advantageous for host survival might influence immune responses in modern critical illness. We will highlight the current evidence for divergent host resilience arising from the adaptations to malaria and summarize how understanding evolutionary red cell adaptations to malaria may provide insight into the heterogeneity of the host response to critical illness, perhaps driving future precision medicine approaches to syndromes affecting the critically ill such as sepsis and acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Jane Dobkin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ling Wu
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nilam S Mangalmurti
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Brittney W, Kozar R, Wei C. EMERGING ROLE OF EXTRACELLULAR RNA IN INNATE IMMUNITY, SEPSIS, AND TRAUMA. Shock 2023; 59:190-199. [PMID: 36730864 PMCID: PMC9957828 DOI: 10.1097/shk.0000000000002032] [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: 02/04/2023]
Abstract
ABSTRACT Sepsis and trauma remain the leading causes of morbidity and mortality. Our understanding of the molecular pathogenesis in the development of multiple organ dysfunction in sepsis and trauma has evolved as more focus is on secondary injury from innate immunity, inflammation, and the potential role of endogenous danger molecules. Studies of the past several decades have generated evidence for extracellular RNAs (exRNAs) as biologically active mediators in health and disease. Here, we review studies on plasma exRNA profiling in mice and humans with sepsis and trauma, the role and mode of action by exRNAs, such as ex-micro(mi)RNAs, in host innate immune response, and their potential implications in various organ injury during sepsis and trauma.
Collapse
Affiliation(s)
- Williams Brittney
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rosemary Kozar
- Shock Trauma Center and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chao Wei
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
25
|
Williams B, Zhu J, Zou L, Chao W. Innate immune TLR7 signaling mediates platelet activation and platelet-leukocyte aggregate formation in murine bacterial sepsis. Platelets 2022; 33:1251-1259. [PMID: 35920588 PMCID: PMC9833650 DOI: 10.1080/09537104.2022.2107627] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Thrombocytopenia is a common complication in sepsis and is associated with higher mortality. Activated platelets express CD62P, which facilitates platelet-leukocyte aggregate (PLA) formation and contributes to thrombocytopenia in sepsis. We have reported that thrombocytopenia in murine sepsis is partly attributable to TLR7 signaling, but the underlying mechanism is unclear. In the current study, we tested the hypothesis that TLR7 mediates platelet activation and PLA formation during sepsis. In vitro, whole blood from WT mice treated with loxoribine, a TLR7 agonist, exhibited a dose-dependent increase in activated platelets compared to the control (PBS with 0.05% DMSO) or loxoribine-treated TLR7-/- whole blood. In a murine model of sepsis, there was a significant increase in platelet activation and PLA formation 24 hours after cecal ligation and puncture (CLP) as evidenced by double positive expression of CD41+/CD62P+ and CD45+/CD62P+, respectively. The sepsis-induced PLA formation was significantly attenuated in TLR7-/- mice. Finally, in ex-vivo experiments, plasma isolated from septic mice induced WT platelet activation, but such effect was significantly attenuated in platelets deficient of TLR7. These findings demonstrate a pivotal role of TLR7 signaling in platelet activation and PLA formation during bacterial sepsis.
Collapse
|
26
|
Huang H, Zhu J, Gu L, Hu J, Feng X, Huang W, Wang S, Yang Y, Cui P, Lin SH, Suen A, Shimada BK, Williams B, Kane MA, Ke Y, Zhang CO, Birukova AA, Birukov KG, Chao W, Zou L. TLR7 Mediates Acute Respiratory Distress Syndrome in Sepsis by Sensing Extracellular miR-146a. Am J Respir Cell Mol Biol 2022; 67:375-388. [PMID: 35679261 PMCID: PMC9447138 DOI: 10.1165/rcmb.2021-0551oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
TLR7 (Toll-like receptor 7), the sensor for single-stranded RNA, contributes to systemic inflammation and mortality in murine polymicrobial sepsis. Recent studies show that extracellular miR-146a-5p serves as a TLR7 ligand and plays an important role in regulating host innate immunity. However, the role of miR-146a-5p and TLR7 signaling in pulmonary inflammation, endothelial activation, and sepsis-associated acute respiratory distress syndrome remains unclear. Here, we show that intratracheal administration of exogenous miR-146a-5p in mice evokes lung inflammation, activates endothelium, and increases endothelial permeability via TLR7-dependent mechanisms. TLR7 deficiency attenuates pulmonary barrier dysfunction and reduces lung inflammatory response in a murine sepsis model. Moreover, the impact of miR-146a-5p-TLR7 signaling on endothelial activation appears to be a secondary effect because TLR7 is undetectable in the human pulmonary artery and microvascular endothelial cells (ECs), which show no response to direct miR-146a-5p treatment in vitro. Both conditioned media of miR-146a-5p-treated macrophages (Mϕ) and septic sera of wild-type mice induce a marked EC barrier disruption in vitro, whereas Mϕ conditioned media or septic sera of TLR7-/- mice do not exhibit such effect. Cytokine array and pathway enrichment analysis of the Mϕ conditioned media and septic sera identify TNFα (tumor necrosis factor α) as the main downstream effector of miR-146a-5p-TLR7 signaling responsible for the EC barrier dysfunction, which is further supported by neutralizing anti-TNFα antibody intervention. Together, these data demonstrate that TLR7 activation elicits pulmonary inflammation and endothelial barrier disruption by sensing extracellular miR-146a-5p and contributes to sepsis-associated acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Huang Huang
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Jing Zhu
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Lili Gu
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Jiang Hu
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Xiujing Feng
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland
| | - Sheng Wang
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Yang Yang
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Ping Cui
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Shao-Hsuan Lin
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Andrew Suen
- Center for Shock, Trauma, and Anesthesiology Research and
| | | | | | - Maureen A. Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland
| | - Yunbo Ke
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Chen-ou Zhang
- Division of Pulmonary and Critical Care Medicine, School of Medicine, and
| | - Anna A. Birukova
- Division of Pulmonary and Critical Care Medicine, School of Medicine, and
| | - Konstantin G. Birukov
- Center for Shock, Trauma, and Anesthesiology Research and
- Division of Pulmonary and Critical Care Medicine, School of Medicine, and
| | - Wei Chao
- Center for Shock, Trauma, and Anesthesiology Research and
| | - Lin Zou
- Center for Shock, Trauma, and Anesthesiology Research and
| |
Collapse
|
27
|
Chen F, Liu Q, Xiong Y, Xu L. Nucleic acid strategies for infectious disease treatments: The nanoparticle-based oral delivery route. Front Pharmacol 2022; 13:984981. [PMID: 36105233 PMCID: PMC9465296 DOI: 10.3389/fphar.2022.984981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Therapies based on orally administrated nucleic acids have significant potential for the treatment of infectious diseases, including chronic inflammatory diseases such as inflammatory bowel disease (IBD)-associated with the gastrointestinal (GI) tract, and infectious and acute contagious diseases like coronavirus disease 2019 (COVID-19). This is because nucleic acids could precisely regulate susceptibility genes in regulating the pro- and anti-inflammatory cytokines expression related to the infections. Unfortunately, gene delivery remains a major hurdle due to multiple intracellular and extracellular barriers. This review thoroughly discusses the challenges of nanoparticle-based nucleic acid gene deliveries and strategies for overcoming delivery barriers to the inflammatory sites. Oral nucleic acid delivery case studies were also present as vital examples of applications in infectious diseases such as IBD and COVID-19.
Collapse
Affiliation(s)
- Fengqian Chen
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yang Xiong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Xu
- Department of Anorectal Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Li Xu,
| |
Collapse
|
28
|
Expression of MicroRNAs in Sepsis-Related Organ Dysfunction: A Systematic Review. Int J Mol Sci 2022; 23:ijms23169354. [PMID: 36012630 PMCID: PMC9409129 DOI: 10.3390/ijms23169354] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a critical condition characterized by increased levels of pro-inflammatory cytokines and proliferating cells such as neutrophils and macrophages in response to microbial pathogens. Such processes lead to an abnormal inflammatory response and multi-organ failure. MicroRNAs (miRNA) are single-stranded non-coding RNAs with the function of gene regulation. This means that miRNAs are involved in multiple intracellular pathways and thus contribute to or inhibit inflammation. As a result, their variable expression in different tissues and organs may play a key role in regulating the pathophysiological events of sepsis. Thanks to this property, miRNAs may serve as potential diagnostic and prognostic biomarkers in such life-threatening events. In this narrative review, we collect the results of recent studies on the expression of miRNAs in heart, blood, lung, liver, brain, and kidney during sepsis and the molecular processes in which they are involved. In reviewing the literature, we find at least 122 miRNAs and signaling pathways involved in sepsis-related organ dysfunction. This may help clinicians to detect, prevent, and treat sepsis-related organ failures early, although further studies are needed to deepen the knowledge of their potential contribution.
Collapse
|
29
|
Lam LM, Mangalmurti NS. ExRNA Takes a Toll in Sepsis-Associated Lung Injury. Am J Respir Cell Mol Biol 2022; 67:271-272. [PMID: 35728049 PMCID: PMC9447133 DOI: 10.1165/rcmb.2022-0237ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Lk Metthew Lam
- University of Pennsylvania Perelman School of Medicine, 14640, Philadelphia, Pennsylvania, United States
| | - Nilam S Mangalmurti
- University of Pennsylvania Perelman School of Medicine, 14640, Philadelphia, Pennsylvania, United States;
| |
Collapse
|