1
|
Zhang J, Xie Z, Zhu X, Xu C, Lin J, Zhao M, Cheng Y. New insights into therapeutic strategies for targeting hepatic macrophages to alleviate liver fibrosis. Int Immunopharmacol 2025; 158:114864. [PMID: 40378438 DOI: 10.1016/j.intimp.2025.114864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Liver fibrosis is a wound-healing response induced by persistent liver damage, resulting from complex multicellular interactions and multifactorial networks. Without intervention, it can progress to cirrhosis and even liver cancer. Current understanding suggests that liver fibrosis is reversible, making it crucial to explore effective therapeutic strategies for its alleviation. Chronic inflammation serves as the primary driver of liver fibrosis, with hepatic macrophages playing a dual role depending on their polarization state. This review summarizes various prevention and therapeutic strategies targeting hepatic macrophages in the context of liver fibrosis. These strategies include inhibition of macrophage recruitment, modulation of macrophage activation and polarization, regulation of macrophage metabolism, and induction of phagocytosis and autophagy in hepatic macrophages. Additionally, we discuss the communication between hepatic macrophages, hepatocytes, and hepatic stellate cells (HSCs), as well as the current clinical application of anti-fibrotic drugs targeting macrophages. The goal is to identify effective therapeutic targets at each stage of macrophage participation in liver fibrosis development, with the aim of using hepatic macrophages as a target for liver fibrosis treatment.
Collapse
Affiliation(s)
- Jialu Zhang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Zhaojing Xie
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Xueyu Zhu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Chenxi Xu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Jiguo Lin
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Mingqi Zhao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
2
|
Cheng X, Kang L, Liu J, Wang Q, Zhang Z, Zhang L, Xie Y, Chang L, Zeng D, Tian L, Zhang L, Xu P, Li Y. Proteomics and phosphoproteomics revealed dysregulated kinases and potential therapy for liver fibrosis. Mol Cell Proteomics 2025:100991. [PMID: 40368138 DOI: 10.1016/j.mcpro.2025.100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025] Open
Abstract
Liver fibrosis is the initial stage of most liver diseases, and it is also a pathological process involving the liver in the late stages of many metabolic diseases. Therefore, it is important to systematically understand the pathological mechanism of liver fibrosis and seek therapeutic approaches for intervention and treatment of liver fibrosis. Disordered proteins and their post-translational modifications, such as phosphorylation, play vital roles in the occurrence and development of liver fibrosis. However, the regulatory mechanisms that govern this process remain poorly understood. In this study, we analyzed and quantified the liver proteome and phosphoproteome of CCl4-induced early liver fibrosis model in mice. Proteomic analysis revealed that the pathways involved in extracellular matrix (ECM) recombination, collagen formation, metabolism and other related disorders, and protein phosphorylation modification pathways were also significantly enriched. In addition, western blotting and phosphoproteomics demonstrated that phosphorylation levels were elevated in the context of liver fibrosis. A total of 13,152 phosphosites were identified, with 952 sites increased while only 156 ones decreased. Furthermore, the upregulated phosphorylation sites, which exhibited no change at the proteome level mainly shared a common [xxxSPxxx] motif. Consequently, the kinases-substrates analysis ascertained the overactive kinases of these up-regulated substrates, which ultimately led to the identification of 13 significantly altered kinases within this dataset. These kinases were mainly catalogued into the STE, CMGC, and CAMK kinase families. Among them, STK4, GSK3α and CDK11B were subsequently validated though cellular and animal experiments, and the results demonstrated that their inhibitors could effectively reduce the activation of hepatic stellate cells and ECM production. These kinases may represent potential therapeutic targets for liver fibrosis, and their inhibitors may serve as promising anti- hepatic fibrosis drugs.
Collapse
Affiliation(s)
- Xinyu Cheng
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Li Kang
- School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Jinfang Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, P. R. China
| | - Qingye Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Zhenpeng Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Li Zhang
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Yuping Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Lei Chang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China
| | - Daobing Zeng
- General Surgery Department, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, P. R. China
| | - Lingqiang Zhang
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China.
| | - Ping Xu
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China; College of Life Sciences, Hebei University, 071002 Baoding, China; TaiKang Medical School (School of Basic Medical Sciences), Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, Wuhan University, Wuhan, P. R. China.
| | - Yanchang Li
- Anhui Medical University School of Basic Medicine, Anhui, P. R. China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing, P. R. China; College of Life Sciences, Hebei University, 071002 Baoding, China.
| |
Collapse
|
3
|
Zhang X, Xiong W, Gao F, Yu Z, Ren F, Lei XG. Impacts and mechanism of liver-specific knockout of selenoprotein I on hepatic phospholipid metabolism, selenogenome expression, redox status, and resistance to CCl 4 toxicity. Free Radic Biol Med 2025; 235:426-442. [PMID: 40345504 DOI: 10.1016/j.freeradbiomed.2025.05.387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/11/2025]
Abstract
Selenoprotein I (SELENOI) was known initially as ethanolamine phosphotransferase 1 (EPT1) and later as a selenoprotein. Because global knockout of Selenoi in mice is embryonically lethal, we generated liver-specific Selenoi knockout (cKO) mice to reveal functions and mechanism of SELENOI in the liver. Compared with control mice, cKO mice (8 weeks old) had no differences in body weight, glucose metabolism, energy expenditure, overall health status, or liver histology. However, these mice had lower (P < 0.05) mRNA levels of 13 selenoprotein genes, contents of Se, GSH, and T-AOC (12-40%), and activities of antioxidant enzymes (17-51%), but higher (P < 0.05) mRNA levels of oxidative stress-related genes (34%-46%) in the liver than the control mice. They had a higher (P < 0.05) ratio of phosphatidylcholine (PC) to phosphatidylethanolamine (PE) due to increases of the former and decreases of the latter, altered PE and PC constituents such as n-6/n-3 PUFA ratios, and elevated mRNA levels (95%-2-fold, P < 0.05) of lipolysis genes, compared with the control mice. The knockout attenuated hepatic injury and fibrosis induced by 14 intraperitoneal injections of CCl4 (0.5 mL/kg). The protection was associated with adaptive cytoprotective mechanisms induced by the overall decline of redox status mediated by SELENOI as a selenoprotein and activations of PPAR signaling, fatty acid desaturase 2 (FADS2), glutathione S-transferase, and lipid peroxide hydrolysis through modulating biosynthesis and(or) constituents of PC, PE, and n-6/n-3 PUFAs mediated by SELENOI as EPT1. Inhibition of FADS2 in CCl4-treated cKO hepatocytes partially removed the protection by the knockout. In conclusion, hepatic SELENOI expression was not essential for survival, but served as a multifunctional regulator of hepatic selenogenome expression, Se metabolism, redox status, biosyntheses and profiles of PC and PE, and resistance to CCI4.
Collapse
Affiliation(s)
- Xu Zhang
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Wei Xiong
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China; Food Laboratory of Zhongyuan, Luohe, Henan, 462300, China.
| | - Fei Gao
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhengquan Yu
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Fazheng Ren
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
4
|
Wu L, Li Z, Wang K, Groleau RR, Rong X, Liu X, Liu C, Lewis SE, Zhu B, James TD. Advances in Organic Small Molecule-Based Fluorescent Probes for Precision Detection of Liver Diseases: A Perspective on Emerging Trends and Challenges. J Am Chem Soc 2025; 147:9001-9018. [PMID: 40036086 PMCID: PMC11926879 DOI: 10.1021/jacs.4c17092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
Liver disease poses a significant challenge to global health, and its early diagnosis is crucial for improving treatment outcomes and patient prognosis. Since fluctuation of key biomarkers during the onset and progression of liver diseases can directly reflect liver health and normal/abnormal function, biomarker-based assays are vital tools for the early detection of liver disease. In this context, small molecule fluorescent probes have undeniably emerged as indispensable tools for diagnosis and analysis, with an ever-growing number of small molecule-based fluorescent probes being developed over recent years, with the sole aim of monitoring relevant biomarkers of liver disease. This perspective will focus on the development and application of probes developed primarily over the last 10 years for diagnosing a range liver disease-related processes. It will outline the foundational design strategies for developing promising probes, their optical response to key biomarkers, and how they have been demonstrated in proof-of-concept imaging applications. Current challenges and new developments in the field will be discussed, with the aim of providing insights and highlighting opportunities in the field.
Collapse
Affiliation(s)
- Luling Wu
- Department
of Chemistry, University of Bath, Bath BA2 7AY, U.K.
| | - Zilu Li
- School
of Water Conservancy and Environment, University
of Jinan, Jinan 250022, China
| | - Kun Wang
- School
of Water Conservancy and Environment, University
of Jinan, Jinan 250022, China
| | - Robin R. Groleau
- Department
of Life Sciences, University of Bath, Bath BA2 7AY, U.K.
| | - Xiaodi Rong
- School
of Water Conservancy and Environment, University
of Jinan, Jinan 250022, China
| | - Xueting Liu
- School
of Water Conservancy and Environment, University
of Jinan, Jinan 250022, China
| | - Caiyun Liu
- School
of Water Conservancy and Environment, University
of Jinan, Jinan 250022, China
| | - Simon E. Lewis
- Department
of Chemistry, University of Bath, Bath BA2 7AY, U.K.
| | - Baocun Zhu
- School
of Water Conservancy and Environment, University
of Jinan, Jinan 250022, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, U.K.
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, People’s
Republic of China
| |
Collapse
|
5
|
Loaiza-Moss J, Braun U, Leitges M. Transcriptome Analysis Suggests PKD3 Regulates Proliferative Glucose Metabolism, Calcium Homeostasis and Microtubule Dynamics After MEF Spontaneous Immortalization. Int J Mol Sci 2025; 26:596. [PMID: 39859313 PMCID: PMC11765705 DOI: 10.3390/ijms26020596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Cell immortalization corresponds to a biologically relevant clinical feature that allows cells to acquire a high proliferative potential during carcinogenesis. In multiple cancer types, Protein Kinase D3 (PKD3) has often been reported as a dysregulated oncogenic kinase that promotes cell proliferation. Using mouse embryonic fibroblasts (MEFs), in a spontaneous immortalization model, PKD3 has been demonstrated as a critical regulator of cell proliferation after immortalization. However, the mechanisms by which PKD3 regulates proliferation in immortalized MEFs require further elucidation. Using a previously validated Prkd3-deficient MEF model, we performed a poly-A transcriptomic analysis to identify putative Prkd3-regulated biological processes and downstream targets in MEFs after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were identified and further analyzed by gene ontology (GO) enrichment and protein-protein interaction (PPI) network analyses to identify potential hub genes. Our results suggest that Prkd3 modulates proliferation through the regulation of gene expression associated with glucose metabolism (Tnf, Ucp2, Pgam2, Angptl4), calcium homeostasis and transport (Calcr and P2rx7) and microtubule dynamics (Stmn2 and Map10). These candidate processes and associated genes represent potential mechanisms involved in Prkd3-induced proliferation in spontaneously immortalized cells as well as clinical targets in several cancer types.
Collapse
Affiliation(s)
| | | | - Michael Leitges
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. Johns, NL A1B 3V6, Canada; (J.L.-M.); (U.B.)
| |
Collapse
|
6
|
Han H, Liu H, Steiner R, Zhao Z, Jin ZG. Inhibition of protein kinase D and its substrate phosphatidylinositol-4 kinase III beta blocks common human coronavirus replication. Microbiol Spectr 2024; 12:e0150124. [PMID: 39540754 PMCID: PMC11619529 DOI: 10.1128/spectrum.01501-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the infection of a coronavirus, named as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coronaviruses can be replicated in the infected host cells. Coronavirus replication involves various steps, including membrane fusion, peri-nuclear particle formation, and matrix vesicle transport to the cell membrane via the endoplasmic reticulum-Golgi-lysosome route. Recent studies have suggested that protein kinase D (PKD) plays a crucial role in regulation of vesicle formation and trafficking in the trans-Golgi network (TGN). Thus, we hypothesize that inhibiting PKD and its associated pathway could be an effective strategy to limit viral replication. Here, we report that molecular and pharmacological inhibition of PKD and its substrate phosphatidylinositol-4 kinase III beta (PI4KIIIβ) significantly diminishes the replication of common human coronaviruses. Specifically, we found that the PKD-silencing siRNA and the PKD inhibitor CRT0066101 have broad-spectrum antiviral activity against HCoV-OC43, HCoV-NL63, and HCoV-229E in cultured cells. Mechanistic studies revealed that the deactivation of PKD reduced the activation of PI4KIIIβ, thereby blocking the transport of viral particles in the host cells. Furthermore, the PI4KIIIβ inhibitor, BQR695, also exhibited antiviral activity against those coronaviruses. In conclusion, PKD and its substrate, PI4KIIIβ, may serve as novel antiviral targets for human coronaviruses and warrant further investigation. IMPORTANCE Human coronaviruses can lead to a range of clinical symptoms, from asymptomatic infection to severe illness and death, with a limited array of antiviral drugs available. Protein kinase D (PKD) is involved in various cellular processes, such as cell proliferation, apoptosis, and membrane fission of the Golgi apparatus. However, the specific role of PKD in the human coronavirus life cycle remains unclear. In this study, we found that PKD inhibitors effectively attenuated human coronavirus replication at the trans-Golgi network (TGN) stage in the viral life cycle. Furthermore, inhibiting PKD reduced PI4KIIIβ activation, thereby blocking viral replication in the host cells. Importantly, PI4KIIIβ inhibitors also blocked human coronavirus replication. Thus, PKD may represent a promising therapeutic target against both current circulating and future emerging coronaviruses.
Collapse
Affiliation(s)
- Huijuan Han
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Zhijun Zhao
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zheng-Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
7
|
Liu H, Sun L, Zhao H, Zhao Z, Zhang S, Jiang S, Cheng T, Wang X, Wang T, Shao Y, Zhu H, Han H, Cao Y, Jiang E, Cao Y, Xu Y. Proteinase 3 depletion attenuates leukemia by promoting myeloid differentiation. Cell Death Differ 2024; 31:697-710. [PMID: 38589495 PMCID: PMC11165011 DOI: 10.1038/s41418-024-01288-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) that have impaired differentiation can transform into leukemic blasts. However, the mechanism that controls differentiation remains elusive. Here, we show that the genetic elimination of Proteinase 3 (PRTN3) in mice led to spontaneous myeloid differentiation. Mechanistically, our findings indicate that PRTN3 interacts with the N-terminal of STAT3, serving as a negative regulator of STAT3-dependent myeloid differentiation. Specifically, PRTN3 promotes STAT3 ubiquitination and degradation, while simultaneously reducing STAT3 phosphorylation and nuclear translocation during G-CSF-stimulated myeloid differentiation. Strikingly, pharmacological inhibition of STAT3 (Stattic) partially counteracted the effects of PRTN3 deficiency on myeloid differentiation. Moreover, the deficiency of PRTN3 in primary AML blasts promotes the differentiation of those cells into functional neutrophils capable of chemotaxis and phagocytosis, ultimately resulting in improved overall survival rates for recipients. These findings indicate PRTN3 exerts an inhibitory effect on STAT3-dependent myeloid differentiation and could be a promising therapeutic target for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Huan Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Lu Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Hongfei Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Zihan Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shiyue Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shan Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tianran Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaohan Wang
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Tong Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Ya Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Haiyan Zhu
- Department of Clinical Lab, Weihai Municipal Hospital, Weihai, 264200, China
| | - Huijuan Han
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750001, China
| | - Yigeng Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, 17165, Sweden.
| | - Yuanfu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
8
|
Liu J, Liu R, Wang H, Zhang Z, Wang J, Wei F. CircPRKD3/miR-6783-3p responds to mechanical force to facilitate the osteogenesis of stretched periodontal ligament stem cells. J Orthop Surg Res 2024; 19:257. [PMID: 38649946 PMCID: PMC11036753 DOI: 10.1186/s13018-024-04727-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The mechanotransduction mechanisms by which cells regulate tissue remodeling are not fully deciphered. Circular RNAs (circRNAs) are crucial to various physiological processes, including cell cycle, differentiation, and polarization. However, the effects of mechanical force on circRNAs and the role of circRNAs in the mechanobiology of differentiation and remodeling in stretched periodontal ligament stem cells (PDLSCs) remain unclear. This article aims to explore the osteogenic function of mechanically sensitive circular RNA protein kinase D3 (circPRKD3) and elucidate its underlying mechanotransduction mechanism. MATERIALS AND METHODS PDLSCs were elongated with 8% stretch at 0.5 Hz for 24 h using the Flexcell® FX-6000™ Tension System. CircPRKD3 was knockdown or overexpressed with lentiviral constructs or plasmids. The downstream molecules of circPRKD3 were predicted by bioinformatics analysis. The osteogenic effect of related molecules was evaluated by quantitative real-time PCR (qRT-PCR) and western blot. RESULTS Mechanical force enhanced the osteogenesis of PDLSCs and increased the expression of circPRKD3. Knockdown of circPRKD3 hindered PDLSCs from osteogenesis under mechanical force, while overexpression of circPRKD3 promoted the early osteogenesis process of PDLSCs. With bioinformatics analysis and multiple software predictions, we identified hsa-miR-6783-3p could act as the sponge of circPRKD3 to indirectly regulate osteogenic differentiation of mechanically stimulated PDLSCs. CONCLUSIONS Our results first suggested that both circPRKD3 and hsa-miR-6783-3p could enhance osteogenesis of stretched PDLSCs. Furthermore, hsa-miR-6783-3p could sponge circPRKD3 to indirectly regulate RUNX2 during the periodontal tissue remodeling process in orthodontic treatment.
Collapse
Affiliation(s)
- Jiani Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Rui Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Hong Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Zijie Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Jixiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, Shandong, 250012, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, Shandong, 250012, China.
| |
Collapse
|
9
|
Luo J, Liu H, Xu Y, Yu N, Steiner RA, Wu X, Si S, Jin ZG. Hepatic Sirt6 activation abrogates acute liver failure. Cell Death Dis 2024; 15:283. [PMID: 38649362 PMCID: PMC11035560 DOI: 10.1038/s41419-024-06537-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 04/25/2024]
Abstract
Acute liver failure (ALF) is a deadly illness due to insufficient detoxification in liver induced by drugs, toxins, and other etiologies, and the effective treatment for ALF is very limited. Among the drug-induced ALF, acetaminophen (APAP) overdose is the most common cause. However, the molecular mechanisms underlying APAP hepatoxicity remain incompletely understood. Sirtuin 6 (Sirt6) is a stress responsive protein deacetylase and plays an important role in regulation of DNA repair, genomic stability, oxidative stress, and inflammation. Here, we report that genetic and pharmacological activation of Sirt6 protects against ALF in mice. We first observed that Sirt6 expression was significantly reduced in the liver tissues of human patients with ALF and mice treated with an overdose of APAP. Then we developed an inducible Sirt6 transgenic mice for Cre-mediated overexpression of the human Sirt6 gene in systemic (Sirt6-Tg) and hepatic-specific (Sirt6-HepTg) manners. Both Sirt6-Tg mice and Sirt6-HepTg mice exhibited the significant protection against APAP hepatoxicity. In contrast, hepatic-specific Sirt6 knockout mice exaggerated APAP-induced liver damages. Mechanistically, Sirt6 attenuated APAP-induced hepatocyte necrosis and apoptosis through downregulation of oxidative stress, inflammation, the stress-activated kinase JNK activation, and apoptotic caspase activation. Moreover, Sirt6 negatively modulated the level and activity of poly (ADP-ribose) polymerase 1 (PARP1) in APAP-treated mouse liver tissues. Importantly, the specific Sirt6 activator MDL-800 exhibited better therapeutic potential for APAP hepatoxicity than the current drug acetylcysteine. Furthermore, in the model of bile duct ligation induced ALF, hepatic Sirt6-KO exacerbated, but Sirt6-HepTg mitigated liver damage. Collectively, our results demonstrate that Sirt6 protects against ALF and suggest that targeting Sirt6 activation could be a new therapeutic strategy to alleviate ALF.
Collapse
Affiliation(s)
- Jinque Luo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Tiantan Xili, Beijing, 100050, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yanni Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Tiantan Xili, Beijing, 100050, China
| | - Nanhui Yu
- The 2nd Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Rebbeca A Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
| | - Xiaoqian Wu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou, China
| | - Shuyi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), No. 1 Tiantan Xili, Beijing, 100050, China.
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA.
| |
Collapse
|
10
|
Liu S, Yin Y, Liu S, Wang C, Sun W, Hu X. Shining a light on liver health: advancements in fluorescence-enhanced enzyme biosensors for early disease detection. Front Bioeng Biotechnol 2024; 12:1392857. [PMID: 38707500 PMCID: PMC11066187 DOI: 10.3389/fbioe.2024.1392857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/05/2024] [Indexed: 05/07/2024] Open
Abstract
Early detection of liver diseases holds paramount importance in optimizing treatment outcomes and prognosis, thereby significantly enhancing the likelihood of recovery while mitigating the risk of progression to liver cancer. Liver diseases encompass a spectrum of conditions, each potentially manifesting distinct enzymatic profiles. Monitoring these enzymes in situ facilitates timely intervention and therapeutic management. In recent years, the field of biosensor technology has witnessed remarkable advancement, owing to strides in biomedicine and computational sciences. Biosensors have garnered widespread utility across medical and biological domains, spanning the detection of disease biomarkers, drug release tracking, ion imaging, and fluorescence imaging within living organisms. These applications have markedly enhanced imaging resolution and have the potential to refine disease diagnosis accuracy for clinicians. A pivotal aspect in the successful application of this technology lies in the construction of fluorescence probes adept at swiftly and selectively identifying target enzymes by amalgamating liver disease enzymes with fluorescence probe technology. However, research in this niche area remains relatively scarce. Building upon this foundational understanding, the present review delineates the utilization of biosensors in the early diagnosis of liver disease. Serving as a theoretical framework, this review envisages the development of high-performance biosensors tailored for the early detection of liver cancer. Furthermore, it offers insights into the potential of biosensor technology to progress and broaden its practical applications, thus contributing to the advancement of diagnostic methodologies in liver disease management.
Collapse
Affiliation(s)
- Shifeng Liu
- Department of the Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yatong Yin
- Qingdao Maternal and Child Health and Family Planning Service Center, Qingdao, China
| | - Shihai Liu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Congxiao Wang
- Department of the Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenshe Sun
- Qingdao Cancer Institute, Qingdao University, Qingdao, China
| | - Xiaokun Hu
- Department of the Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Gutiérrez-Galindo E, Yilmaz ZH, Hausser A. Membrane trafficking in breast cancer progression: protein kinase D comes into play. Front Cell Dev Biol 2023; 11:1173387. [PMID: 37293129 PMCID: PMC10246754 DOI: 10.3389/fcell.2023.1173387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Protein kinase D (PKD) is a serine/threonine kinase family that controls important cellular functions, most notably playing a key role in the secretory pathway at the trans-Golgi network. Aberrant expression of PKD isoforms has been found mainly in breast cancer, where it promotes various cellular processes such as growth, invasion, survival and stem cell maintenance. In this review, we discuss the isoform-specific functions of PKD in breast cancer progression, with a particular focus on how the PKD controlled cellular processes might be linked to deregulated membrane trafficking and secretion. We further highlight the challenges of a therapeutic approach targeting PKD to prevent breast cancer progression.
Collapse
Affiliation(s)
| | - Zeynep Hazal Yilmaz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
12
|
Shao M, Wang Y, Dong H, Wang L, Zhang X, Han X, Sang X, Bao Y, Peng M, Cao G. From liver fibrosis to hepatocarcinogenesis: Role of excessive liver H2O2 and targeting nanotherapeutics. Bioact Mater 2023; 23:187-205. [PMID: 36406254 PMCID: PMC9663332 DOI: 10.1016/j.bioactmat.2022.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/23/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022] Open
Abstract
Liver fibrosis and hepatocellular carcinoma (HCC) have been worldwide threats nowadays. Liver fibrosis is reversible in early stages but will develop precancerosis of HCC in cirrhotic stage. In pathological liver, excessive H2O2 is generated and accumulated, which impacts the functionality of hepatocytes, Kupffer cells (KCs) and hepatic stellate cells (HSCs), leading to genesis of fibrosis and HCC. H2O2 accumulation is associated with overproduction of superoxide anion (O2•−) and abolished antioxidant enzyme systems. Plenty of therapeutics focused on H2O2 have shown satisfactory effects against liver fibrosis or HCC in different ways. This review summarized the reasons of liver H2O2 accumulation, and the role of H2O2 in genesis of liver fibrosis and HCC. Additionally, nanotherapeutics targeting H2O2 were summarized for further consideration of antifibrotic or antitumor therapy. Liver fibrosis and HCC are closely related because ROS induced liver damage and inflammation, especially over-cumulated H2O2. Excess H2O2 diffusion in pathological liver was due to increased metabolic rate and diminished cellular antioxidant systems. Freely diffused H2O2 damaged liver-specific cells, thereby leading to fibrogenesis and hepatocarcinogenesis. Nanotherapeutics targeting H2O2 are summarized for treatment of liver fibrosis and HCC, and also challenges are proposed.
Collapse
|
13
|
Sun Y, Weng J, Chen X, Ma S, Zhang Y, Zhang F, Zhang Z, Wang F, Shao J, Zheng S. Oroxylin A activates ferritinophagy to induce hepatic stellate cell senescence against hepatic fibrosis by regulating cGAS-STING pathway. Biomed Pharmacother 2023; 162:114653. [PMID: 37086511 DOI: 10.1016/j.biopha.2023.114653] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/24/2023] Open
Abstract
In recent study, the pathological mechanism of liver fibrosis has been associated with hepatic stellate cell (HSC) senescence. Targeted induction of HSC senescence is considered as a new strategy to remove activated HSC. Nevertheless, little is known about the role of ferritinophagy in cell senescence. In this study, we reported that Oroxylin A from Scutellaria baicalensis Georgi can regulate HSC senescence induced by ferritinophagy through the cGAS-STING pathway to reduce liver fibrosis. We first found that Oroxylin A treatment alleviated the pathological changes of liver fibrosis, reduced collagen deposition, and significantly inhibited liver fibrosis. Interestingly, Oroxylin A treatment can activate HSC ferritinophagy and further induce HSC senescence. It is noteworthy that ferritinophagy is mediated by nuclear receptor coactivator 4 (NCOA4), an important selective mediator for ferritin degradation. NCOA4 siRNA causes Oroxylin A to reduce the degree of telomerase activity in HSCs and induce the expression of senescence markers, such as SA-β-Gal and related marker proteins. Importantly, the cGAS-STING pathway is crucial to the activation of HSC ferritinophagy by Oroxylin A. Specifically, Oroxylin A can promote the secretion of cytokines like IFN-β by the cGAS-STING pathway to regulate ferritinophagy. cGAS siRNA resulted in a dose-dependent decrease in the expression of NCOA4, a significant reduction in the expression level of autophagy-related phenotype, and a decrease in the content of ROS and iron ions in HSCs. In conclusion, we identified the new role of ferritinophagy and the GAS-STING pathway in Oroxylin A -mediated anti-hepatic fibrosis.
Collapse
Affiliation(s)
- Ying Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingdan Weng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaolei Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuyao Ma
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuxin Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
14
|
Burciaga SD, Saavedra F, Fischer L, Johnstone K, Jensen ED. Protein kinase D3 conditional knockout impairs osteoclast formation and increases trabecular bone volume in male mice. Bone 2023; 172:116759. [PMID: 37044359 DOI: 10.1016/j.bone.2023.116759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Studies using kinase inhibitors have shown that the protein kinase D (PRKD) family of serine/threonine kinases are required for formation and function of osteoclasts in culture. However, the involvement of individual protein kinase D genes and their in vivo significance to skeletal dynamics remains unclear. In the current study we present data indicating that protein kinase D3 is the primary form of PRKD expressed in osteoclasts. We hypothesized that loss of PRKD3 would impair osteoclast formation, thereby decreasing bone resorption and increasing bone mass. Conditional knockout (cKO) of Prkd3 using a murine Cre/Lox system driven by cFms-Cre revealed that its loss in osteoclast-lineage cells reduced osteoclast differentiation and resorptive function in culture. Examination of the Prkd3 cKO mice showed that bone parameters were unaffected in the femur at 4 weeks of age, but consistent with our hypothesis, Prkd3 conditional knockout resulted in 18 % increased trabecular bone mass in male mice at 12 weeks and a similar increase at 6 months. These effects were not observed in female mice. As a further test of our hypothesis, we asked if Prkd3 cKO could protect against bone loss in a ligature-induced periodontal disease model but did not see any reduction in bone destruction in this system. Together, our data indicate that PRKD3 promotes osteoclastogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Samuel D Burciaga
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Flavia Saavedra
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Lori Fischer
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Karen Johnstone
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA
| | - Eric D Jensen
- Department of Diagnostic & Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN 55455, USA.
| |
Collapse
|
15
|
Veazey JM, Wong GS, Eliseeva SI, Smyth TR, Chapman TJ, Lim K, Kim M, Georas SN. Protein kinase D3 promotes neutrophil migration during viral infection. Immunol Cell Biol 2023; 101:130-141. [PMID: 36318273 PMCID: PMC10112008 DOI: 10.1111/imcb.12603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/13/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
Protein kinase D (PKD) is a serine/threonine kinase family with three isoforms (PKD1-3) that are expressed in most cells and implicated in a wide array of signaling pathways, including cell growth, differentiation, transcription, secretion, polarization and actin turnover. Despite growing interest in PKD, relatively little is known about the role of PKD in immune responses. We recently published that inhibiting PKD limits proinflammatory cytokine secretion and leukocyte accumulation in mouse models of viral infection, and that PKD3 is highly expressed in the murine lung and immune cell populations. Here we focus on the immune-related phenotypes of PKD3 knockout mice. We report that PKD3 is necessary for maximal neutrophil accumulation in the lung following challenge with inhaled polyinosinic:polycytidylic acid, a double-stranded RNA, as well as following influenza A virus infection. Using reciprocal bone marrow chimeras, we found that PKD3 is required in the hematopoietic compartment for optimal neutrophil migration to the lung. Ex vivo transwell and chemokinesis assays confirmed that PKD3-/- neutrophils possess an intrinsic motility defect, partly because of reduced surface expression of CD18, which is critical for leukocyte migration. Finally, the peak of neutrophilia was significantly reduced in PKD3-/- mice after lethal influenza A virus infection. Together, these results demonstrate that PKD3 has an essential, and nonredundant, role in promoting neutrophil recruitment to the lung. A better understanding of the isoform-specific and cell type-specific activities of PKD has the potential to lead to novel therapeutics for respiratory illnesses.
Collapse
Affiliation(s)
- Janelle M Veazey
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Gordon S Wong
- Department of Medicine, Yale New Haven Health, Greenwich Hospital, Greenwich, CT, USA
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
| | - Sophia I Eliseeva
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
| | - Timothy R Smyth
- Department of Toxicology, University of North Caroline, Chapel Hill, NC, USA
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Timothy J Chapman
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
- Merck, Kenilworth, NJ, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Steve N Georas
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Rochester, NY, USA
| |
Collapse
|
16
|
Chen L, Zhao J, Chao Y, Roy A, Guo W, Qian J, Xu W, Domsic RT, Lafyatis R, Lu B, Deng F, Wang QJ. Loss of Protein Kinase D2 Activity Protects Against Bleomycin-Induced Dermal Fibrosis in Mice. J Transl Med 2023; 103:100018. [PMID: 37039152 PMCID: PMC10507682 DOI: 10.1016/j.labinv.2022.100018] [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: 05/14/2022] [Revised: 08/22/2022] [Accepted: 09/20/2022] [Indexed: 01/11/2023] Open
Abstract
Protein kinase D (PKD) has been linked to inflammatory responses in various pathologic conditions; however, its role in inflammation-induced dermal fibrosis has not been evaluated. In this study, we aimed to investigate the roles and mechanisms of protein kinase D2 (PKD2) in inflammation-induced dermal fibrosis and evaluate the therapeutic potential of PKD inhibitors in this disease. Using homozygous kinase-dead PKD2 knock-in (KI) mice, we examined whether genetic ablation or pharmacologic inhibition of PKD2 activity affected dermal inflammation and fibrosis in a bleomycin (BLM)-induced skin fibrosis model. Our data showed that dermal thickness and collagen fibers were significantly reduced in BLM-treated PKD2 KI mice compared with that in wild-type mice, and so was the expression of α-smooth muscle actin and collagens and the mRNA levels of transforming growth factor-β1 and interleukin-6 in the KI mice. Corroboratively, pharmacologic inhibition of PKD by CRT0066101 also significantly blocked BLM-induced dermal fibrosis and reduced α-smooth muscle actin, collagen, and interleukin-6 expression. Further analyses indicated that loss of PKD2 activity significantly blocked BLM-induced infiltration of monocytes/macrophages and neutrophils in the dermis. Moreover, using bone marrow-derived macrophages, we demonstrated that PKD activity was required for cytokine production and migration of macrophages. We have further identified Akt as a major downstream target of PKD2 in the early inflammatory phase of the fibrotic process. Taken together, our findings indicate that PKD2 promotes dermal fibrosis via regulating immune cell infiltration, cytokine production, and downstream activation of Akt in lesional skin, and targeted inhibition of PKD2 may benefit the treatment of this condition.
Collapse
Affiliation(s)
- Liping Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinjun Zhao
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Wenjing Guo
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiabi Qian
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wanfu Xu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Robyn T Domsic
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Lafyatis
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
17
|
Varga A, Nguyen MT, Pénzes K, Bátai B, Gyulavári P, Gurbi B, Murányi J, Csermely P, Csala M, Vántus T, Sőti C. Protein Kinase D3 (PKD3) Requires Hsp90 for Stability and Promotion of Prostate Cancer Cell Migration. Cells 2023; 12:cells12020212. [PMID: 36672148 PMCID: PMC9857065 DOI: 10.3390/cells12020212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/12/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Prostate cancer metastasis is a significant cause of mortality in men. PKD3 facilitates tumor growth and metastasis, however, its regulation is largely unclear. The Hsp90 chaperone stabilizes an array of signaling client proteins, thus is an enabler of the malignant phenotype. Here, using different prostate cancer cell lines, we report that Hsp90 ensures PKD3 conformational stability and function to promote cancer cell migration. We found that pharmacological inhibition of either PKDs or Hsp90 dose-dependently abrogated the migration of DU145 and PC3 metastatic prostate cancer cells. Hsp90 inhibition by ganetespib caused a dose-dependent depletion of PKD2, PKD3, and Akt, which are all involved in metastasis formation. Proximity ligation assay and immunoprecipitation experiments demonstrated a physical interaction between Hsp90 and PKD3. Inhibition of the chaperone-client interaction induced misfolding and proteasomal degradation of PKD3. PKD3 siRNA combined with ganetespib treatment demonstrated a specific involvement of PKD3 in DU145 and PC3 cell migration, which was entirely dependent on Hsp90. Finally, ectopic expression of PKD3 enhanced migration of non-metastatic LNCaP cells in an Hsp90-dependent manner. Altogether, our findings identify PKD3 as an Hsp90 client and uncover a potential mechanism of Hsp90 in prostate cancer metastasis. The molecular interaction revealed here may regulate other biological and pathological functions.
Collapse
Affiliation(s)
- Attila Varga
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (A.V.); (C.S.)
| | - Minh Tu Nguyen
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Kinga Pénzes
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- Institute of Medical Microbiology, Semmelweis University, 1089 Budapest, Hungary
| | - Bence Bátai
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- HCEMM-SU Molecular Oncohematology Research Group, Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Pál Gyulavári
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
- IQVIA Hungary, 1117 Budapest, Hungary
| | - Bianka Gurbi
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - József Murányi
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Péter Csermely
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Tibor Vántus
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- MTA-SE Pathobiochemistry Research Group, Semmelweis University, 1094 Budapest, Hungary
| | - Csaba Sőti
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (A.V.); (C.S.)
| |
Collapse
|
18
|
Wu X, Liu H, Brooks A, Xu S, Luo J, Steiner R, Mickelsen DM, Moravec CS, Jeffrey AD, Small EM, Jin ZG. SIRT6 Mitigates Heart Failure With Preserved Ejection Fraction in Diabetes. Circ Res 2022; 131:926-943. [PMID: 36278398 PMCID: PMC9669223 DOI: 10.1161/circresaha.121.318988] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 10/13/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a growing health problem without effective therapies. Epidemiological studies indicate that diabetes is a strong risk factor for HFpEF, and about 45% of patients with HFpEF are suffering from diabetes, yet the underlying mechanisms remain elusive. METHODS Using a combination of echocardiography, hemodynamics, RNA-sequencing, molecular biology, in vitro and in vivo approaches, we investigated the roles of SIRT6 (sirtuin 6) in regulation of endothelial fatty acid (FA) transport and HFpEF in diabetes. RESULTS We first observed that endothelial SIRT6 expression was markedly diminished in cardiac tissues from heart failure patients with diabetes. We then established an experimental mouse model of HFpEF in diabetes induced by a combination of the long-term high-fat diet feeding and a low-dose streptozocin challenge. We also generated a unique humanized SIRT6 transgenic mouse model, in which a single copy of human SIRT6 transgene was engineered at mouse Rosa26 locus and conditionally induced with the Cre-loxP technology. We found that genetically restoring endothelial SIRT6 expression in the diabetic mice ameliorated diastolic dysfunction concurrently with decreased cardiac lipid accumulation. SIRT6 gain- or loss-of-function studies showed that SIRT6 downregulated endothelial FA uptake. Mechanistically, SIRT6 suppressed endothelial expression of PPARγ through SIRT6-dependent deacetylation of histone H3 lysine 9 around PPARγ promoter region; and PPARγ reduction mediated SIRT6-dependent inhibition of endothelial FA uptake. Importantly, oral administration of small molecule SIRT6 activator MDL-800 to diabetic mice mitigated cardiac lipid accumulation and diastolic dysfunction. CONCLUSIONS The impairment of endothelial SIRT6 expression links diabetes to HFpEF through the alteration of FA transport across the endothelial barrier. Genetic and pharmacological strategies that restored endothelial SIRT6 function in mice with diabetes alleviated experimental HFpEF by limiting FA uptake and improving cardiac metabolism, thus warranting further clinical evaluation.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Huan Liu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Alan Brooks
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jinque Luo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Deanne M. Mickelsen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christine S. Moravec
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Alexis D. Jeffrey
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Eric M. Small
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
19
|
You Y, Gao C, Wu J, Qu H, Xiao Y, Kang Z, Li J, Hong J. Enhanced Expression of ARK5 in Hepatic Stellate Cell and Hepatocyte Synergistically Promote Liver Fibrosis. Int J Mol Sci 2022; 23:ijms232113084. [PMID: 36361872 PMCID: PMC9655442 DOI: 10.3390/ijms232113084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 11/22/2022] Open
Abstract
AMPK-related protein kinase 5 (ARK5) is involved in a broad spectrum of physiological and cell events, and aberrant expression of ARK5 has been observed in a wide variety of solid tumors, including liver cancer. However, the role of ARK5 in liver fibrosis remains largely unexplored. We found that ARK5 expression was elevated in mouse fibrotic livers, and showed a positive correlation with the progression of liver fibrosis. ARK5 was highly expressed not only in activated hepatic stellate cells (HSCs), but also in hepatocytes. In HSCs, ARK5 prevents the degradation of transforming growth factor β type I receptor (TβRI) and mothers against decapentaplegic homolog 4 (Smad4) proteins by inhibiting the expression of Smad ubiquitin regulatory factor 2 (Smurf2), thus maintaining the continuous transduction of the transforming growth factor β (TGF-β) signaling pathway, which is essential for cell activation, proliferation and survival. In hepatocytes, ARK5 induces the occurrence of epithelial-mesenchymal transition (EMT), and also promotes the secretion of inflammatory factors. Inflammatory factors, in turn, further enhance the activation of HSCs and deepen the degree of liver fibrosis. Notably, we demonstrated in a mouse model that targeting ARK5 with the selective inhibitor HTH-01-015 attenuates CCl4-induced liver fibrosis in mice. Taken together, the results indicate that ARK5 is a critical driver of liver fibrosis, and promotes liver fibrosis by synergy between HSCs and hepatocytes.
Collapse
Affiliation(s)
- Yang You
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
| | - Chongqing Gao
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
| | - Junru Wu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
| | - Hengdong Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
| | - Yang Xiao
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
- Department of Hepatological Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Ziwei Kang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
| | - Jinying Li
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510630, China
- Department of Hepatological Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
- Correspondence: ; Tel.: +86-20-8522-0253
| |
Collapse
|
20
|
Clusterin negatively modulates mechanical stress-mediated ligamentum flavum hypertrophy through TGF-β1 signaling. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1549-1562. [PMID: 36131026 PMCID: PMC9534863 DOI: 10.1038/s12276-022-00849-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/20/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022]
Abstract
Ligamentum flavum hypertrophy (LFH) is a major cause of lumbar spinal canal stenosis (LSCS). The pathomechanisms for LFH have not been fully elucidated. Isobaric tags for relative and absolute quantitation (iTRAQ) technology, proteomics assessments of human ligamentum flavum (LF), and successive assays were performed to explore the effect of clusterin (CLU) upregulation on LFH pathogenesis. LFH samples exhibited higher cell positive rates of the CLU, TGF-β1, α-SMA, ALK5 and p-SMAD3 proteins than non-LFH samples. Mechanical stress and TGF-β1 initiated CLU expression in LF cells. Notably, CLU inhibited the expression of mechanical stress-stimulated and TGF-β1-stimulated COL1A2 and α-SMA. Mechanistic studies showed that CLU inhibited mechanical stress-stimulated and TGF-β1-induced SMAD3 activities through suppression of the phosphorylation of SMAD3 and by inhibiting its nuclear translocation by competitively binding to ALK5. PRKD3 stabilized CLU protein by inhibiting lysosomal distribution and degradation of CLU. CLU attenuated mechanical stress-induced LFH in vivo. In summary, the findings showed that CLU attenuates mechanical stress-induced LFH by modulating the TGF-β1 pathways in vitro and in vivo. These findings imply that CLU is induced by mechanical stress and TGF-β1 and inhibits LF fibrotic responses via negative feedback regulation of the TGF-β1 pathway. These findings indicate that CLU is a potential treatment target for LFH. The protein clusterin regulates the body’s response to lower back pain induced by mechanical stress and could be a target for treatments. Lower back pain is common and is exacerbated by our upright stance. A major cause of the pain is excessive cell growth (hypertrophy) in the ligaments between vertebrae. This growth narrows the spinal canal and compresses nerves. Using a unique mouse model bred to walk upright, Zhongmin Zhang and Liang Wang at Southern Medical University in Guangzhou, China, and co-workers showed that clusterin, a protein involved in regulation of cell survival, can reduce the hypertrophy caused by mechanical stresses, and could be used in back pain treatments. Clusterin regulates the activity of the growth factor TGF-β1, which plays a role in synthesizing new tissues after injury, but can spur excessive growth.
Collapse
|
21
|
Nie G, Zhou Y, Song M, Xu J, Cui Z, Feng Y, Wang H, Chen D, Zhang Y, Wang K. NIR-II imaging-guided diagnosis and evaluation of the therapeutic effect on acute alcoholic liver injury via a nanoprobe. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:1847-1855. [PMID: 35412537 DOI: 10.1039/d2ay00279e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Acute alcoholic liver injury (AALI) is hard to diagnose on account of no obvious clinical symptoms, and thereby it easily develops into serious liver diseases and threatens people's health. However, traditional methods for detecting AALI are far from satisfactory due to the low sensitivity, invasiveness and non-visualization, and the development of new techniques is in urgent demand. Near-infrared (NIR)-II fluorescence imaging has been widely studied in biochemistry and biomedicine. As the blood flow velocity of the liver is closely related to the progression of AALI, herein, a NIR-II fluorescent nanoprobe, NTPB-NPs, was applied to diagnose AALI by monitoring the fluorescence intensity changes in the liver caused by the variations of the blood flow velocity. More importantly, when medication was applied to alleviate the liver injury of AALI mice, NTPB-NPs could also track the therapeutic effect in situ. In this study, the relationship between hepatic vascular velocity and the progression of AALI was confirmed with NTPB-NPs via NIR-II imaging. To the best of our knowledge, this is the first time that a NIR-II fluorescence imaging technique has been used to diagnose AALI mice and evaluate the therapeutic effect on AALI mice. This study may also provide a potential NIR-II imaging agent for clinical research to improve the management of liver injury related diseases.
Collapse
Affiliation(s)
- Gang Nie
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| | - Yinxing Zhou
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| | - Mengzi Song
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| | - Jingya Xu
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| | - Zheng Cui
- Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yangzhen Feng
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China.
| | - Huiling Wang
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, Chemical Biology Center, College of Chemistry, International Joint Research Center for Intelligent Biosensing Technology and Health, Central China Normal University, Wuhan, China
| | - Dugang Chen
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China.
| | - Yu Zhang
- Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Kaiping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
Koutník J, Neururer V, Gruber T, Peer S, Hermann-Kleiter N, Olson WJ, Labi V, Leitges M, Baier G, Siegmund K. Addressing the role of PKD3 in the T cell compartment with knockout mice. Cell Commun Signal 2022; 20:54. [PMID: 35440091 PMCID: PMC9020081 DOI: 10.1186/s12964-022-00864-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Protein kinase D3 (PKD3) has been implicated in signal transduction downstream of the T cell receptor (TCR). However, its role for the activation of primary T lymphocytes has not been elucidated so far. METHODS Expression of PKD isoforms in primary murine T cells was determined by RT-PCR and SDS-Page. A germline PKD3-knockout mouse line was analyzed for its immune response to OVA/alum intraperitoneal immunization. Phenotyping of the T cell compartment ex vivo as well as upon stimulation in vitro was performed by flow cytometry. Additionally, cytokine expression was assessed by flow cytometry, RT-PCR and Luminex technology. RESULTS PKD expression in T cells is modulated by TCR stimulation, leading to a rapid down-regulation on mRNA and on protein level. PKD3-deficient mice respond to immunization with enhanced T follicular helper cell generation. Furthermore, peripheral PKD3-deficient CD4+ T cells express more interleukin-2 than wild type CD4+ T cells upon TCR stimulation ex vivo. However, purified naïve CD4+ T cells do not differ in their phenotype upon differentiation in vitro from wild type T cells. Moreover, we observed a shift towards an effector/memory phenotype of splenic T cells at steady state, which might explain the contradictory results obtained with pan-T cells ex vivo and naïve-sorted T cells. CONCLUSION While PKD3-deficiency in vivo in mice leads to a skewing of the T cell compartment towards a more activated phenotype, this kinase seems to be dispensable for naïve CD4+ T cell differentiation in vitro. Video Abstract.
Collapse
Affiliation(s)
- Jiří Koutník
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria
| | - Verena Neururer
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria
- Apoptosis, Cancer, and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon1, 69008, Lyon, France
| | - Thomas Gruber
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastian Peer
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria
| | | | - William J Olson
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Verena Labi
- Institute of Developmental Immunology, Medical University Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Michael Leitges
- Division of BioMedical Sciences, Faculty of Medicine, Craig L Dobbin Genetics Research Centre, Memorial University of Newfoundland Health Science Centre, 300 Prince Philip Drive, St. John's, NF, A1B 3V6, Canada
| | - Gottfried Baier
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria
| | - Kerstin Siegmund
- Institute of Cell Genetics, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
23
|
Cheng D, Chai J, Wang H, Fu L, Peng S, Ni X. Hepatic macrophages: Key players in the development and progression of liver fibrosis. Liver Int 2021; 41:2279-2294. [PMID: 33966318 DOI: 10.1111/liv.14940] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/15/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022]
Abstract
Hepatic fibrosis is a common pathological process involving persistent liver injury with various etiologies and subsequent inflammatory responses that occur in chronic liver diseases. If left untreated, liver fibrosis can progress to liver cirrhosis, hepatocellular carcinoma and eventually, liver failure. Unfortunately, to date, there is no effective treatment for liver fibrosis, with the exception of liver transplantation. Although the pathophysiology of liver fibrosis is multifactorial and includes the activation of hepatic stellate cells, which are known to drive liver fibrogenesis, hepatic macrophages have emerged as central players in the development of liver fibrosis and regression. Hepatic macrophages, which consist of resident macrophages (Kupffer cells) and monocyte-derived macrophages, have been shown to play an intricate role in the initiation of inflammatory responses to liver injury, progression of fibrosis, and promotion of fibrosis resolution. These features have made hepatic macrophages uniquely attractive therapeutic targets in the fight against hepatic fibrosis. In this review, we synthesised the literature to highlight the functions and regulation of heterogeneity in hepatic macrophages. Furthermore, using the existing findings, we attempt to offer insights into the molecular mechanisms underlying the phenotypic switch from fibrogenic macrophages to restorative macrophages, the regulation of heterogeneity, and modes of action for hepatic macrophages. A better understanding of these mechanisms may guide the development of novel anti-fibrotic therapies (eg macrophage subset-targeted treatments) to combat liver fibrosis in the future.
Collapse
Affiliation(s)
- Da Cheng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
| | - Jin Chai
- Cholestatic Liver Diseases Center, Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huiwen Wang
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
- International Collaborative Research Center for Medical Metabolomics, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
24
|
The PKD-Dependent Biogenesis of TGN-to-Plasma Membrane Transport Carriers. Cells 2021; 10:cells10071618. [PMID: 34203456 PMCID: PMC8303525 DOI: 10.3390/cells10071618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 01/30/2023] Open
Abstract
Membrane trafficking is essential for processing and transport of proteins and lipids and to establish cell compartmentation and tissue organization. Cells respond to their needs and control the quantity and quality of protein secretion accordingly. In this review, we focus on a particular membrane trafficking route from the trans-Golgi network (TGN) to the cell surface: protein kinase D (PKD)-dependent pathway for constitutive secretion mediated by carriers of the TGN to the cell surface (CARTS). Recent findings highlight the importance of lipid signaling by organelle membrane contact sites (MCSs) in this pathway. Finally, we discuss our current understanding of multiple signaling pathways for membrane trafficking regulation mediated by PKD, G protein-coupled receptors (GPCRs), growth factors, metabolites, and mechanosensors.
Collapse
|
25
|
Loza-Valdes A, Mayer AE, Kassouf T, Trujillo-Viera J, Schmitz W, Dziaczkowski F, Leitges M, Schlosser A, Sumara G. A phosphoproteomic approach reveals that PKD3 controls PKA-mediated glucose and tyrosine metabolism. Life Sci Alliance 2021; 4:4/8/e202000863. [PMID: 34145024 PMCID: PMC8321662 DOI: 10.26508/lsa.202000863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase D3 (PKD3) regulates hepatic metabolism in a PKA-dependent manner and reveals many other putative PKD3 targets in the liver. Members of the protein kinase D (PKD) family (PKD1, 2, and 3) integrate hormonal and nutritional inputs to regulate complex cellular metabolism. Despite the fact that a number of functions have been annotated to particular PKDs, their molecular targets are relatively poorly explored. PKD3 promotes insulin sensitivity and suppresses lipogenesis in the liver of animals fed a high-fat diet. However, its substrates are largely unknown. Here we applied proteomic approaches to determine PKD3 targets. We identified more than 300 putative targets of PKD3. Furthermore, biochemical analysis revealed that PKD3 regulates cAMP-dependent PKA activity, a master regulator of the hepatic response to glucagon and fasting. PKA regulates glucose, lipid, and amino acid metabolism in the liver, by targeting key enzymes in the respective processes. Among them the PKA targets phenylalanine hydroxylase (PAH) catalyzes the conversion of phenylalanine to tyrosine. Consistently, we showed that PKD3 is activated by glucagon and promotes glucose and tyrosine levels in hepatocytes. Therefore, our data indicate that PKD3 might play a role in the hepatic response to glucagon.
Collapse
Affiliation(s)
- Angel Loza-Valdes
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany.,Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Alexander E Mayer
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Toufic Kassouf
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jonathan Trujillo-Viera
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Werner Schmitz
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Filip Dziaczkowski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Michael Leitges
- Tier 1, Canada Research Chair in Cell Signaling and Translational Medicine, Division of BioMedical Sciences/Faculty of Medicine, Craig L Dobbin Genetics Research Centre, Memorial University of Newfoundland, Health Science Centre, St. Johns, Canada
| | - Andreas Schlosser
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Grzegorz Sumara
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany .,Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
26
|
Trujillo‐Viera J, El‐Merahbi R, Schmidt V, Karwen T, Loza‐Valdes A, Strohmeyer A, Reuter S, Noh M, Wit M, Hawro I, Mocek S, Fey C, Mayer AE, Löffler MC, Wilhelmi I, Metzger M, Ishikawa E, Yamasaki S, Rau M, Geier A, Hankir M, Seyfried F, Klingenspor M, Sumara G. Protein Kinase D2 drives chylomicron-mediated lipid transport in the intestine and promotes obesity. EMBO Mol Med 2021; 13:e13548. [PMID: 33949105 PMCID: PMC8103097 DOI: 10.15252/emmm.202013548] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Lipids are the most energy-dense components of the diet, and their overconsumption promotes obesity and diabetes. Dietary fat content has been linked to the lipid processing activity by the intestine and its overall capacity to absorb triglycerides (TG). However, the signaling cascades driving intestinal lipid absorption in response to elevated dietary fat are largely unknown. Here, we describe an unexpected role of the protein kinase D2 (PKD2) in lipid homeostasis. We demonstrate that PKD2 activity promotes chylomicron-mediated TG transfer in enterocytes. PKD2 increases chylomicron size to enhance the TG secretion on the basolateral side of the mouse and human enterocytes, which is associated with decreased abundance of APOA4. PKD2 activation in intestine also correlates positively with circulating TG in obese human patients. Importantly, deletion, inactivation, or inhibition of PKD2 ameliorates high-fat diet-induced obesity and diabetes and improves gut microbiota profile in mice. Taken together, our findings suggest that PKD2 represents a key signaling node promoting dietary fat absorption and may serve as an attractive target for the treatment of obesity.
Collapse
Affiliation(s)
- Jonathan Trujillo‐Viera
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Rabih El‐Merahbi
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Vanessa Schmidt
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Till Karwen
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Angel Loza‐Valdes
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Akim Strohmeyer
- Chair for Molecular Nutritional MedicineTechnical University of MunichTUM School of Life Sciences WeihenstephanFreisingGermany
- EKFZ ‐ Else Kröner‐Fresenius‐Center for Nutritional MedicineTechnical University of MunichMunichGermany
- ZIEL ‐ Institute for Food & HealthTechnical University of MunichFreisingGermany
| | - Saskia Reuter
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Minhee Noh
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Magdalena Wit
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Izabela Hawro
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| | - Sabine Mocek
- Chair for Molecular Nutritional MedicineTechnical University of MunichTUM School of Life Sciences WeihenstephanFreisingGermany
- EKFZ ‐ Else Kröner‐Fresenius‐Center for Nutritional MedicineTechnical University of MunichMunichGermany
- ZIEL ‐ Institute for Food & HealthTechnical University of MunichFreisingGermany
| | - Christina Fey
- Fraunhofer Institute for Silicate Research (ISC)Translational Center Regenerative Therapies (TLC‐RT)WürzburgGermany
| | - Alexander E Mayer
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Mona C Löffler
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
| | - Ilka Wilhelmi
- Department of Experimental DiabetologyGerman Institute of Human Nutrition Potsdam‐RehbrueckeNuthetalGermany
- German Center for Diabetes Research (DZD)München‐NeuherbergGermany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research (ISC)Translational Center Regenerative Therapies (TLC‐RT)WürzburgGermany
| | - Eri Ishikawa
- Molecular ImmunologyResearch Institute for Microbial Diseases (RIMD)Osaka UniversitySuitaJapan
- Molecular ImmunologyImmunology Frontier Research Center (IFReC)Osaka UniversitySuitaJapan
| | - Sho Yamasaki
- Molecular ImmunologyResearch Institute for Microbial Diseases (RIMD)Osaka UniversitySuitaJapan
- Molecular ImmunologyImmunology Frontier Research Center (IFReC)Osaka UniversitySuitaJapan
| | - Monika Rau
- Division of HepatologyUniversity Hospital WürzburgWürzburgGermany
| | - Andreas Geier
- Division of HepatologyUniversity Hospital WürzburgWürzburgGermany
| | - Mohammed Hankir
- Department of General, Visceral, Transplant, Vascular and Pediatric SurgeryUniversity Hospital WürzburgWürzburgGermany
| | - Florian Seyfried
- Department of General, Visceral, Transplant, Vascular and Pediatric SurgeryUniversity Hospital WürzburgWürzburgGermany
| | - Martin Klingenspor
- Chair for Molecular Nutritional MedicineTechnical University of MunichTUM School of Life Sciences WeihenstephanFreisingGermany
- EKFZ ‐ Else Kröner‐Fresenius‐Center for Nutritional MedicineTechnical University of MunichMunichGermany
- ZIEL ‐ Institute for Food & HealthTechnical University of MunichFreisingGermany
| | - Grzegorz Sumara
- Rudolf‐Virchow‐ZentrumCenter for Integrative and Translational BioimagingUniversity of WürzburgWürzburgGermany
- Nencki Institute of Experimental BiologyPolish Academy of SciencesWarszawaPoland
| |
Collapse
|
27
|
Fan N, Wu C, Zhou Y, Wang X, Li P, Liu Z, Tang B. Rapid Two-Photon Fluorescence Imaging of Monoamine Oxidase B for Diagnosis of Early-Stage Liver Fibrosis in Mice. Anal Chem 2021; 93:7110-7117. [PMID: 33909401 DOI: 10.1021/acs.analchem.1c00815] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Liver fibrosis could induce cirrhosis and liver cancer, causing serious damages to liver function and even death. Early diagnosis of fibrosis is extremely requisite for optimizing treatment schedule to improve cure rate. In early-stage fibrosis, overexpressed monoamine oxidase B (MAO-B) can serve as a biomarker, which greatly contributes to the diagnosis of early liver fibrosis. However, there is still a lack of desired strategy to precisely monitor MAO-B in situ. In this work, we established a two-photon fluorescence imaging method for in vivo detection of MAO-B activity counting on a simply prepared probe, BiPhAA. The BiPhAA could be activated by MAO-B within 10 min and fluoresced brightly. To our knowledge, this BiPhAA-based imaging platform for MAO-B is more rapid than other current detection methods. Furthermore, BiPhAA allowed the dynamic observation of endogenous MAO-B level changes in hepatic stellate cells (LX-2). Through two-photon fluorescence imaging, we observed six times higher fluorescence brightness in the liver tissue of fibrosis mice than that of normal mice, thus successfully distinguishing mice with liver fibrosis from normal mice. Our work offers a simple, fast, and highly sensitive approach for imaging MAO-B in situ and paves a way to the diagnosis of early liver fibrosis with accuracy.
Collapse
Affiliation(s)
- Nannan Fan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| | - Chuanchen Wu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| | - Yongqing Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| | - Zhenzhen Liu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Science, Shandong Normal University, Jinan 250014, P.R. China
| |
Collapse
|
28
|
Chen L, Guo P, Li W, Fang F, Zhu W, Fan J, Wang F, Gao Y, Zhao Q, Wang Q, Xiao Y, Xing X, Li D, Shi T, Yu D, Aschner M, Zhang L, Chen W. Perturbation of Specific Signaling Pathways Is Involved in Initiation of Mouse Liver Fibrosis. Hepatology 2021; 73:1551-1569. [PMID: 32654205 DOI: 10.1002/hep.31457] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS To identify the regulatory role of protein phosphatase 2A (PP2A) in the development of liver disease, we generated a mouse model with hepatocyte-specific deletion of Ppp2r1a gene (encoding PP2A Aα subunit). APPROACH AND RESULTS Homozygote (HO) mice and matched wild-type littermates were investigated at 3, 6, 9, 12, 15, and 18 months of age. Pathological examination showed that PP2A Aα deficiency in hepatocytes resulted in progressive liver fibrosis phenotype from 9 months of age. No hepatocyte death was observed in HO mice. However, perturbation of pathways including epidermal growth factor receptor 1 (EGFR1), amino acid metabolism, and translation factors as well as leptin and adiponectin led to pronounced hepatic fibrosis. In vitro studies demonstrated the involvement of specific B subunit complexes in the regulation of EGFR1 signaling pathway and cross talk between defected hepatocytes and stimulation of interstitial hyperplasia. It is noteworthy that HO mice failed to develop hepatocellular carcinoma for as long as 22 months of age. We further demonstrate that PP2A Aβ-containing holoenzymes played a critical role in preventing hepatocyte apoptosis and antagonizing tumorigenesis through specific pathways on Aα loss. Furthermore, PP2A Aα and Aβ were functionally distinct, and the Aβ isoform failed to substitute for Aα in the development of inflammation and liver fibrosis. CONCLUSIONS These observations identify pathways that contribute to the pathogenesis of liver fibrosis and provide putative therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Liping Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ping Guo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Wenxue Li
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Fei Fang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, National Chromatographic Research and Analysis Center, Dalian, China
| | - Wei Zhu
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Junling Fan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fangping Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Gao
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Qun Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, National Chromatographic Research and Analysis Center, Dalian, China
| | - Qing Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yongmei Xiao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xiumei Xing
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Daochuan Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Tieliu Shi
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY
| | - Lihua Zhang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Science, National Chromatographic Research and Analysis Center, Dalian, China
| | - Wen Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
29
|
Zhang X, Connelly J, Chao Y, Wang QJ. Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021; 11:biom11030483. [PMID: 33807058 PMCID: PMC8005150 DOI: 10.3390/biom11030483] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase D (PKD) is a family of serine/threonine protein kinases operating in the signaling network of the second messenger diacylglycerol. The three family members, PKD1, PKD2, and PKD3, are activated by a variety of extracellular stimuli and transduce cell signals affecting many aspects of basic cell functions including secretion, migration, proliferation, survival, angiogenesis, and immune response. Dysregulation of PKD in expression and activity has been detected in many human diseases. Further loss- or gain-of-function studies at cellular levels and in animal models provide strong support for crucial roles of PKD in many pathological conditions, including cancer, metabolic disorders, cardiac diseases, central nervous system disorders, inflammatory diseases, and immune dysregulation. Complexity in enzymatic regulation and function is evident as PKD isoforms may act differently in different biological systems and disease models, and understanding the molecular mechanisms underlying these differences and their biological significance in vivo is essential for the development of safer and more effective PKD-targeted therapies. In this review, to provide a global understanding of PKD function, we present an overview of the PKD family in several major human diseases with more focus on cancer-associated biological processes.
Collapse
|
30
|
Kolczynska K, Loza-Valdes A, Hawro I, Sumara G. Diacylglycerol-evoked activation of PKC and PKD isoforms in regulation of glucose and lipid metabolism: a review. Lipids Health Dis 2020; 19:113. [PMID: 32466765 PMCID: PMC7257441 DOI: 10.1186/s12944-020-01286-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Protein kinase C (PKC) and Protein kinase D (PKD) isoforms can sense diacylglycerol (DAG) generated in the different cellular compartments in various physiological processes. DAG accumulates in multiple organs of the obese subjects, which leads to the disruption of metabolic homeostasis and the development of diabetes as well as associated diseases. Multiple studies proved that aberrant activation of PKCs and PKDs contributes to the development of metabolic diseases. DAG-sensing PKC and PKD isoforms play a crucial role in the regulation of metabolic homeostasis and therefore might serve as targets for the treatment of metabolic disorders such as obesity and diabetes.
Collapse
Affiliation(s)
- Katarzyna Kolczynska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Angel Loza-Valdes
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Izabela Hawro
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Grzegorz Sumara
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warszawa, Poland.
| |
Collapse
|