1
|
Khan A, Mallick M, Ladke JS, Bhandari R. The ring rules the chain - inositol pyrophosphates and the regulation of inorganic polyphosphate. Biochem Soc Trans 2024; 52:567-580. [PMID: 38629621 DOI: 10.1042/bst20230256] [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: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
The maintenance of phosphate homeostasis serves as a foundation for energy metabolism and signal transduction processes in all living organisms. Inositol pyrophosphates (PP-InsPs), composed of an inositol ring decorated with monophosphate and diphosphate moieties, and inorganic polyphosphate (polyP), chains of orthophosphate residues linked by phosphoanhydride bonds, are energy-rich biomolecules that play critical roles in phosphate homeostasis. There is a complex interplay between these two phosphate-rich molecules, and they share an interdependent relationship with cellular adenosine triphosphate (ATP) and inorganic phosphate (Pi). In eukaryotes, the enzymes involved in PP-InsP synthesis show some degree of conservation across species, whereas distinct enzymology exists for polyP synthesis among different organisms. In fact, the mechanism of polyP synthesis in metazoans, including mammals, is still unclear. Early studies on PP-InsP and polyP synthesis were conducted in the slime mould Dictyostelium discoideum, but it is in the budding yeast Saccharomyces cerevisiae that a clear understanding of the interplay between polyP, PP-InsPs, and Pi homeostasis has now been established. Recent research has shed more light on the influence of PP-InsPs on polyP in mammals, and the regulation of both these molecules by cellular ATP and Pi levels. In this review we will discuss the cross-talk between PP-InsPs, polyP, ATP, and Pi in the context of budding yeast, slime mould, and mammals. We will also highlight the similarities and differences in the relationship between these phosphate-rich biomolecules among this group of organisms.
Collapse
Affiliation(s)
- Azmi Khan
- Laboratory of Cell Signalling, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India
| | - Manisha Mallick
- Laboratory of Cell Signalling, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India
- Graduate Studies, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Jayashree S Ladke
- Laboratory of Cell Signalling, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India
- Graduate Studies, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Rashna Bhandari
- Laboratory of Cell Signalling, Centre for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India
| |
Collapse
|
2
|
Kang QM, Wang J, Chen SM, Song SR, Yu SC. Glioma-associated mesenchymal stem cells. Brain 2024; 147:755-765. [PMID: 37850820 DOI: 10.1093/brain/awad360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 09/06/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
Recent studies have revealed that glioma-associated mesenchymal stem cells play instrumental roles in tumorigenesis and tumour progression and cannot be ignored as a cellular component of the glioma microenvironment. Nevertheless, the origin of these cells and their roles are poorly understood. The only relevant studies have shown that glioma-associated mesenchymal stem cells play a large role in promoting tumour proliferation, invasion and angiogenesis. This review provides a comprehensive summary of their discovery and definition, origin, differences from other tissue-derived mesenchymal stem cells, spatial distribution, functions and prognostic and therapeutic opportunities to deepen the understanding of these cells and provide new insight into the treatment of glioma.
Collapse
Affiliation(s)
- Qing-Mei Kang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Shi-Man Chen
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Si-Rong Song
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| |
Collapse
|
3
|
Qi J, Shi L, Zhu L, Chen Y, Zhu H, Cheng W, Chen AF, Fu C. Functions, Mechanisms, and therapeutic applications of the inositol pyrophosphates 5PP-InsP 5 and InsP 8 in mammalian cells. J Cardiovasc Transl Res 2024; 17:197-215. [PMID: 37615888 DOI: 10.1007/s12265-023-10427-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023]
Abstract
Water-soluble myo-inositol phosphates have long been characterized as second messengers. The signaling properties of these compounds are determined by the number and arrangement of phosphate groups on the myo-inositol backbone. Recently, higher inositol phosphates with pyrophosphate groups were recognized as signaling molecules. 5-Diphosphoinositol 1,2,3,4,6-pentakisphosphate (5PP-InsP5) is the most abundant isoform, constituting more than 90% of intracellular inositol pyrophosphates. 5PP-InsP5 can be further phosphorylated to 1,5-bisdiphosphoinositol 2,3,4,6-tetrakisphosphate (InsP8). These two molecules, 5PP-InsP5 and InsP8, are present in various subcellular compartments, where they participate in regulating diverse cellular processes such as cell death, energy homeostasis, and cytoskeletal dynamics. The synthesis and metabolism of inositol pyrophosphates are subjected to tight regulation, allowing for their highly specific functions. Blocking the 5PP-InsP5/InsP8 signaling pathway by inhibiting the biosynthesis of 5PP-InsP5 demonstrates therapeutic benefits in preclinical studies, and thus holds promise as a therapeutic approach for certain diseases treatment, such as metabolic disorders.
Collapse
Affiliation(s)
- Ji Qi
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Linhui Shi
- Department of Critical Care Unit, Ningbo Medical Center Li Huili Hospital, Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Limei Zhu
- Department of Trauma Orthopedics, Ningbo No.6 Hospital, Ningbo, 315040, China
| | - Yuanyuan Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hong Zhu
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Weiwei Cheng
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Chenglai Fu
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
4
|
Lao J, Hu P, Li J, Li J, Zhang X, Yang H, Qi C. Acupuncture Combined with Bushen-Jianpi Decoction Ameliorates the Ovarian Function of Diminished Ovarian Reserve Rats by Regulating Phosphoinositide 3-Kinase/AKT Signaling. Comb Chem High Throughput Screen 2024; 27:2402-2418. [PMID: 38178681 DOI: 10.2174/0113862073264971231113061204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/22/2023] [Accepted: 10/11/2023] [Indexed: 01/06/2024]
Abstract
OBJECTIVE This study aimed to explore the therapeutic efficiency as well as mechanism of acupuncture combined with Bushen-Jianpi decoction (BJD) to treat rats with diminished ovarian reserve (DOR). METHODS A DOR rat model was constructed using zona pellucida 3 peptide, and acupuncture, BJD, and their combination were administered as therapeutic interventions. We measured changes in the ovarian indexes, the number of follicles at all levels, the serum levels of sex hormones and immune factors, the expression levels of phosphoinositide 3-kinase (PI3K), AKT, p-AKT, and caspase-3, and the changes in the proportions of splenic T cell subtypes, including T-helper 17 (Th17), Tc17, regulatory T (Treg), CD4+, and CD8+ cells. RESULTS Acupuncture combined with BJD induced a decrease in the levels of follicle-stimulating and luteinizing hormones, and the effect was greater than that elicited by BJD or acupuncture alone (P < 0.05). Additionally, this combination treatment effectively abrogated the increase in the levels of interleukin-2 (IL-2), IL-17, anti-zona pellucida antibody, and cleaved caspase-3 (P < 0.05), while promoting the regulation of IL-6 and p-AKT (P < 0.01). Furthermore, treatment with acupuncture combined with BJD restored the proportions of CD4+ cells and the CD4+ / CD8+ T cell ratio (P < 0.01), decreased the proportion of CD8+ T and Th17 cells (P < 0.01), and increased the proportions of Tc17 and Treg cells (P < 0.01). CONCLUSION Combining acupuncture with BJD can enhance ovarian function in DOR rats. The regulation of sex hormone levels and immune function in rats may be attributed to the adjustment of the mRNA and proteins levels of PI3K, AKT, and caspase-3 in the PI3K/AKT signaling pathway, which leads to an improvement in the immune function of DOR rats.
Collapse
Affiliation(s)
- Jianting Lao
- Department of Gynaecology, Shanghai Hospital of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Panwei Hu
- Department of Gynaecology, Shanghai Shuguang Hospital, Shanghai, 200071, China
| | - Jia Li
- Department of Gynaecology, Shanghai Shuguang Hospital, Shanghai, 200071, China
| | - Jiahui Li
- Department of Gynaecology, Shanghai Shuguang Hospital, Shanghai, 200071, China
| | - Xiaole Zhang
- Department of Gynaecology, Shanghai Shuguang Hospital, Shanghai, 200071, China
| | - Hong Yang
- Department of Gynaecology, Shanghai Hospital of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Cong Qi
- Department of Gynaecology, Shanghai Shuguang Hospital, Shanghai, 200071, China
| |
Collapse
|
5
|
Du J, Dong Y, Zhu H, Deng Y, Sa C, Yu Q, Li M. DNA damage-induced autophagy is regulated by inositol polyphosphate synthetases in Candida albicans. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119622. [PMID: 37913846 DOI: 10.1016/j.bbamcr.2023.119622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
DNA damage-induced autophagy is a new type of autophagy that differs from traditional macroautophagy; however, this type of autophagy has not been identified in the pathogenic fungus Candida albicans. Inositol polyphosphates are involved in the regulation of DNA damage repair and macroautophagy; however, whether inositol polyphosphates are involved in the regulation of DNA damage-induced autophagy remains unclear. In this study, we identified DNA damage-induced autophagy in C. albicans and systematically investigated the mechanisms of inositol polyphosphate pathway regulation. We found that the core machinery of macro autophagy is also essential for DNA damage-induced autophagy, and that inositol polyphosphate synthetases Kcs1, Ipk1, and Vip1 play a critical role in autophagy. In this study, we focused on Kcs1 and Vip1, which are responsible for the synthesis of inositol pyrophosphate. The kcs1Δ/Δ and vip1Δ/Δ strains exhibited reduced number of phagophore assembly sites (PAS) and autophagic bodies. The recruitment of autophagy-related gene 1 (Atg1) to PAS was significantly affected in the kcs1Δ/Δ and vip1Δ/Δ strains. Target of rapamycin complex 1 kinase activity was elevated in kcs1Δ/Δ and vip1Δ/Δ strains, which significantly inhibited the initiation of autophagy. Atg18 Localization was altered in these mutants. The absence of Kcs1 or Vip1 caused the downregulation of RAD53, a key gene in the DNA damage response. These data provide further understanding of the mechanism of autophagy regulation in C. albicans.
Collapse
Affiliation(s)
- Jiawen Du
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yixuan Dong
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ying Deng
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chula Sa
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
6
|
Sun Z, Cai Y, Chen Y, Jin Q, Zhang Z, Zhang L, Li Y, Huang L, Wang J, Yang Y, Lv Q, Han Z, Xie M, Zhu X. Ultrasound-targeted microbubble destruction promotes PDGF-primed bone mesenchymal stem cell transplantation for myocardial protection in acute Myocardial Infarction in rats. J Nanobiotechnology 2023; 21:481. [PMID: 38102643 PMCID: PMC10725038 DOI: 10.1186/s12951-023-02204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Ultrasound-targeted microbubble destruction (UTMD) has emerged as a promising strategy for the targeted delivery of bone marrow mesenchymal stem cells (MSCs) to the ischemic myocardium. However, the limited migration capacity and poor survival of MSCs remains a major therapeutic barrier. The present study was performed to investigate the synergistic effect of UTMD with platelet-derived growth factor BB (PDGF-BB) on the homing of MSCs for acute myocardial infarction (AMI). METHODS MSCs from male donor rats were treated with PDGF-BB, and a novel microbubble formulation was prepared using a thin-film hydration method. In vivo, MSCs with or without PDGF-BB pretreatment were transplanted by UTMD after inducing AMI in experimental rats. The therapeutic efficacy of PDGF-BB-primed MSCs on myocardial apoptosis, angiogenesis, cardiac function and scar repair was estimated. The effects and molecular mechanisms of PDGF-BB on MSC migration and survival were explored in vitro. RESULTS The results showed that the biological effects of UTMD increased the local levels of stromal-derived factor-1 (SDF-1), which promoted the migration of transplanted MSCs to the ischemic region. Compared with UTMD alone, UTMD combined with PDGF-BB pretreatment significantly increased the cardiac homing of MSCs, which subsequently reduced myocardial apoptosis, promoted neovascularization and tissue repair, and increased cardiac function 30 days after MI. The vitro results demonstrated that PDGF-BB enhanced MSC migration and protected these cells from H2O2-induced apoptosis. Mechanistically, PDGF-BB pretreatment promoted MSC migration and inhibited H2O2-induced MSC apoptosis via activation of the phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/Akt) pathway. Furthermore, crosstalk between PDGF-BB and stromal-derived factor-1/chemokine receptor 4 (SDF-1/CXCR4) is involved in the PI3K/AKT signaling pathway. CONCLUSION The present study demonstrated that UTMD combined with PDGF-BB treatment could enhance the homing ability of MSCs, thus alleviating AMI in rats. Therefore, UTMD combined with PDGF-BB pretreatment may offer exciting therapeutic opportunities for strengthening MSC therapy in ischemic diseases.
Collapse
Grants
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 2018CFB568 National Natural Science Foundation of Hubei
Collapse
Affiliation(s)
- Zhenxing Sun
- Anhui Medical University, Hefei, 230031, China
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yu Cai
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Ziming Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yuman Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Lei Huang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Zhengyang Han
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
| | - Xiangming Zhu
- Anhui Medical University, Hefei, 230031, China.
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| |
Collapse
|
7
|
Zhang H, Wan X, Tian J, An Z, Liu L, Zhao X, Zhou Y, Zhang L, Ge C, Song X. The therapeutic efficacy and clinical translation of mesenchymal stem cell-derived exosomes in cardiovascular diseases. Biomed Pharmacother 2023; 167:115551. [PMID: 37783149 DOI: 10.1016/j.biopha.2023.115551] [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: 06/19/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023] Open
Abstract
Exosomes, mainly derived from mesenchymal stem cells, provide a good reference for cardiac function repair and clinical application in cardiac and vascular diseases by regulating cardiomyocyte viability, inflammatory levels, angiogenesis, and ventricular remodeling after a heart injury. This review presents the cardioprotective efficacy of mesenchymal stem cell-originated exosomes and explores the underlying molecular mechanisms. Furthermore, we expound on several efficient approaches to transporting exosomes into the heart in clinical application and comment on the advantages and disadvantages of each method.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Xueqi Wan
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China; The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, PR China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Yuquan Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Lijun Zhang
- Department of Radiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China
| | - Changjiang Ge
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
8
|
Eisenbeis VB, Qiu D, Gorka O, Strotmann L, Liu G, Prucker I, Su XB, Wilson MSC, Ritter K, Loenarz C, Groß O, Saiardi A, Jessen HJ. β-lapachone regulates mammalian inositol pyrophosphate levels in an NQO1- and oxygen-dependent manner. Proc Natl Acad Sci U S A 2023; 120:e2306868120. [PMID: 37579180 PMCID: PMC10450438 DOI: 10.1073/pnas.2306868120] [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: 05/02/2023] [Accepted: 07/13/2023] [Indexed: 08/16/2023] Open
Abstract
Inositol pyrophosphates (PP-InsPs) are energetic signaling molecules with important functions in mammals. As their biosynthesis depends on ATP concentration, PP-InsPs are tightly connected to cellular energy homeostasis. Consequently, an increasing number of studies involve PP-InsPs in metabolic disorders, such as type 2 diabetes, aspects of tumorigenesis, and hyperphosphatemia. Research conducted in yeast suggests that the PP-InsP pathway is activated in response to reactive oxygen species (ROS). However, the precise modulation of PP-InsPs during cellular ROS signaling is unknown. Here, we report how mammalian PP-InsP levels are changing during exposure to exogenous (H2O2) and endogenous ROS. Using capillary electrophoresis electrospray ionization mass spectrometry (CE-ESI-MS), we found that PP-InsP levels decrease upon exposure to oxidative stressors in HCT116 cells. Application of quinone drugs, particularly β-lapachone (β-lap), under normoxic and hypoxic conditions enabled us to produce ROS in cellulo and to show that β-lap treatment caused PP-InsP changes that are oxygen-dependent. Experiments in MDA-MB-231 breast cancer cells deficient of NAD(P)H:quinone oxidoreductase-1 (NQO1) demonstrated that β-lap requires NQO1 bioactivation to regulate the cellular metabolism of PP-InsPs. Critically, significant reductions in cellular ATP concentrations were not directly mirrored in reduced PP-InsP levels as shown in NQO1-deficient MDA-MB-231 cells treated with β-lap. The data presented here unveil unique aspects of β-lap pharmacology and its impact on PP-InsP levels. The identification of different quinone drugs as modulators of PP-InsP synthesis will allow the overall impact on cellular function of such drugs to be better appreciated.
Collapse
Affiliation(s)
- Verena B. Eisenbeis
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Danye Qiu
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg79106, Germany
| | - Lisa Strotmann
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Guizhen Liu
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Isabel Prucker
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Xue Bessie Su
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, United Kingdom
| | - Miranda S. C. Wilson
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, United Kingdom
| | - Kevin Ritter
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Christoph Loenarz
- Faculty of Chemistry and Pharmacy, Institute for Pharmaceutical Sciences, Pharmaceutical and Medicinal Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| | - Olaf Groß
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg79106, Germany
| | - Adolfo Saiardi
- Medical Research Council, Laboratory for Molecular Cell Biology, University College London, WC1E 6BTLondon, United Kingdom
| | - Henning J. Jessen
- Faculty of Chemistry and Pharmacy, Institute of Organic Chemistry, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
- The Center for Integrative Biological Signaling Studies, Albert-Ludwigs-Universität Freiburg, Freiburg im Breisgau79104, Germany
| |
Collapse
|
9
|
Zhang T, Lu L, Li M, Zhang D, Yu P, Zhang X, Zhang Z, Lei C. Exosome from BMMSC Attenuates Cardiopulmonary Bypass-Induced Acute Lung Injury Via YAP/β-Catenin Pathway: Downregulation of Pyroptosis. Stem Cells 2022; 40:1122-1133. [PMID: 36063391 DOI: 10.1093/stmcls/sxac063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/22/2022] [Indexed: 01/04/2023]
Abstract
Acute lung injury (ALI) accompanied with systemic inflammatory response is an important complication after cardiopulmonary bypass (CPB). Pyroptosis, which is induced by the secretion of inflammatory factors, has been implicated in ALI. However, recent studies have suggested that bone marrow mesenchymal stem cell-derived exosomes (BMMSC-Exo) can ameliorate ALI, but the mechanism is poorly understood. Therefore, we aim to examine the effects of BMMSC-Exo in CPB-induced ALI, and its underlying mechanism. CPB rat models (male Sprague-Dawley rats) were administered BMMSC-Exo intravenously before induction of ALI. Lung tissue, bronchoalveolar lavage fluid (BALF), and alveolar macrophage (AM) were collected after the treatments for further analysis, and rat AM NR8383 cells were used for in vitro study. HE staining was performed to detect macrophage infiltration. Western blot was used to detect related proteins expression. And ELISA assay was performed to investigate secretion of inflammatory factors. These results showed that BMMSC-Exo treatment ameliorated macrophage infiltration and oxidative stress, and downregulated expression of pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and GSDMD-N, in the lung tissue and AM, as well as decreased the secretion of IL-18 and IL-1β in BALF. Moreover, BMMSC-Exo activated YAP/β-catenin signaling pathway. Overall, these findings of this study indicated that BMMSC-Exo suppressed CPB-induced pyroptosis in ALI by activating YAP/β-catenin axis, which could be a novel strategy for lung protection during CPB.
Collapse
Affiliation(s)
- Taoyuan Zhang
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.,Department of Anesthesiology, Rizhao International Heart Hospital, Rizhao, Shandong, People's Republic of China
| | - Linhe Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Man Li
- Central Medical Branch of PLA General Hospital, Beijing, People's Republic of China
| | - Ding Zhang
- Department of Anesthesiology, Rizhao International Heart Hospital, Rizhao, Shandong, People's Republic of China
| | - Peng Yu
- Department of Anesthesiology, Rizhao Traditional Chinese Medicine Hospital, Rizhao, Shandong, People's Republic of China
| | - Xinhao Zhang
- Department of Anesthesiology, Rizhao International Heart Hospital, Rizhao, Shandong, People's Republic of China
| | - Zheng Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Chong Lei
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
10
|
Inositol Alleviates Pulmonary Fibrosis by Promoting Autophagy via Inhibiting the HIF-1 α-SLUG Axis in Acute Respiratory Distress Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1030238. [PMID: 36589681 PMCID: PMC9803570 DOI: 10.1155/2022/1030238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/18/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022]
Abstract
The effective remission of acute respiratory distress syndrome- (ARDS-) caused pulmonary fibrosis determines the recovery of lung function. Inositol can relieve lung injuries induced by ARDS. However, the mechanism of myo-inositol in the development of ARDS is unclear, which limits its use in the clinic. We explored the role and mechanism of myo-inositol in the development of ARDS by using an in vitro lipopolysaccharide- (LPS-) established alveolar epithelial cell inflammation model and an in vivo ARDS mouse model. Our results showed that inositol can alleviate the progression of pulmonary fibrosis. More significantly, we found that inositol can induce autophagy to inhibit the progression pulmonary fibrosis caused by ARDS. In order to explore the core regulators of ARDS affected by inositol, mRNA-seq sequencing was performed. Those results showed that transcription factor HIF-1α can regulate the expression of SLUG, which in turn can regulate the key gene E-Cadherin involved in cell epithelial-mesenchymal transition (EMT) as well as N-cadherin expression, and both were regulated by inositol. Our results suggest that inositol activates autophagy to inhibit EMT progression induced by the HIF-1α/SLUG signaling pathway in ARDS, and thereby alleviates pulmonary fibrosis.
Collapse
|
11
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 314] [Impact Index Per Article: 104.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
12
|
Ning H, Chen H, Deng J, Xiao C, Xu M, Shan L, Yang C, Zhang Z. Exosomes secreted by FNDC5-BMMSCs protect myocardial infarction by anti-inflammation and macrophage polarization via NF-κB signaling pathway and Nrf2/HO-1 axis. Stem Cell Res Ther 2021; 12:519. [PMID: 34583757 PMCID: PMC8480009 DOI: 10.1186/s13287-021-02591-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/09/2021] [Indexed: 12/17/2022] Open
Abstract
Background Exosomes are considered a substitute for stem cell-based therapy for myocardial infarction (MI). FNDC5, a transmembrane protein located in the cytoplasm, plays a crucial role in inflammation diseases and MI repair. Furthermore, our previous study found that FNDC5 pre-conditioning bone marrow-derived mesenchymal stem cells (BMMSCs) could secrete more exosomes, but little was known on MI repair. Methods Exosomes isolated from BMMSCs with or without FNDC5-OV were injected into infarcted hearts. Then, cardiomyocytes apoptosis and inflammation responses were detected. Furthermore, exosomes were administrated to RAW264.7 macrophage with LPS treatment to investigate its effect on inflammation and macrophage polarization. Results Compared with MSCs-Exo, FNDC5-MSCs-Exo had superior therapeutic effects on anti-inflammation and anti-apoptosis, as well as polarizing M2 macrophage in vivo. Meanwhile, the in vitro results also showed that FNDC5-MSCs-Exo decreased pro-inflammatory secretion and increased anti-inflammatory secretion under LPS stimulation, which partly depressed NF‐κB signaling pathway and upregulated Nrf2/HO-1 Axis. Conclusions FNDC5-BMMSCs-derived exosomes play anti-inflammation effects and promote M2 macrophage polarization via NF-κB signaling pathway and Nrf2/HO-1 Axis, which may develop a promising cell-free therapy for MI. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02591-4.
Collapse
Affiliation(s)
- Hongjuan Ning
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Haixu Chen
- Institute of Geriatrics, Health Care Office, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jingyu Deng
- Air Force Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Chun Xiao
- PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Moyan Xu
- Institute of Geriatrics, Health Care Office, National Clinical Research Center of Geriatrics Disease, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lina Shan
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Chao Yang
- PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China.
| | - Zheng Zhang
- PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China.
| |
Collapse
|
13
|
Wu G, Chang F, Fang H, Zheng X, Zhuang M, Liu X, Hou W, Xu L, Chen Z, Tang C, Wu Y, Sun Y, Zhu F. Non-muscle myosin II knockdown improves survival and therapeutic effects of implanted bone marrow-derived mesenchymal stem cells in lipopolysaccharide-induced acute lung injury. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:262. [PMID: 33708889 PMCID: PMC7940885 DOI: 10.21037/atm-20-4851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Bone marrow-derived mesenchymal stem cells (BMSCs) have been shown to have some beneficial effects in acute lung injury (ALI), but the therapeutic effects are limited due to apoptosis or necrosis after transplantation into injured lungs. Here, we aim to explore whether Non-muscle myosin II (NM-II) knockdown could enhance BMSCs survival and improve therapeutic effects in ALI. Methods MSCs, isolated from rat bone marrow, were transfected with the small interfering RNA (siRNA) targeted to NM-II mRNA by a lentivirus vector. Rats were equally randomized to four groups: the control group was given normal saline via tail vein; the other three groups underwent intratracheal lipopolysaccharide (LPS) instillation followed by administration with either normal saline, BMSCs transduced with lentivirus-enhanced green fluorescent protein (eGFP) empty vector, or BMSCs transduced with lentivirus-eGFP NM-II siRNA. Hematoxylin and eosin staining was used to evaluate lung histopathologic changes and Masson trichrome staining was used to assess lung fibrosis. The myeloperoxidase activity was also tested in lung tissues. The mRNA expression of inflammatory cytokines in lung tissues was determined via quantitative reverse transcription PCR. Sex-determining region of the Y chromosome gene expression was measured by fluorescence in situ hybridization (FISH) assay. The expression of self-renewal activity and apoptosis-associated proteins were measured by Western blot. Results Transplantation of NM-II siRNA-modified BMSCs could improve histopathological morphology, decrease inflammatory infiltrates, down-regulate the expression levels of inflammatory cytokines, and reduce pulmonary interstitial edema. NM-II siRNA-modified BMSCs showed antifibrotic properties and alleviated the degrees of pulmonary fibrosis induced by endotoxin. In addition, NM-II knockdown BMSCs showed slightly better therapeutic effect on lung inflammation when compared with control BMSCs. The beneficial effects of NM-II siRNA-modified BMSCs may be attributed to enhanced self-renewal activity and decreased apoptosis. Conclusions NM-II knockdown could inhibit the apoptosis of implanted BMSCs in lung tissues and improve its self-renewal activity. NM-II siRNA-modified BMSCs have a slightly enhanced ability to attenuate lung injury after LPS challenge.
Collapse
Affiliation(s)
- Guosheng Wu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Fei Chang
- Department of Burn and Plastic Surgery, The Affiliated Zhang Jiagang Hospital of Soochow University, Suzhou, China
| | - He Fang
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xingfeng Zheng
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mingzhu Zhuang
- Clinical BioBank, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaobin Liu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wenjia Hou
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Long Xu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhengli Chen
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chenqi Tang
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yu Wu
- Department of Anesthesiology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Yu Sun
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Feng Zhu
- Department of Burn Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Chen Y, Zhou F, Liu H, Li J, Che H, Shen J, Luo E. SIRT1, a promising regulator of bone homeostasis. Life Sci 2021; 269:119041. [PMID: 33453243 DOI: 10.1016/j.lfs.2021.119041] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 12/16/2022]
Abstract
Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide-dependent deacetylase, epigenetically regulates various cell metabolisms, including inflammation, tumorigenesis, and bone metabolism. Many clinical studies have found the potential of SIRT1 in predicting and treating bone-related disorders, such as osteoporosis and osteonecrosis, suggesting that SIRT1 might be a regulator of bone homeostasis. In order to identify the mechanisms that underlie the pivotal role of SIRT1 in bone homeostasis, many studies revealed that SIRT1 could maintain the balance between bone formation and absorption via regulating the ratio of osteoblasts to osteoclasts. SIRT1 controls the differentiation of mesenchymal stem cells (MSCs) and bone marrow-derived macrophages, increasing osteogenesis and reducing osteoclastogenesis. Besides, SIRT1 can enhance bone-forming cells' viability, including MSCs and osteoblasts under adverse conditions by resisting senescence, suppressing apoptosis, and promoting autophagy in favor of osteogenesis. Furthermore, the effect on bone vasculature homeostasis enables SIRT1 to become a valuable strategy for ischemic osteonecrosis and senile osteoporosis. The review systemically discusses SIRT1 pathways and the critical role in bone homeostasis and assesses whether SIRT1 is a potential target for manipulation and therapy, to lay a solid foundation for further researches in the future.
Collapse
Affiliation(s)
- Ye Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA
| | - Jiaxuan Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Huiling Che
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiaqi Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - En Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
15
|
Li X, Xu J, Dai B, Wang X, Guo Q, Qin L. Targeting autophagy in osteoporosis: From pathophysiology to potential therapy. Ageing Res Rev 2020; 62:101098. [PMID: 32535273 DOI: 10.1016/j.arr.2020.101098] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Osteoporosis is a highly prevalent disorder characterized by the loss of bone mass and microarchitecture deterioration of bone tissue, attributed to various factors, including menopause (primary), aging (primary) and adverse effects of relevant medications (secondary). In recent decades, knowledge regarding the etiological mechanisms underpinning osteoporosis emphasizes that bone cellular homeostasis, including the maintenance of cell functions, differentiation, and the response to stress, is tightly regulated by autophagy, which is a cell survival mechanism for eliminating and recycling damaged proteins and organelles. With the important roles in the maintenance of cellular homeostasis and organ function, autophagy has emerged as a potential target for the prevention and treatment of osteoporosis. In this review, we update and discuss the pathophysiology of autophagy in normal bone cell life cycle and metabolism. Then, the alternations of autophagy in primary and secondary osteoporosis, and the accompanied pathological process are discussed. Finally, we discuss current strategies, limitations, and challenges involved in targeting relevant pathways and propose strategies by which such hurdles may be circumvented in the future for their translation into clinical validations and applications for the prevention and treatment of osteoporosis.
Collapse
|
16
|
Deng J, Zhang N, Chen F, Yang C, Ning H, Xiao C, Sun K, Liu Y, Yang M, Hu T, Zhang Z, Jiang W. Irisin ameliorates high glucose-induced cardiomyocytes injury via AMPK/mTOR signal pathway. Cell Biol Int 2020; 44:2315-2325. [PMID: 32770767 DOI: 10.1002/cbin.11441] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/29/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022]
Abstract
High glucose (HG)-induced cardiomyocytes (CMs) injury is a leading cause of diabetic cardiomyopathy with little treatment options. Irisin, a new myokine, which is cleaved from its precursor fibronectin type III domain-containing protein 5 (FNDC5), has aroused great attention as an essential cardioprotective factor and glucose metabolism regulator but little was known on diabetic cardiomyopathy yet. Here, we aim to clarify the role of irisin in the HG-induced CMs injury. Neonatal Sprague-Dawley rat CMs were cultured in a normal or HG medium for 12, 24, and 48 hr, respectively before exposing to irisin. The apoptosis level was determined by terminal-deoxynucleotidyl transferase-mediated-dUTP nick end-labeling assay. Cell viability was measured with the conventional methyl thiazolyl tetrazolium assay. Moreover, reactive oxygen species production was evaluated by dihydroethidium staining. Inflammatory factors, namely tumor necrosis factor-α, interleukin-6, interleukin-1β were determined by enzyme-linked immunosorbent assay kits. Furthermore, protein and messenger RNA (mRNA) expressions were measured by western blot and quantitative real-time polymerase chain reaction, respectively. HG increases the apoptosis of CMs and activated the inflammatory responses and oxidative stress in CMs. Meanwhile, the mRNA and protein expressions of FNDC5 are decreased after HG exposure. Nevertheless, the increased apoptosis is alleviated by irisin treatment. Notably, irisin suppresses the inflammatory responses and oxidative stress in injured CMs. Mechanically, after the administration of Compound C, AMP-activated protein kinase (AMPK) inhibitor, these cardioprotective effects resulting from irisin are reversed. Irisin plays a significant role in antiapoptosis, anti-inflammation, antioxidative stress in HG-induced CMs via AMPK/mammalian target of the rapamycin signaling pathway.
Collapse
Affiliation(s)
- Jingyu Deng
- Department of Cardiology, Postgraduate Training Base in PLA Rocket Force Characteristic Medical Center, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ning Zhang
- Department of surgery, Central Medical District of Chinese PLA General Hospital, Beijing, China
| | - Feng Chen
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Chao Yang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hongjuan Ning
- Department of Respiratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chun Xiao
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ke Sun
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yongfei Liu
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Ming Yang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Taohong Hu
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zheng Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Wei Jiang
- Department of Cardiology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| |
Collapse
|
17
|
β-arrestin1 inhibits hypoxic injury-induced autophagy in human pulmonary artery endothelial cells via the Akt/mTOR signaling pathway. Int J Biochem Cell Biol 2020; 125:105791. [PMID: 32544529 DOI: 10.1016/j.biocel.2020.105791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/20/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022]
Abstract
Autophagy has been greatly implicated in injured endothelial cells during pulmonary arterial hypertension (PAH). β-arrestin1, a multifunctional cytoplasmic protein, has attracted considerable attention as an essential protective factor in PAH. However, its role in autophagy of injured pulmonary arterial endothelial cells (PAECs) remains to be determined. Here, we investigated the potential effects of β-arrestin1 on autophagy and apoptosis in human PAECs (hPAECs) under hypoxic stress. Hypoxic stimuli increases autophagy and decreases the level of β-arrestin1 in hPAECs. Furthermore, pathologic changes, namely increased proliferation, migration, and apoptosis resistance, are observed after hypoxia exposure. These are reversed after β-arrestin1 overexpression (β-arrestin1-OV) or treatment with 3-MA, an autophagy inhibitor. Finally, β-arrestin1 suppresses the increase in autophagy and apoptosis resistance of hypoxic hPAECs. Mechanistically, β-arrestin1 upregulates the activity of the Akt/mTOR signaling pathway and downregulates the expression of BNIP3 and Nix after hypoxic stress. Collectively, we have demonstrated, for the first time, that β-arrestin1 reduces excessive autophagy and apoptosis resistance by activating the Akt/mTOR axis in hypoxic hPAECs. This knowledge suggests a promising therapeutic target for PAH.
Collapse
|
18
|
Deng J, Zhang N, Wang Y, Yang C, Wang Y, Xin C, Zhao J, Jin Z, Cao F, Zhang Z. FNDC5/irisin improves the therapeutic efficacy of bone marrow-derived mesenchymal stem cells for myocardial infarction. Stem Cell Res Ther 2020; 11:228. [PMID: 32522253 PMCID: PMC7288492 DOI: 10.1186/s13287-020-01746-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/08/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background The beneficial functions of bone marrow mesenchymal stem cells (BM-MSCs) decline with decreased cell survival, limiting their therapeutic efficacy for myocardial infarction (MI). Irisin, a novel myokine which is cleaved from its precursor fibronectin type III domain-containing protein 5 (FNDC5), is believed to be involved in a cardioprotective effect, but little was known on injured BM-MSCs and MI repair yet. Here, we investigated whether FNDC5 or irisin could improve the low viability of transplanted BM-MSCs and increase their therapeutic efficacy after MI. Methods BM-MSCs, isolated from dual-reporter firefly luciferase and enhanced green fluorescent protein positive (Fluc+–eGFP+) transgenic mice, were exposed to normoxic condition and hypoxic stress for 12 h, 24 h, and 48 h, respectively. In addition, BM-MSCs were treated with irisin (20 nmol/L) and overexpression of FNDC5 (FNDC5-OV) in serum deprivation (H/SD) injury. Furthermore, BM-MSCs were engrafted into infarcted hearts with or without FNDC5-OV. Results Hypoxic stress contributed to increased apoptosis, decreased cell viability, and paracrine effects of BM-MSCs while irisin or FNDC5-OV alleviated these injuries. Longitudinal in vivo bioluminescence imaging and immunofluorescence results illustrated that BM-MSCs with overexpression of FNDC5 treatment (FNDC5-MSCs) improved the survival of transplanted BM-MSCs, which ameliorated the increased apoptosis and decreased angiogenesis of BM-MSCs in vivo. Interestingly, FNDC5-OV elevated the secretion of exosomes in BM-MSCs. Furthermore, FNDC5-MSC therapy significantly reduced fibrosis and alleviated injured heart function. Conclusions The present study indicated that irisin or FNDC5 improved BM-MSC engraftment and paracrine effects in infarcted hearts, which might provide a potential therapeutic target for MI.
Collapse
Affiliation(s)
- Jingyu Deng
- Department of Cardiology, Postgraduate Training Base in PLA Rocket Force Characteristic Medical Center, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Ning Zhang
- Central Beijing Medical District, Chinese PLA General Hospital, Fuxing-Road, Haidian, Beijing, 100853, China
| | - Yong Wang
- Department of Nuclear Medicine, the Fifth Medical Center,, Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing, 100071, China
| | - Chao Yang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Yabin Wang
- National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Chao Xin
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Jinming Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zhitao Jin
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Feng Cao
- National Clinical Research Center for Geriatric Diseases, Second Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Zheng Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China.
| |
Collapse
|
19
|
Inositol Pyrophosphates: Signaling Molecules with Pleiotropic Actions in Mammals. Molecules 2020; 25:molecules25092208. [PMID: 32397291 PMCID: PMC7249018 DOI: 10.3390/molecules25092208] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Inositol pyrophosphates (PP-IPs) such as 5-diphosphoinositol pentakisphosphate (5-IP7) are inositol metabolites containing high-energy phosphoanhydride bonds. Biosynthesis of PP-IPs is mediated by IP6 kinases (IP6Ks) and PPIP5 kinases (PPIP5Ks), which transfer phosphate to inositol hexakisphosphate (IP6). Pleiotropic actions of PP-IPs are involved in many key biological processes, including growth, vesicular remodeling, and energy homeostasis. PP-IPs function to regulate their target proteins through allosteric interactions or protein pyrophosphorylation. This review summarizes the current understanding of how PP-IPs control mammalian cellular signaling networks in physiology and disease.
Collapse
|
20
|
Mukherjee S, Haubner J, Chakraborty A. Targeting the Inositol Pyrophosphate Biosynthetic Enzymes in Metabolic Diseases. Molecules 2020; 25:molecules25061403. [PMID: 32204420 PMCID: PMC7144392 DOI: 10.3390/molecules25061403] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
In mammals, a family of three inositol hexakisphosphate kinases (IP6Ks) synthesizes the inositol pyrophosphate 5-IP7 from IP6. Genetic deletion of Ip6k1 protects mice from high fat diet induced obesity, insulin resistance and fatty liver. IP6K1 generated 5-IP7 promotes insulin secretion from pancreatic β-cells, whereas it reduces insulin signaling in metabolic tissues by inhibiting the protein kinase Akt. Thus, IP6K1 promotes high fat diet induced hyperinsulinemia and insulin resistance in mice while its deletion has the opposite effects. IP6K1 also promotes fat accumulation in the adipose tissue by inhibiting the protein kinase AMPK mediated energy expenditure. Genetic deletion of Ip6k3 protects mice from age induced fat accumulation and insulin resistance. Accordingly, the pan IP6K inhibitor TNP [N2-(m-trifluorobenzyl), N6-(p-nitrobenzyl)purine] ameliorates obesity, insulin resistance and fatty liver in diet induced obese mice by improving Akt and AMPK mediated insulin sensitivity and energy expenditure. TNP also protects mice from bone loss, myocardial infarction and ischemia reperfusion injury. Thus, the IP6K pathway is a potential target in obesity and other metabolic diseases. Here, we summarize the studies that established IP6Ks as a potential target in metabolic diseases. Further studies will reveal whether inhibition of this pathway has similar pleiotropic benefits on metabolic health of humans.
Collapse
|
21
|
Mu M, Zhao H, Wang Y, Liu J, Fei D, Xing M. Arsenic trioxide or/and copper sulfate co-exposure induce glandular stomach of chicken injury via destruction of the mitochondrial dynamics and activation of apoptosis as well as autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 185:109678. [PMID: 31557571 DOI: 10.1016/j.ecoenv.2019.109678] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/27/2019] [Accepted: 09/12/2019] [Indexed: 06/10/2023]
Abstract
Arsenic and copper are naturally occurring element. Contamination from natural processes and anthropogenic activities can be discovered all over the world and their unique interactions with the environment lead to widespread toxicity. When the content was excessive, the organism would be hurt seriously. The glandular stomach is an important organ of the poultry gastrointestinal tract. This study was aimed to investigate the toxicity of arsenic trioxide or/and copper sulfate (As or/and Cu) on chicken glandular stomach. Seventy-two 1-day-old Hy-Line chickens were randomly divided into control (C) group, arsenic trioxide (As) group, copper sulfate (Cu) group and arsenic trioxide and copper sulfate (AsCu) group, and exposed to 30 mg/kg arsenic trioxide or/and 300 mg/kg copper sulphates for 12 weeks. The indicators of mitochondrial dynamics, apoptosis and autophagy were tested in the glandular stomach. The results showed that exposure to As or/and Cu caused mitochondrial dynamic imbalance. Additionally, the levels of pro-apoptosis and autophagy indicators were increased and the levels of anti-apoptosis indicators were decreased in the treatment groups. Beyond that, in the treatment groups, we could clearly see karyopyknosis and chromatin condensation were associated with increased apoptosis rate, as well as the disappearance of the nuclear membrane, the swelling of mitochondria and the accumulation of autophagosomes were involved in the death of cells. It was worth noting that the glandular stomach lesions were time-dependent, and the combination of As and Cu were worse than the As and Cu alone. Collectively, our results suggest that As or/and Cu aggravate mitochondrial dysfunction, apoptosis and autophagy in a time-dependent manner, and the combined toxicity of As and Cu was higher.
Collapse
Affiliation(s)
- Mengyao Mu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Yu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Juanjuan Liu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Dongxue Fei
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, 150040, Heilongjiang, PR China.
| |
Collapse
|
22
|
Wang Z, Tang J, Li Y, Wang Y, Guo Y, Tu Q, Chen J, Wang C. AdipoRon promotes diabetic fracture repair through endochondral ossification-based bone repair by enhancing survival and differentiation of chondrocytes. Exp Cell Res 2019; 387:111757. [PMID: 31838062 DOI: 10.1016/j.yexcr.2019.111757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022]
Abstract
Diabetic bone defects may exhibit impaired endochondral ossification (ECO) leading to delayed bone repair. AdipoRon, a receptor agonist of adiponectin polymers, can ameliorate diabetes and related complications, as well as overcome the disadvantages of the unstable structure of artificial adiponectin polymers. Here, the effects of AdipoRon on the survival and differentiation of chondrocytes in a diabetic environment were explored focusing on related mechanisms in gene and protein levels. In vivo, AdipoRon was applied to diet-induced-obesity (DIO) mice, a model of obesity and type 2 diabetes, with femoral fracture. Sequential histological evaluations and micro-CT were examined for further verification. We found that AdipoRon could ameliorate cell viability, apoptosis, and reactive oxygen species (ROS) production and promote mRNA expression of chondrogenic markers and cartilaginous matrix production of ATDC5 cells in high glucose medium via activating ERK1/2 pathway. Additionally, DIO mice with intragastric AdipoRon administration had more neocartilage and accelerated new bone formation. These data suggest that AdipoRon could stimulate bone regeneration via ECO in diabetes.
Collapse
Affiliation(s)
- Zhongyi Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Jinxin Tang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Ying Li
- Department of Stomatology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, China
| | - Yu Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Yanyang Guo
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Qisheng Tu
- Tufts School of Dental Medicine, Sackler School of Graduate Biomedical Sciences, Tufts School of Medicine, Boston, 02111, USA
| | - Jake Chen
- Tufts School of Dental Medicine, Sackler School of Graduate Biomedical Sciences, Tufts School of Medicine, Boston, 02111, USA.
| | - Chen Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|