1
|
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
|
2
|
Panda SP, Singh V. The Dysregulated MAD in Mad: A Neuro-theranostic Approach Through the Induction of Autophagic Biomarkers LC3B-II and ATG. Mol Neurobiol 2023; 60:5214-5236. [PMID: 37273153 DOI: 10.1007/s12035-023-03402-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023]
Abstract
The word mad has historically been associated with the psyche, emotions, and abnormal behavior. Dementia is a common symptom among psychiatric disorders or mad (schizophrenia, depression, bipolar disorder) patients. Autophagy/mitophagy is a protective mechanism used by cells to get rid of dysfunctional cellular organelles or mitochondria. Autophagosome/mitophagosome abundance in autophagy depends on microtubule-associated protein light chain 3B (LC3B-II) and autophagy-triggering gene (ATG) which functions as an autophagic biomarker for phagophore production and quick mRNA disintegration. Defects in either LC3B-II or the ATG lead to dysregulated mitophagy-and-autophagy-linked dementia (MAD). The impaired MAD is closely associated with schizophrenia, depression, and bipolar disorder. The pathomechanism of psychosis is not entirely known, which is the severe limitation of today's antipsychotic drugs. However, the reviewed circuit identifies new insights that may be especially helpful in targeting biomarkers of dementia. Neuro-theranostics can also be achieved by manufacturing either bioengineered bacterial and mammalian cells or nanocarriers (liposomes, polymers, and nanogels) loaded with both imaging and therapeutic materials. The nanocarriers must cross the BBB and should release both diagnostic agents and therapeutic agents in a controlled manner to prove their effectiveness against psychiatric disorders. In this review, we highlighted the potential of microRNAs (miRs) as neuro-theranostics in the treatment of dementia by targeting autophagic biomarkers LC3B-II and ATG. Focus was also placed on the potential for neuro-theranostic nanocells/nanocarriers to traverse the BBB and induce action against psychiatric disorders. The neuro-theranostic approach can provide targeted treatment for mental disorders by creating theranostic nanocarriers.
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Uttar Pradesh, Mathura, India.
| | - Vikrant Singh
- Research Scholar, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh, Mathura, India
| |
Collapse
|
3
|
Wang Y, Dong N, Zhou Y, Li H, Qin G, Li H, Zhao Q, Zhang M, Su Y. Effects of Emodin on Protein Expression Related to Autophagy of Interstitial Cells of Cajal in Diabetic Rats. Chem Pharm Bull (Tokyo) 2023; 71:129-133. [PMID: 36464270 DOI: 10.1248/cpb.c22-00596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
This work aims to investigate the effects and mechanism of emodin in treating diabetic gastroenteropathy and colonic dysmotility in STZ + HS/HF diet induced diabetic gastroenteropathy rats. Diabetic colonic dysmotility model was established by high-fat/high-glucose (HS/HF) feeding combined with streptozotocin (STZ). Emodin was divided into high, medium and low dose groups. After eight weeks of intervention, fasting blood glucose (FBG) and body weight were measured. Gastrointestinal transmission time was evaluated. Serum vasoactive intestinal peptide (VIP) and substance P (SP) were detected. Colonic protein expression of selective autophagy adaptor proteins p62 and beclin1 were detected by immunohistochemistry. Colonic protein expression of beclin1, autophagy related gene 5 (Atg5), C-kit and p62 were detected by Western blot. After treating with emodin, gastrointestinal transmission rate was improved. The expression of serum SP was increased and serum VIP was decreased. Colonic c-kit and p62 were up-regulated. The expressions of beclin1 and Atg5 were down-regulated. Emodin can improve colonic dysmotility and promote the recovery of colonic motility and intestinal defecation in diabetic rats. Its mechanism may involved with up-regulating the expression of C-kit and P62, down-regulating the expression of Beclin1 and Atg5 in colon, which are associated with colon over-autophagy of Cajal interstitial cell (ICC).
Collapse
Affiliation(s)
| | - Na Dong
- Shaanxi University of Chinese Medicine
| | - Yuan Zhou
- Shaanxi University of Chinese Medicine
| | - Hongyan Li
- Affiliated hospital of Shaanxi University of Chinese Medicine
| | - Gangxin Qin
- Affiliated hospital of Shaanxi University of Chinese Medicine
| | - Hui Li
- Shaanxi University of Chinese Medicine
| | | | | | - Yanjin Su
- Affiliated hospital of Shaanxi University of Chinese Medicine
| |
Collapse
|
4
|
Zhu R, Luo Y, Li S, Wang Z. The role of microglial autophagy in Parkinson's disease. Front Aging Neurosci 2022; 14:1039780. [PMID: 36389074 PMCID: PMC9664157 DOI: 10.3389/fnagi.2022.1039780] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/10/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. Studies have shown that abnormal accumulation of α-synuclein (α-Syn) in the substantia nigra is a specific pathological characteristic of PD. Abnormal accumulation of α-Syn in PD induces the activation of microglia. Microglia, which are immune cells in the central nervous system, are involved in the function and regulation of inflammation in PD by autophagy. The role of microglial autophagy in the pathophysiology of PD has become a hot-pot issue. This review outlines the pathways of microglial autophagy, and explores the key factor of microglial autophagy in the mechanism of PD and the possibility of microglial autophagy as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Rui Zhu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yuyi Luo
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China,*Correspondence: Zhengbo Wang,
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China,*Correspondence: Zhengbo Wang,
| |
Collapse
|
5
|
Li C, Ren J, Zhang M, Wang H, Yi F, Wu J, Tang Y. The heterogeneity of microglial activation and its epigenetic and non-coding RNA regulations in the immunopathogenesis of neurodegenerative diseases. Cell Mol Life Sci 2022; 79:511. [PMID: 36066650 DOI: 10.1007/s00018-022-04536-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 12/15/2022]
Abstract
Microglia are resident immune cells in the brain and play a central role in the development and surveillance of the nervous system. Extensive gliosis is a common pathological feature of several neurodegenerative diseases, such as Alzheimer's disease (AD), the most common cause of dementia. Microglia can respond to multiple inflammatory insults and later transform into different phenotypes, such as pro- and anti-inflammatory phenotypes, thereby exerting different functions. In recent years, an increasing number of studies based on both traditional bulk sequencing and novel single-cell/nuclear sequencing and multi-omics analysis, have shown that microglial phenotypes are highly heterogeneous and dynamic, depending on the severity and stage of the disease as well as the particular inflammatory milieu. Thus, redirecting microglial activation to beneficial and neuroprotective phenotypes promises to halt the progression of neurodegenerative diseases. To this end, an increasing number of studies have focused on unraveling heterogeneous microglial phenotypes and their underlying molecular mechanisms, including those due to epigenetic and non-coding RNA modulations. In this review, we summarize the epigenetic mechanisms in the form of DNA and histone modifications, as well as the general non-coding RNA regulations that modulate microglial activation during immunopathogenesis of neurodegenerative diseases and discuss promising research approaches in the microglial era.
Collapse
Affiliation(s)
- Chaoyi Li
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jie Ren
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mengfei Zhang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huakun Wang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Fang Yi
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Junjiao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Tang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China.
- The Biobank of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Li H, Li X, Xu Z, Lu J, Cao C, You W, Yu Z, Shen H, Chen G. Unbalanced Regulation of Sec22b and Ykt6 Blocks Autophagosome Axonal Retrograde Flux in Neuronal Ischemia-Reperfusion Injury. J Neurosci 2022; 42:5641-5654. [PMID: 35654605 PMCID: PMC9295843 DOI: 10.1523/jneurosci.2030-21.2022] [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: 10/08/2021] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/21/2022] Open
Abstract
Cerebral ischemia-reperfusion (I/R) injury in ischemic penumbra is accountable for poor outcome of ischemic stroke patients receiving recanalization therapy. Compelling evidence previously demonstrated a dual role of autophagy in stroke. This study aimed to understand the traits of autophagy in the ischemic penumbra and the potential mechanism that switches the dual role of autophagy. We found that autophagy induction by rapamycin and lithium carbonate performed before ischemia reduced neurologic deficits and infarction, while autophagy induction after reperfusion had the opposite effect in the male murine middle cerebral artery occlusion/reperfusion (MCAO/R) model, both of which were eliminated in mice lacking autophagy (Atg7flox/flox; Nestin-Cre). Autophagic flux determination showed that reperfusion led to a blockage of axonal autophagosome retrograde transport in neurons, which then led to autophagic flux damage. Then, we found that I/R induced changes in the protein levels of Sec22b and Ykt6 in neurons, two autophagosome transport-related factors, in which Sec22b significantly increased and Ykt6 significantly decreased. In the absence of exogenous autophagy induction, Sec22b knock-down and Ykt6 overexpression significantly alleviated autophagic flux damage, infarction, and neurologic deficits in neurons or murine exposed to cerebral I/R in an autophagy-dependent manner. Furthermore, Sec22b knock-down and Ykt6 overexpression switched the outcome of rapamycin posttreatment from deterioration to neuroprotection. Thus, Sec22b and Ykt6 play key roles in neuronal autophagic flux, and modest regulation of Sec22b and Ykt6 may help to reverse the failure of targeting autophagy induction to improve the prognosis of ischemic stroke.SIGNIFICANCE STATEMENT The highly polarized architecture of neurons with neurites presents challenges for material transport, such as autophagosomes, which form at the neurite tip and need to be transported to the cell soma for degradation. Here, we demonstrate that Sec22b and Ykt6 act as autophagosome porters and play an important role in maintaining the integrity of neuronal autophagic flux. Ischemia-reperfusion (I/R)-induced excess Sec22b and loss of Ykt6 in neurons lead to axonal autophagosome retrograde trafficking failure, autophagic flux damage, and finally neuronal injury. Facilitated axonal autophagosome retrograde transport by Sec22b knock-down and Ykt6 overexpression may reduce I/R-induced neuron injury and extend the therapeutic window of pharmacological autophagy induction for neuroprotection.
Collapse
Affiliation(s)
- Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Zhongmou Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Chang Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215006, China
| |
Collapse
|
7
|
Kho W, von Haefen C, Paeschke N, Nasser F, Endesfelder S, Sifringer M, González-López A, Lanzke N, Spies CD. Dexmedetomidine Restores Autophagic Flux, Modulates Associated microRNAs and the Cholinergic Anti-inflammatory Pathway upon LPS-Treatment in Rats. J Neuroimmune Pharmacol 2022; 17:261-276. [PMID: 34357471 PMCID: PMC9726767 DOI: 10.1007/s11481-021-10003-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/26/2021] [Indexed: 12/29/2022]
Abstract
Infections and perioperative stress can lead to neuroinflammation, which in turn is linked to cognitive impairments such as postoperative delirium or postoperative cognitive dysfunctions. The α2-adrenoceptor agonist dexmedetomidine (DEX) prevents cognitive impairments and has organo-protective and anti-inflammatory properties. Macroautophagy (autophagy) regulates many biological processes, but its role in DEX-mediated anti-inflammation and the underlying mechanism of DEX remains largely unclear. We were interested how a pretreatment with DEX protects against lipopolysaccharide (LPS)-induced inflammation in adult male Wistar rats. We used Western blot and activity assays to study how DEX modulated autophagy- and apoptosis-associated proteins as well as molecules of the cholinergic anti-inflammatory pathway, and qPCR to analyse the expression of autophagy and inflammation-associated microRNAs (miRNA) in the spleen, cortex and hippocampus at different time points (6 h, 24 h, 7 d). We showed that a DEX pretreatment prevents LPS-induced impairments in autophagic flux and attenuates the LPS-induced increase in the apoptosis-associated protein cleaved poly(ADP-ribose)-polymerase (PARP) in the spleen. Both, DEX and LPS altered miRNA expression and molecules of the cholinergic anti-inflammatory pathway in the spleen and brain. While only a certain set of miRNAs was up- and/or downregulated by LPS in each tissue, which was prevented or attenuated by a DEX pretreatment in the spleen and hippocampus, all miRNAs were up- and/or downregulated by DEX itself - independent of whether or not they were altered by LPS. Our results indicate that the organo-protective effect of DEX may be mediated by autophagy, possibly by acting on associated miRNAs, and the cholinergic anti-inflammatory pathway. Preventive effects of DEX on LPS-induced inflammation. DEX restores the LPS-induced impairments in autophagic flux, attenuates PARP cleavage and alters molecules of the cholinergic system in the spleen. Furthermore, DEX alters and prevents LPS-induced miRNA expression changes in the spleen and brain along with LPS.
Collapse
Affiliation(s)
- Widuri Kho
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nadine Paeschke
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Fatme Nasser
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Sifringer
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Adrián González-López
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany ,CIBER-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Nadine Lanzke
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia D. Spies
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Gao S, Zhou Q, Jin H, Shi N, Wang X, Zhang L, Yan M. Effect of pyrroloquinoline quinone on lipopolysaccharide-induced autophagy in HAPI microglia cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1377. [PMID: 34733929 PMCID: PMC8506552 DOI: 10.21037/atm-21-730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/04/2021] [Indexed: 12/01/2022]
Abstract
Background Pyrroloquinoline quinone (PQQ) is involved in various physiological and biochemical processes, including antioxidant, cell proliferation, and mitochondrial formation. It plays a vital role in protecting neurons. However, the effect of PQQ on microglia, an inflammatory cell of the central nervous system (CNS), is still unclear. This study aimed to investigate the biological role and neuroprotective mechanism of PQQ in HAPI microglial cells exposed to lipopolysaccharide (LPS). Methods Western blot (WB) was used to detect apoptosis and autophagy-related molecules Bax, Bcl2, active-caspase-3, caspase-3, LC3, lysosomal associated membrane protein 2 (LAMP2), AKT, tumor necrosis factor receptor (TNFR) 1, and TNFR2 expression. The phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor LY294002 was used to block the Akt pathway. WB detected the effects of PI3K on autophagy and TNFR1 and TNFR2 expression. The localization of active-caspase-3, caspase-3, LC3, LAMP2, TNFR1, and TNFR2 in cells was observed by immunofluorescence staining. The effect of PQQ on the cell cycle was examined by flow cytometry. We used 5-Ethynyl-2’-deoxyuridine (EdU) assay to detect cell proliferation. The migration ability of cells under different conditions was detected by scratch test and Transwell assay. Results Our results showed that there were different effects on the apoptosis-related molecules Bcl2/Bax and active-caspase-3/caspase in HAPI microglial cells treated with PQQ at different times. PQQ had no significant effect on the LC3b/a ratio in the early stage, which was upregulated in the later stage. The expression of LAMP2 was significantly increased in both early and late stages after PQQ treatment. At the same time, we found that PQQ can reverse the translocation of LAMP2 from the cytoplasm to the nucleus in LPS-induced HAPI microglia. After PQQ treatment, TNFR1 was significantly decreased, but TNFR2 increased in LPS-induced HAPI microglia. It may be that PQQ works through the PI3K/Akt signaling pathway to up-regulate LC3, LAMP2, and TNFR1 and down-regulate TNFR2 in LPS-induced HAPI microglia. However, PQQ has little effect on LPS-induced proliferation, cell cycle, and migration of HAPI microglia. Conclusions In LPS-induced HAPI microglia, PQQ reduces the apoptosis level and increases that of autophagy. In addition, PQQ changes the distribution of LAMP2 in the cytoplasm and nucleus, which is regulated through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Shumei Gao
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Qiao Zhou
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Hui Jin
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Naiqi Shi
- School of Chemistry and Molecular Biosciences, the University of Queensland, Brisbane, Queensland, Australia
| | - Xiaoyu Wang
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Li Zhang
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Meijuan Yan
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
9
|
Pan X, Wang G, Wang B. MicroRNA-1182 and let-7a exert synergistic inhibition on invasion, migration and autophagy of cholangiocarcinoma cells through down-regulation of NUAK1. Cancer Cell Int 2021; 21:161. [PMID: 33750398 PMCID: PMC7942015 DOI: 10.1186/s12935-021-01797-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
Background Cholangiocarcinoma (CCA) is the second most common primary liver malignancy worldwide. Several microRNAs (miRNAs) have been implicated as potential tumor suppressors in CCA. This study aims to explore the potential effects of miR-1182 and let-7a on CCA development. Methods Bioinformatics analysis was conducted to screen differentially expressed genes in CCA, Western blot analysis detected NUAK1 protein expression and RT-qPCR detected miR-1182, let-7a and NUAK1 expression in CCA tissues and cell lines. Dual luciferase reporter gene assay and RIP were applied to validate the relationship between miR-1182 and NUAK1 as well as between let-7a and NUAK1. Functional experiment was conducted to investigate the role of miR-1182, let-7a and NUAK1 in cell migration, proliferation and autophagy. Then, the CCA cells that received various treatments were implanted to mice to establish animal model, followed by tumor observation and HE staining to evaluate lung metastasis. Results CCA tissues and cells were observed to have a high expression of NUAK1 and poor expression of miR-1182 and let-7a. NUAK1 was indicated as a target gene of miR-1182 and let-7a. Importantly, upregulation of either miR-1182 or let-7a induced autophagy, and inhibited cell progression and in vivo tumor growth and lung metastasis; moreover, combined treatment of miR-1182 and let-7a overexpression presented with enhanced inhibitory effect on NUAK1 expression and CCA progression, but such synergistic effect could be reversed by overexpression of NUAK1. Conclusion Taken together, the findings suggest the presence of a synergistic antitumor effect of miR-1182 and let-7a on the development of CCA via the down-regulation of NUAK1, providing novel insight into the targeted therapy against CCA.
Collapse
Affiliation(s)
- Xin Pan
- Interventional Department, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, People's Republic of China.
| | - Gang Wang
- Interventional Department, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, People's Republic of China
| | - Baoming Wang
- Interventional Department, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, People's Republic of China
| |
Collapse
|
10
|
Brites D. Regulatory function of microRNAs in microglia. Glia 2020; 68:1631-1642. [PMID: 32463968 DOI: 10.1002/glia.23846] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/14/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
Microglia are CNS-resident cells involved in immune surveillance and maintenance of intercellular homeostasis, while also contributing to neurologic pathologies. MicroRNAs (miRNAs) are a class of small (~22 nucleotides) single-stranded noncoding RNAs that participate in gene regulation at the post-transcriptional level. miRNAs typically bind to the untranslated region (3' UTR) of RNAs. It has been shown that miRNAs are important players in controlling inflammation and that their abnormal expression is linked to cancer and ageing, and to the onset and progression of neurodegenerative disease. Furthermore, miRNAs participate in intercellular trafficking. Thus, miRNAs are released from cells in a free form, bound to proteins or packaged within extracellular vesicles (EVs), exerting paracrine and long distance signaling. In this review, recent findings on the role of miRNAs as drivers of microglia phenotypic changes and their cotribution in neurological disease are addressed. MAIN POINTS: miRNAs have a key role in microglia function/dysfunction, polarization, and restoration. Microglia are both a source and recipient of extracellular vesicles (EVs) containing miRNAs. Extracellular miRNAs may be found as soluble (free and EV cargo) and protein complexes.
Collapse
Affiliation(s)
- Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Lee JW, Nam H, Kim LE, Jeon Y, Min H, Ha S, Lee Y, Kim SY, Lee SJ, Kim EK, Yu SW. TLR4 (toll-like receptor 4) activation suppresses autophagy through inhibition of FOXO3 and impairs phagocytic capacity of microglia. Autophagy 2018; 15:753-770. [PMID: 30523761 DOI: 10.1080/15548627.2018.1556946] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Macroautophagy/autophagy is a lysosome-dependent catabolic process for the turnover of proteins and organelles in eukaryotes. Autophagy plays an important role in immunity and inflammation, as well as metabolism and cell survival. Diverse immune and inflammatory signals induce autophagy in macrophages through pattern recognition receptors, such as toll-like receptors (TLRs). However, the physiological role of autophagy and its signaling mechanisms in microglia remain poorly understood. Microglia are phagocytic immune cells that are resident in the central nervous system and share many characteristics with macrophages. Here, we show that autophagic flux and expression of autophagy-related (Atg) genes in microglia are significantly suppressed upon TLR4 activation by lipopolysaccharide (LPS), in contrast to their stimulation by LPS in macrophages. Metabolomics analysis of the levels of phosphatidylinositol (PtdIns) and its 3-phosphorylated form, PtdIns3P, in combination with bioinformatics prediction, revealed an LPS-induced reduction in the synthesis of PtdIns and PtdIns3P in microglia but not macrophages. Interestingly, inhibition of PI3K, but not MTOR or MAPK1/3, restored autophagic flux with concomitant dephosphorylation and nuclear translocation of FOXO3. A constitutively active form of FOXO3 also induced autophagy, suggesting FOXO3 as a downstream target of the PI3K pathway for autophagy inhibition. LPS treatment impaired phagocytic capacity of microglia, including MAP1LC3B/LC3-associated phagocytosis (LAP) and amyloid β (Aβ) clearance. PI3K inhibition restored LAP and degradation capacity of microglia against Aβ. These findings suggest a unique mechanism for the regulation of microglial autophagy and point to the PI3K-FOXO3 pathway as a potential therapeutic target to regulate microglial function in brain disorders. Abbreviations: Atg: autophagy-related gene; Aβ: amyloid-β; BafA1: bafilomycin A1; BECN1: beclin 1, autophagy related; BMDM: bone marrow-derived macrophage; CA: constitutively active; CNS: central nervous system; ZFYVE1/DFCP1: zinc finger, FYVE domain containing 1; FOXO: forkhead box O; ELISA:enzyme-linked immunosorbent assay; HBSS: Hanks balanced salt solution; LAP: LC3-associated phagocytosis; MAP1LC3B: microtubule-associated protein 1 light chain 3; LPS: lipopolysaccharide; LY: LY294002; MTOR: mechanistic target of rapamycin kinase; Pam3CSK4: N-palmitoyl-S-dipalmitoylglyceryl Cys-Ser-(Lys)4; PtdIns: phosphatidylinositol; PtdIns3P: phosphatidylinositol-3-phosphate; PLA: proximity ligation assay; Poly(I:C): polyinosinic-polycytidylic acid; qRT-PCR: quantitative real-time polymerase chain reaction; RPS6KB1: ribosomal protein S6 kinase, polypeptide 1; TLR: Toll-like receptor; TNF: tumor necrosis factor; TFEB: transcription factor EB; TSPO: translocator protein.
Collapse
Affiliation(s)
- Ji-Won Lee
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Hyeri Nam
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Leah Eunjung Kim
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Yoonjeong Jeon
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea.,b Neurometabolomics Research Center , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Hyunjung Min
- c Department of Neuroscience and Physiology , Dental Research Institute, School of Dentistry, Seoul National University , Seoul , Republic of Korea
| | - Shinwon Ha
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Younghwan Lee
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Seon-Young Kim
- d Gene Editing Research Center , KRIBB , Daejeon , Republic of Korea
| | - Sung Joong Lee
- c Department of Neuroscience and Physiology , Dental Research Institute, School of Dentistry, Seoul National University , Seoul , Republic of Korea
| | - Eun-Kyoung Kim
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea.,b Neurometabolomics Research Center , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| | - Seong-Woon Yu
- a Department of Brain and Cognitive Sciences , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea.,b Neurometabolomics Research Center , Daegu Gyeongbuk Institute of Science and Technology (DGIST) , Daegu , Republic of Korea
| |
Collapse
|
12
|
Immune Cells After Ischemic Stroke Onset: Roles, Migration, and Target Intervention. J Mol Neurosci 2018; 66:342-355. [DOI: 10.1007/s12031-018-1173-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/14/2018] [Indexed: 01/09/2023]
|
13
|
Cheray M, Joseph B. Epigenetics Control Microglia Plasticity. Front Cell Neurosci 2018; 12:243. [PMID: 30123114 PMCID: PMC6085560 DOI: 10.3389/fncel.2018.00243] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 01/31/2023] Open
Abstract
Microglia, resident immune cells of the central nervous system, fulfill multiple functions in the brain throughout life. These microglial functions range from participation in innate and adaptive immune responses, involvement in the development of the brain and its homeostasis maintenance, to contribution to degenerative, traumatic, and proliferative diseases; and take place in the developing, the aging, the healthy, or the diseased brain. Thus, an impressive level of cellular plasticity, appears as a requirement for the pleiotropic biological functions of microglia. Epigenetic changes, including histone modifications or DNA methylation as well as microRNA expression, are important modifiers of gene expression, and have been involved in cell phenotype regulation and reprogramming and are therefore part of the mechanisms regulating cellular plasticity. Here, we review and discuss the epigenetic mechanisms, which are emerging as contributors to this microglial cellular plasticity and thereby can constitute interesting targets to modulate microglia associated brain diseases, including developmental diseases, neurodegenerative diseases as well as cancer.
Collapse
Affiliation(s)
- Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
14
|
Systemic inflammation induced by lipopolysaccharide aggravates inherited retinal dystrophy. Cell Death Dis 2018; 9:350. [PMID: 29500424 PMCID: PMC5834451 DOI: 10.1038/s41419-018-0355-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/16/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
Retinal neurodegenerative diseases involve a scenario of inflammation and cell death that leads to morphological alterations and visual impairment. Non-ocular inflammatory processes could affect neurodegenerative retinal disorders and their progression, at least in part by activating microglial cells and releasing pro-inflammatory cytokines. Our purpose was to study the consequences of a systemic inflammatory process in the progression of retinal degeneration in P23H rats, a retinitis pigmentosa (RP) model. In order to induce a mild chronic systemic inflammation, we administered low doses of lipopolysaccharide (LPS) from age P20 to P60 to dystrophic P23H rats and healthy SD rats. Visual responsiveness was assessed by electroretinography (ERG). The morphological state of the retinas was analyzed by fluorescent immunohistochemistry (IHC), evaluating the number, morphology, and connectivity of different neuronal populations by means of cell type-specific markers. Microglia density, distribution, and degree of activation were evaluated by IHC and flow cytometry. The expression levels of inflammation- and apoptosis-related genes were analyzed by qRT-PCR arrays. Low-dose LPS administration did not induce significant functional or morphological changes in the retina of SD rats, although at the molecular level, we detected expression changes in genes related to apoptosis. Otherwise, systemic injection of LPS into P23H rats induced a further deterioration in the ERG response, with greater loss of photoreceptors and worsening of synaptic connectivity, accompanied by increasing numbers of microglial cells, which also showed a more intense activation state. Several inflammation- and apoptosis-related genes were upregulated. Our results indicate that chronic exacerbation of the inflammatory response in response to LPS accelerates neurodegeneration in dystrophic P23H rats, suggesting that in patients with ocular neurodegenerative diseases, peripheral damage, as a systemic infection or chronic inflammatory process, could accelerate disease progression, and should be taken into account in order to select an appropriate therapy to revert, block or slow-down the degenerative process.
Collapse
|
15
|
Şekerdağ E, Solaroğlu I, Gürsoy-Özdemir Y. Cell Death Mechanisms in Stroke and Novel Molecular and Cellular Treatment Options. Curr Neuropharmacol 2018; 16:1396-1415. [PMID: 29512465 PMCID: PMC6251049 DOI: 10.2174/1570159x16666180302115544] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/18/2017] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
As a result of ischemia or hemorrhage, blood supply to neurons is disrupted which subsequently promotes a cascade of pathophysiological responses resulting in cell loss. Many mechanisms are involved solely or in combination in this disorder including excitotoxicity, mitochondrial death pathways, and the release of free radicals, protein misfolding, apoptosis, necrosis, autophagy and inflammation. Besides neuronal cell loss, damage to and loss of astrocytes as well as injury to white matter contributes also to cerebral injury. The core problem in stroke is the loss of neuronal cells which makes recovery difficult or even not possible in the late states. Acute treatment options that can be applied for stroke are mainly targeting re-establishment of blood flow and hence, their use is limited due to the effective time window of thrombolytic agents. However, if the acute time window is exceeded, neuronal loss starts due to the activation of cell death pathways. This review will explore the most updated cellular death mechanisms leading to neuronal loss in stroke. Ischemic and hemorrhagic stroke as well as subarachnoid hemorrhage will be debated in the light of cell death mechanisms and possible novel molecular and cellular treatment options will be discussed.
Collapse
Affiliation(s)
- Emine Şekerdağ
- Address correspondence to this author at the Neuroscience Research Lab, Research Center for Translational Medicine, Koç University, Istanbul, Turkey; Tel: +90 850 250 8250; E-mail:
| | | | | |
Collapse
|
16
|
Abstract
Microglia serve as brain-resident myeloid cells that affect cerebral development, ischemia, neurodegeneration, and neuro-viral infection. MicroRNAs play a key role in central nervous system disease through post-transcriptional regulation. Indeed, evidence shows that microRNAs are one of the most important regulators mediating microglial activation, polarization, and autophagy, and subsequently affecting neuroinflammation and the outcome of central nervous system disease. In this review, we provide insight into the function of microRNAs, which may be an attractive strategy and influential treatment for microglia-related central nervous system dysfunction. Moreover, we comprehensively describe how microglia fight against central nervous system disease via multiple functional microRNAs.
Collapse
Affiliation(s)
- Xiao-Hua Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Tian-Long Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University; Institute of Geriatrics; National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
17
|
Jia D, Niu Y, Li D, Zhang Q. MicroRNA-223 alleviates lipopolysaccharide-induced PC-12 cells apoptosis and autophagy by targeting RPH1 in spinal cord injury. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9223-9232. [PMID: 31966794 PMCID: PMC6965896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/20/2017] [Indexed: 06/10/2023]
Abstract
Spinal cord injury (SCI) is one of the most devastating diseases. MicroRNAs (miRNAs) are recognized as key regulators in SCI; however, the role of miR-223 in SCI remains unclear. Herein, our study aimed to explore the effect of miR-223 on lipopolysaccharide (LPS)-induced injury to PC-12 cells. PC-12 cells were treated with different concentrations of LPS, and then cell viability, apoptosis, apoptosis-related factors and autophagy-related factors were analyzed by CCK-8, flow cytometry and western blot. Subsequently, miR-223 mimic, miR-223 inhibitor, pEX-RPH1, sh-RPH1 and corresponding controls were transfected into PC-12 cells followed by 5 μg/ml of LPS treatment. Cell viability, apoptosis, apoptosis-related and autophagy-related factors were analyzed again. A target gene of miR-223 was validated by dual-luciferase assay. Besides, the main factors expressions of mTOR and NF-κB signal pathways were measured by western blot. LPS reduced cell viability but increased apoptotic cells rate, up-regulated Bax, cleaved-caspase-3, cleaved-caspase-9, LC-II and Beclin-1, and down-regulated Bcl-2 and p62 expressions in a dose-dependent way. Additionally, miR-223 overexpression promoted cell viability but inhibited apoptosis, and autophagy in LPS-stimulated PC-12 cells. RPH1 was a direct target of miR-223, and RPH1 exhibited contrary impacts to miR-223 on LPS-induced cell apoptosis and autophagy. Besides, the promoting effects of miR-223 suppression on cell apoptosis and autophagy were relieved by RPH1 silence. Furthermore, miR-223 blocked LPS-induced mTOR and NF-κB pathways by down-regulation of RPH1. MiR-223 improved cell viability but declined apoptosis and autophagy by targeting RPH1 and blocked mTOR and NF-κB pathways in LPS challenged PC-12 cells.
Collapse
Affiliation(s)
- Daofu Jia
- Department of Emergency Medicine, Jinan Central Hospital Jinan 250013, China
| | - Yanping Niu
- Clinic Service Centre, Shandong Provincial Qianfoshan Hospital Jinan 250014, China
| | - Dongling Li
- Department of Operation Room, Children Hospital of Jinan Jinan 250022, China
| | - Qingguo Zhang
- Department of Orthopedic Surgery, Jinan Central Hospital Jinan 250013, China
| |
Collapse
|
18
|
miR-Let7A Controls the Cell Death and Tight Junction Density of Brain Endothelial Cells under High Glucose Condition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6051874. [PMID: 28680530 PMCID: PMC5478855 DOI: 10.1155/2017/6051874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/07/2017] [Accepted: 05/21/2017] [Indexed: 12/13/2022]
Abstract
Hyperglycemia-induced stress in the brain of patients with diabetes triggers the disruption of blood-brain barrier (BBB), leading to diverse neurological diseases including stroke and dementia. Recently, the role of microRNA becomes an interest in the research for deciphering the mechanism of brain endothelial cell damage under hyperglycemia. Therefore, we investigated whether mircoRNA Let7A (miR-Let7A) controls the damage of brain endothelial (bEnd.3) cells against high glucose condition. Cell viability, cell death marker expressions (p-53, Bax, and cleaved poly ADP-ribose polymerase), the loss of tight junction proteins (ZO-1 and claudin-5), proinflammatory response (interleukin-6, tumor necrosis factor-α), inducible nitric oxide synthase, and nitrite production were confirmed using MTT, reverse transcription-PCR, quantitative-PCR, Western blotting, immunofluorescence, and Griess reagent assay. miR-Let7A overexpression significantly prevented cell death and loss of tight junction proteins and attenuated proinflammatory response and nitrite production in the bEnd.3 cells under high glucose condition. Taken together, we suggest that miR-Let7A may attenuate brain endothelial cell damage by controlling cell death signaling, loss of tight junction proteins, and proinflammatory response against high glucose stress. In the future, the manipulation of miR-Let7A may be a novel solution in controlling BBB disruption which leads to the central nervous system diseases.
Collapse
|
19
|
Li H, Wu J, Shen H, Yao X, Liu C, Pianta S, Han J, Borlongan CV, Chen G. Autophagy in hemorrhagic stroke: Mechanisms and clinical implications. Prog Neurobiol 2017; 163-164:79-97. [PMID: 28414101 DOI: 10.1016/j.pneurobio.2017.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/13/2017] [Accepted: 04/08/2017] [Indexed: 02/07/2023]
Abstract
Accumulating evidence advances the critical role of autophagy in brain pathology after stroke. Investigations employing autophagy induction or inhibition using pharmacological tools or autophagy-related gene knockout mice have recently revealed the biological significance of intact and functional autophagy in stroke. Most of the reported cases attest to a pro-survival role for autophagy in stroke, by facilitating removal of damaged proteins and organelles, which can be recycled for energy generation and cellular defenses. However, these observations are difficult to reconcile with equally compelling evidence demonstrating stroke-induced upregulation of brain cell death index that parallels enhanced autophagy. This begs the question of whether drug-induced autophagy during stroke culminates in improved or worsened pathological outcomes. A corollary fascinating hypothesis, but presents as a tricky conundrum, involves the effects of autophagy on cell death and inflammation, which are two main culprits in the disease progression of stroke-induced brain injury. Evidence has extended the roles of autophagy in inflammation via cytokine regulation in an unconventional secretion manner or by targeting inflammasomes for degradation. Moreover, in the recently concluded Vancouver Autophagy Symposium (VAS) held in 2014, the potential of selective autophagy for clinical treatment has been recognized. The role of autophagy in ischemic stroke has been reviewed previously in detail. Here, we evaluate the strength of laboratory and clinical evidence by providing a comprehensive summary of the literature on autophagy, and thereafter we offer our perspectives on exploiting autophagy as a drug target for cerebral ischemia, especially in hemorrhagic stroke.
Collapse
Affiliation(s)
- Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Xiyang Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Chenglin Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - S Pianta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - J Han
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - C V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China.
| |
Collapse
|
20
|
Zhang Z, Tian H, Miao Y, Feng X, Li Y, Wang H, Song X. Upregulation of p72 Enhances Malignant Migration and Invasion of Glioma Cells by Repressing Beclin1 Expression. BIOCHEMISTRY (MOSCOW) 2017; 81:574-82. [PMID: 27301285 DOI: 10.1134/s0006297916060031] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
p72 is the member of the DEAD-box RNA helicase family, which can unwind double-stranded RNA and is efficient for microRNA (miRNA, miR) processing. However, its specific role in glioma has not been elucidated. First, the expression of p72 in glioma cell lines and tissues was explored using Western blot. To explore the role of p72 on glioma progression, adenovirus inhibiting p72 was transfected into A172 and T98G cells. Cell autophagy was determined using GFP-LC3 dots, and cell apoptosis was determined using flow cytometry. The effect of Beclin1 was explored using GFP-LC3 dots, flow cytometry, and colony formation. The possible miRNAs that target the 3'-untranslated region (3'-UTR) of Beclin1 were predicted using TargetScan. Dual luciferase reporter assay was applied to determine whether these miRNAs bind to the 3'-UTR of Beclin1. The expression of p72 was significantly increased in glioma cell lines and tissues. Autophagy-related protein Beclin1 was found to be significantly enhanced when p72 was inhibited. The accumulation of GFP-LC3 dots was significant in cells transfected with ad-sh-p72 compared with ad-con. Colony formation capacity and cell apoptosis were also found to be significantly decreased with p72 inhibition. Furthermore, upregulation of Beclin1 contributes to A172 cell autophagy, invasion, and apoptosis. Overexpression of p72 induces increased miR-34-5p and miR-5195-3p expression in A172 and T98G cells. Beclin1 was the target gene of miR-34-5p and miR-5195-3p. In conclusion, we found for the first time that overexpression of p72 decreased Beclin1 expression partially by increasing miR-34-5p and miR-5195-3p expression in A172 and T98G cells.
Collapse
Affiliation(s)
- Zhenxing Zhang
- Liaoning Medical University, Department of Neurosurgery, Jinzhou, 121001, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Li B, Chen P, Chang Y, Qi J, Fu H, Guo H. Let-7a inhibits tumor cell growth and metastasis by directly targeting RTKN in human colon cancer. Biochem Biophys Res Commun 2016; 478:739-45. [PMID: 27498032 DOI: 10.1016/j.bbrc.2016.08.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 08/03/2016] [Indexed: 11/29/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide, with high morbidity. MicroRNAs (miRNAs) are endogenous small RNAs that play important roles in regulating multiple biological and pathologic processes. The differential expression of miRNAs in CRC was first reported in 2003. Accumulated evidence indicates that lethal-7a (let-7a, miRNA) generally functions as a tumor suppressor in several human cancers. However, the role of let-7a in human colon cancer remains unclear. The aim of this study was to investigate the biological functions of let-7a and its potential role in colon cancer. We first discovered that let-7a level was significantly decreased in colon cancer tissues and cell lines (HT-29, HCT-116, LoVo, SW480, and SW620). To explore the effects of let-7a on colon cancer, let-7a over-expressed HCT-116 and SW620 cells were constructed. Further studies demonstrated that over-expressed let-7a could remarkably inhibit HCT-116 and SW620 cell growth and metastasis by directly down-regulating Rhotekin (RTKN). When RTKN was reintroduced into let-7a mimic transfected HCT-116 or SW620 cells, the inhibition effects of let-7a on colon cancer cell growth and metastasis were markedly reversed. In conclusion, our research shows that let-7a can inhibit tumor cell growth and metastasis by directly targeting RTKN in human colon cancer.
Collapse
Affiliation(s)
- Bin Li
- Department of Oncology, The First Affiliated Hospital of Xi'an Medical University, Shaanxi, Xi'an 710077, China.
| | - Peng Chen
- Institution of Basic Medical Science, Xi'an Medical University, Shaanxi, Xi'an 710021, China; School of Life Sciences, Northwest University, Shaanxi, Xi'an 710069, China
| | - Yanxiang Chang
- Department of Oncology, The First Affiliated Hospital of Xi'an Medical University, Shaanxi, Xi'an 710077, China
| | - Jingpeng Qi
- Department of Oncology, The First Affiliated Hospital of Xi'an Medical University, Shaanxi, Xi'an 710077, China
| | - Hui Fu
- Institution of Basic Medical Science, Xi'an Medical University, Shaanxi, Xi'an 710021, China
| | - Huifang Guo
- Institution of Basic Medical Science, Xi'an Medical University, Shaanxi, Xi'an 710021, China
| |
Collapse
|
22
|
Ziółkowska B, Woźniak M, Ziółkowski P. Co-expression of autophagic markers following photodynamic therapy in SW620 human colon adenocarcinoma cells. Mol Med Rep 2016; 14:2548-54. [PMID: 27485939 PMCID: PMC4991753 DOI: 10.3892/mmr.2016.5541] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/31/2016] [Indexed: 12/19/2022] Open
Abstract
Photodynamic therapy (PDT) is a minimally invasive cancer treatment. It involves the combination of a photosensitizer and light of a specific wavelength to generate singlet oxygen and other reactive oxygen species that lead to tumor cell death. Autophagy is one of the pathways that tumor cells undergo during photodamage and it is common in photodynamic therapy. The aim of this study was to examine the effect of in vitro PDT on the expression of autophagy‑related proteins, autophagy related 7 (Atg7), light chain 3 (LC3) and Beclin‑1. Human SW620 colon carcinoma cells were treated with 5-aminolevulinic acid (ALA)‑based PDT at a dose of 3 mM. The irradiation was performed using 4.5 J/cm2 total light and a fluence rate of 60 mW/cm2. Autophagy was evaluated by immunocytochemistry using specific antibodies to Atg7, Beclin‑1 and LC3. The evaluation was repeated at several time points (0, 4, 8 and 24 h) following irradiation. The induction of autophagy was observed directly following the 5‑ALA‑mediated PDT procedure with the strongest expression of autophagy-related proteins at 4 and 8 h after irradiation as demonstrated using immunocytochemistry. It was characterized by significantly increased expression of Beclin‑1, Atg7 and LC3. To the best of our knowledge this is the first study to analyze Beclin‑1, Atg7 and LC3 expression in a PDT‑related experiment. This study enhances the understanding of the role of autophagy in PDT, which may contribute to better and more effective tumor responses to this therapy.
Collapse
Affiliation(s)
- Barbara Ziółkowska
- Department of Pathology, Wrocław Medical University, 50‑368 Wrocław, Poland
| | - Marta Woźniak
- Department of Pathology, Wrocław Medical University, 50‑368 Wrocław, Poland
| | - Piotr Ziółkowski
- Department of Pathology, Wrocław Medical University, 50‑368 Wrocław, Poland
| |
Collapse
|
23
|
Song J, Jun M, Ahn MR, Kim OY. Involvement of miR-Let7A in inflammatory response and cell survival/apoptosis regulated by resveratrol in THP-1 macrophage. Nutr Res Pract 2016; 10:377-84. [PMID: 27478543 PMCID: PMC4958639 DOI: 10.4162/nrp.2016.10.4.377] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/25/2016] [Accepted: 04/14/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND/OBJECTIVES Resveratrol, a natural polyphenol, has multiple functions in cellular responses including apoptosis, survival, and differentiation. It also participates in the regulation of inflammatory response and oxidative stress. MicroRNA-Let-7A (miR-Let7A), known as a tumor suppressor miRNA, was recently reported to play a crucial role in both inflammation and apoptosis. Therefore, we examined involvement of miR-Let7A in the modulation of inflammation and cell survival/apoptosis regulated by resveratrol. MATERIALS/METHODS mRNA expression of pro-/anti-inflammatory cytokines and sirtuin 1 (SIRT1), and protein expression of apoptosis signal-regulating kinase 1 (ASK1), p-ASK1, and caspase-3 and cleaved caspase-3 were measured, and cell viability and Hoechst/PI staining for apoptosis were observed in Lipopolysaccharide (LPS)-stimulated human THP-1 macrophages with the treatment of resveratrol and/or miR-Let7A overexpression. RESULTS Pre-treatment with resveratrol (25-200 µM) resulted in significant recovery of the reduced cell viabilities under LPS-induced inflammatory condition and in markedly increased expression of miR-Let7A in non-stimulated or LPS-stimulated cells. Increased mRNA levels of tumor necrosis factor-α and interleukin (IL)-6 induced by LPS were significantly attenuated, and decreased levels of IL-10 and brain-derived neurotrophic factor were significantly restored by resveratrol and miR-Let7A overexpression, respectively, or in combination. Decreased expression of IL-4 mRNA by LPS stimulation was also significantly increased by miR-Let7A overexpression co-treated with resveratrol. In addition, decreased SIRT1 mRNA levels, and increased p-ASK1 levels and PI-positive cells by LPS stimulation were significantly restored by resveratrol and miR-Let7A overexpression, respectively, or in combination. CONCLUSIONS miR-Let7A may be involved in the inflammatory response and cell survival/apoptosis modulated by resveratrol in human THP-1 macrophages.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Biomedical Engineering, Dongguk University, Gyeonggi 10326, Korea.; Department of Biology, York University, Toronto, Ontario, Canada
| | - Mira Jun
- Department of Food Science and Nutrition, Dong-A University, 37 Nakdong-daero 550beon-gil, Saha-gu, Busan 49315, Korea
| | - Mok-Ryeon Ahn
- Department of Food Science and Nutrition, Dong-A University, 37 Nakdong-daero 550beon-gil, Saha-gu, Busan 49315, Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, 37 Nakdong-daero 550beon-gil, Saha-gu, Busan 49315, Korea
| |
Collapse
|
24
|
Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol 2016; 157:247-272. [PMID: 26851161 DOI: 10.1016/j.pneurobio.2016.01.005] [Citation(s) in RCA: 514] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/22/2015] [Accepted: 01/10/2016] [Indexed: 12/16/2022]
Abstract
Microglia are brain resident macrophages originated from primitive progenitor cells in the yolk sac. Microglia can be activated within hours and recruited to the lesion site. Traditionally, microglia activation is considered to play a deleterious role in ischemic stroke, as inhibition of microglia activation attenuates ischemia induced brain injury. However, increasing evidence show that microglia activation is critical for attenuating neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after cerebral ischemia. Differential polarization of microglia could likely explain the biphasic role of microglia in ischemia. We comprehensively reviewed the mechanisms involved in regulating microglia activation and polarization. The latest discoveries of microRNAs in modulating microglia function are discussed. In addition, the interaction between microglia and other cells including neurons, astrocytes, oligodendrocytes, and stem cells were also reviewed. Future therapies targeting microglia may not exclusively aim at suppressing microglia activation, but also at modulating microglia polarization at different stages of ischemic stroke. More work is needed to elucidate the cellular and molecular mechanisms of microglia polarization under ischemic environment. The roles of microRNAs and transplanted stem cells in mediating microglia activation and polarization during brain ischemia also need to be further studied.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jixian Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Rehabilitation, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
25
|
The role of autophagy in modulation of neuroinflammation in microglia. Neuroscience 2016; 319:155-67. [PMID: 26827945 DOI: 10.1016/j.neuroscience.2016.01.035] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 01/05/2023]
Abstract
Microglia have multiple functions in regulating homeostasis in the central nervous system (CNS), and microglial inflammation is thought to play a role in the etiology of the neurodegenerative diseases. When endogenous or exogenous stimuli trigger disorders in microenvironmental homeostasis in CNS, microglia critically determine the fate of other neural cells. Recently, it was reported that autophagy might influence inflammation and activation of microglia. Though the interaction between autophagy and macrophages has been reported and reviewed in length, the role of autophagy in microglia has yet to be reviewed. Herein, we will highlight recent advances on the emerging role of autophagy in microglia, focusing on the regulation of autophagy during microglial inflammation, and the possible mechanism involved.
Collapse
|