1
|
Shaw P, Dey Bhowmik A, Gopinatha Pillai MS, Robbins N, Dwivedi SKD, Rao G. Anoikis resistance in Cancer: Mechanisms, therapeutic strategies, potential targets, and models for enhanced understanding. Cancer Lett 2025; 624:217750. [PMID: 40294841 DOI: 10.1016/j.canlet.2025.217750] [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: 01/21/2025] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Anoikis, defined as programmed cell death triggered by the loss of cell-extracellular matrix (ECM) and cell-cell interactions, is crucial for maintaining tissue homeostasis and preventing aberrant cell migration. Cancer cells, however, display anoikis resistance (AR) which in turn enables cancer metastasis. AR results from alterations in apoptotic signaling, metabolic reprogramming, autophagy modulation, and epigenetic changes, allowing cancer cells to survive in detached conditions. In this review we describe the mechanisms underlying both anoikis and AR, focusing on intrinsic and extrinsic pathways, disrupted cell-ECM interactions, and autophagy in cancer. Recent findings (i.e., between 2014 and 2024) on epigenetic regulation of AR and its role in metastasis are discussed. Therapeutic strategies targeting AR, including chemical inhibitors, are highlighted alongside a network analysis of 122 proteins reported to be associated with AR which identifies 53 hub proteins as potential targets. We also evaluate in vitro and in vivo models for studying AR, emphasizing their role in advancing metastasis research. Our overall goal is to guide future studies and therapeutic developments to counter cancer metastasis.
Collapse
Affiliation(s)
- Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Nathan Robbins
- James E. Hurley School of Science and Mathematics, Oklahoma Baptist University, Shawnee, OK, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA.
| |
Collapse
|
2
|
Mori K, Togo A, Yamashita K, Sakuragi S, Bannai H, Umezawa T, Ohta K, Asahi T, Nozaki C, Kataoka K. Mitochondrial damage and ER stress in CB1 receptor antagonist-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neuropharmacology 2025; 273:110440. [PMID: 40185361 DOI: 10.1016/j.neuropharm.2025.110440] [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: 01/03/2025] [Revised: 03/13/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Cannabinoid receptor type 1 (CB1R) is the key modulator of neuronal viability. CB1R antagonists provide neuroprotective effects on neurotoxicity caused by e.g. neuronal injury. However, the underlying mechanisms and potential limitations of CB1R antagonism remain unclear. Here we investigated the impact of environmental conditions on CB1R antagonist effects. We have found that cell-permeable CB1R antagonists, rimonabant and AM251, induced cell death in human neuroblastoma SH-SY5Y cells under serum-free conditions. Mitochondrial morphological analysis revealed mitochondrial swelling characterized by their network fragmentation and cristae reduction. Phosphoproteomics analysis showed the ER stress signaling pathway PERK/eIF2α/ATF4/CHOP, leading to caspase-dependent apoptosis. These results suggest that CB1R antagonists promote apoptosis via mitochondrial damage and ER stress under serum-free conditions in SH-SY5Y cells. Our findings indicate that while CB1R antagonists may be neuroprotective in certain conditions, they may also pose a neurotoxic risk in environments characterized by cellular stress or nutrient deprivation.
Collapse
Affiliation(s)
- Kazuaki Mori
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Akinobu Togo
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Kota Yamashita
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Shigeo Sakuragi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hiroko Bannai
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Taishi Umezawa
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Keisuke Ohta
- Advanced Imaging Research Center, Kurume University School of Medicine, 67 Asahi-cho, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Toru Asahi
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan
| | - Chihiro Nozaki
- Global Center for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Kosuke Kataoka
- Comprehensive Research Organization, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo, 162-0041, Japan; Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan.
| |
Collapse
|
3
|
Hanne N, Hu D, Vidal-García M, Allen C, Shakir MB, Liu W, Hallgrímsson B, Marcucio R. Downstream branches of receptor tyrosine kinase signaling act interdependently to shape the face. Dev Dyn 2025. [PMID: 40391979 DOI: 10.1002/dvdy.70041] [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: 12/11/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Previously we found that increasing fibroblast growth factor (FGF) signaling in the neural crest cells within the frontonasal process (FNP) of the chicken embryo caused dysmorphology that was correlated with reduced proliferation, disrupted cellular orientation, and lower MAPK activation but no change in PLCγ and PI3K activation. This suggests RTK signaling may drive craniofacial morphogenesis through specific downstream effectors that affect cellular activities. In this study we inhibited three downstream branches of RTK signaling to determine their role in regulating cellular activities and how these changes affect morphogenesis of the FNP. RESULTS Small molecule inhibitors of MEK1/2, PI3K, and PLCγ were delivered individually and in tandem to the right FNP of chicken embryos. All treatments caused asymmetric proximodistal truncation on the treated side and a mild expansion on the untreated side compared to DMSO control treated FNPs. Inhibiting each pathway caused similar decreased proliferation and disrupted cellular orientation, and only mildly increased apoptosis. CONCLUSIONS Since RTK signaling is a ubiquitous and tightly regulated biochemical system, we conclude that the downstream pathways are robust to developmental perturbation through redundant signaling systems.
Collapse
Affiliation(s)
- Nicholas Hanne
- Department of Orthopaedic Surgery, University of California-San Francisco, San Francisco, California, USA
| | - Diane Hu
- Department of Orthopaedic Surgery, University of California-San Francisco, San Francisco, California, USA
| | - Marta Vidal-García
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Charlie Allen
- Cardiovascular Research Institute, University of California-San Francisco, San Francisco, California, USA
| | - M Bilal Shakir
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Wei Liu
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Benedikt Hallgrímsson
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
4
|
Li C, Syed MU, Nimbalkar A, Shen Y, Vieira MD, Fraser C, Inde Z, Qin X, Ouyang J, Kreuzer J, Clark SE, Kelley G, Hensley EM, Morris R, Lazaro R, Belmonte B, Oh A, Walcott M, Nabel CS, Caenepeel S, Saiki AY, Rex K, Lipford JR, Heist RS, Lin JJ, Haas W, Sarosiek K, Hughes PE, Hata AN. LKB1 regulates JNK-dependent stress signaling and apoptotic dependency of KRAS-mutant lung cancers. Nat Commun 2025; 16:4112. [PMID: 40316540 PMCID: PMC12048556 DOI: 10.1038/s41467-025-58753-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025] Open
Abstract
The efficacy of molecularly targeted therapies may be limited by co-occurring mutations within a tumor. Conversely, these alterations may confer collateral vulnerabilities that can be therapeutically leveraged. KRAS-mutant lung cancers are distinguished by recurrent loss of the tumor suppressor STK11/LKB1. Whether LKB1 modulates cellular responses to therapeutic stress seems unknown. Here we show that in LKB1-deficient KRAS-mutant lung cancer cells, inhibition of KRAS or its downstream effector MEK leads to hyperactivation of JNK due to loss of NUAK-mediated PP1B phosphatase activity. JNK-mediated inhibitory phosphorylation of BCL-XL rewires apoptotic dependencies, rendering LKB1-deficient cells vulnerable to MCL-1 inhibition. These results uncover an unknown role for LKB1 in regulating stress signaling and mitochondrial apoptosis independent of its tumor suppressor activity mediated by AMPK and SIK. Additionally, our study reveals a therapy-induced vulnerability in LKB1-deficient KRAS-mutant lung cancers that could be exploited as a genotype-informed strategy to improve the efficacy of KRAS-targeted therapies.
Collapse
Affiliation(s)
- Chendi Li
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Yi Shen
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Cameron Fraser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zintis Inde
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xingping Qin
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jian Ouyang
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Biochemistry & Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Johannes Kreuzer
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah E Clark
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Grace Kelley
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Emily M Hensley
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Robert Morris
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Raul Lazaro
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Audris Oh
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Makeba Walcott
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Christopher S Nabel
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Koch Institute for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Anne Y Saiki
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | - Karen Rex
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Rebecca S Heist
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Kristopher Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Lab for Systems Pharmacology, Harvard Program in Therapeutics Science, Harvard Medical School, Boston, MA, USA
| | | | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Qian AS, Kluck GEG, Yu P, Gonzalez L, Balint E, Trigatti BL. Apolipoprotein A1 deficiency increases macrophage apoptosis and necrotic core development in atherosclerotic plaques in a Bim-dependent manner. J Lipid Res 2025; 66:100782. [PMID: 40120762 PMCID: PMC12051063 DOI: 10.1016/j.jlr.2025.100782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
In advanced atherosclerotic lesions, macrophage apoptosis contributes to plaque progression and the formation of necrotic cores, rendering plaques vulnerable to rupture. The proapoptotic protein B-cell lymphoma 2 [Bcl-2] interacting mediator of cell death (Bim) plays a crucial role in mediating apoptosis in macrophages under prolonged endoplasmic reticulum stress. HDL has been shown to suppress macrophage apoptosis induced by endoplasmic reticulum stressors. To investigate the impact of apolipoprotein A1 (ApoA1) deficiency, associated with reduced HDL levels, on necrotic core growth and plaque apoptosis, we introduced ApoA1 deficiency into low-density lipoprotein receptor (LDLR) knockout mice and fed them a high-fat diet for 10 weeks. ApoA1-deficient Ldlr KO mice developed advanced plaques characterized by large necrotic cores, increased apoptosis, and elevated Bim expression in macrophages within the plaques. To assess whether deletion of Bim could mitigate this development, mice underwent bone marrow transplantation with bone marrow from either Bim-deficient mice or from mice with a deletion of myeloid-derived Bim driven by LyzM-cre. Inhibiting Bim in all bone marrow-derived cells led to leukocytosis, reductions in plasma cholesterol and triglyceride levels, and decreased plaque apoptosis, necrotic core, and plaque sizes in ApoA1 and Ldlr double-KO mice but not in Ldlr KO mice. Likewise, conditional deletion of Bim in the myeloid compartment of ApoA1 and Ldlr double-KO mice also reduced apoptosis, necrotic core sizes, and plaque sizes, without inducing leukocytosis or lowering plasma cholesterol levels. These findings suggest that ApoA1 deficiency triggers apoptosis in myeloid cells through a Bim-dependent pathway, significantly contributing to the development of necrotic cores and the progression of atherosclerotic plaques.
Collapse
Affiliation(s)
- Alexander S Qian
- Thrombosis and Atherosclerosis Research Institute, Centre for Metabolism, Obesity and Diabetes Research, and Department of Biochemistry and Biomedical Sciences, McMaster University and, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - George E G Kluck
- Thrombosis and Atherosclerosis Research Institute, Centre for Metabolism, Obesity and Diabetes Research, and Department of Biochemistry and Biomedical Sciences, McMaster University and, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Pei Yu
- Thrombosis and Atherosclerosis Research Institute, Centre for Metabolism, Obesity and Diabetes Research, and Department of Biochemistry and Biomedical Sciences, McMaster University and, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Leticia Gonzalez
- Thrombosis and Atherosclerosis Research Institute, Centre for Metabolism, Obesity and Diabetes Research, and Department of Biochemistry and Biomedical Sciences, McMaster University and, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Elizabeth Balint
- Thrombosis and Atherosclerosis Research Institute, Centre for Metabolism, Obesity and Diabetes Research, and Department of Biochemistry and Biomedical Sciences, McMaster University and, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Bernardo L Trigatti
- Thrombosis and Atherosclerosis Research Institute, Centre for Metabolism, Obesity and Diabetes Research, and Department of Biochemistry and Biomedical Sciences, McMaster University and, Hamilton Health Sciences, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
Li X, Hu F, Lu T, Wu S, Ma G, Lin Y, Zhang H. Endoplasmic reticulum stress in non-small cell lung cancer. Am J Cancer Res 2025; 15:1829-1851. [PMID: 40371139 PMCID: PMC12070083 DOI: 10.62347/rgrq7608] [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: 01/21/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
The Endoplasmic reticulum (ER), an organelle present in various eukaryotic cells, is responsible for protein synthesis, modification, folding, and transport, as well as for the regulation of lipid metabolism and Ca2+ homeostasis. ER stress plays a pivotal role in the pathogenesis and therapeutic response of non-small cell lung cancer (NSCLC), significantly influencing cellular fate decisions through its unique sensing and regulatory mechanisms. This review aims to elucidate the key role of ER stress sensors and to explore how they mediate cell autophagy, apoptosis, and non-apoptotic modes of cell death in the context of drug-treated NSCLC. This investigation lays a solid foundation for optimizing future treatment strategies for NSCLC.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Fangning Hu
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Tong Lu
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Shuo Wu
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Guanqiang Ma
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Yani Lin
- Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, School of Laboratory Animal and Shandong Laboratory Animal Center, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
| | - Hua Zhang
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| |
Collapse
|
7
|
Howe J, Barbar EJ. Dynamic interactions of dimeric hub proteins underlie their diverse functions and structures: A comparative analysis of 14-3-3 and LC8. J Biol Chem 2025; 301:108416. [PMID: 40107617 PMCID: PMC12017986 DOI: 10.1016/j.jbc.2025.108416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/22/2025] Open
Abstract
Hub proteins interact with a host of client proteins and regulate multiple cellular functions. Dynamic hubs have a single binding interface for one client at a time resulting in competition among clients with the highest affinity. Dynamic dimeric hubs with two identical sites bind either two different client proteins or two chains of the same client to form homogenous complexes and could also form heterogeneous mixtures of interconverting complexes. Here, we review the interactions of the dimeric hubs 14-3-3 and LC8. 14-3-3 is a phosphoserine/threonine binding protein involved in structuring client proteins and regulating their phosphorylation. LC8 is involved in promoting the dimerization of client peptides and the rigidification of their disordered regions. Both 14-3-3 and LC8 are essential genes, with 14-3-3 playing a crucial role in apoptosis and cell cycle regulation, while LC8 is critical for the assembly of proteins involved in transport, DNA repair, and transcription. Interestingly, both protein dimers can dissociate by phosphorylation, which results in their interactome-wide changes. Their interactions are also regulated by the phosphorylation of their clients. Both form heterogeneous complexes with various functions including phase separation, signaling, and viral hijacking where they restrict the conformational heterogeneity of their dimeric clients that bind nucleic acids. This comparative analysis highlights the importance of dynamic protein-protein interactions in the diversity of functions of 14-3-3 and LC8 and how small differences in structures of interfaces explain why 14-3-3 is primarily involved in the regulation of phosphorylation states while LC8 is primarily involved in the regulation of assembly of large dynamic complexes.
Collapse
Affiliation(s)
- Jesse Howe
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA
| | - Elisar J Barbar
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA.
| |
Collapse
|
8
|
Vogler M, Braun Y, Smith VM, Westhoff MA, Pereira RS, Pieper NM, Anders M, Callens M, Vervliet T, Abbas M, Macip S, Schmid R, Bultynck G, Dyer MJ. The BCL2 family: from apoptosis mechanisms to new advances in targeted therapy. Signal Transduct Target Ther 2025; 10:91. [PMID: 40113751 PMCID: PMC11926181 DOI: 10.1038/s41392-025-02176-0] [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: 11/07/2024] [Revised: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The B cell lymphoma 2 (BCL2) protein family critically controls apoptosis by regulating the release of cytochrome c from mitochondria. In this cutting-edge review, we summarize the basic biology regulating the BCL2 family including canonical and non-canonical functions, and highlight milestones from basic research to clinical applications in cancer and other pathophysiological conditions. We review laboratory and clinical development of BH3-mimetics as well as more recent approaches including proteolysis targeting chimeras (PROTACs), antibody-drug conjugates (ADCs) and tools targeting the BH4 domain of BCL2. The first BCL2-selective BH3-mimetic, venetoclax, showed remarkable efficacy with manageable toxicities and has transformed the treatment of several hematologic malignancies. Following its success, several chemically similar BCL2 inhibitors such as sonrotoclax and lisaftoclax are currently under clinical evaluation, alone and in combination. Genetic analysis highlights the importance of BCL-XL and MCL1 across different cancer types and the possible utility of BH3-mimetics targeting these proteins. However, the development of BH3-mimetics targeting BCL-XL or MCL1 has been more challenging, with on-target toxicities including thrombocytopenia for BCL-XL and cardiac toxicities for MCL1 inhibitors precluding clinical development. Tumor-specific BCL-XL or MCL1 inhibition may be achieved by novel targeting approaches using PROTACs or selective drug delivery strategies and would be transformational in many subtypes of malignancy. Taken together, we envision that the targeting of BCL2 proteins, while already a success story of translational research, may in the foreseeable future have broader clinical applicability and improve the treatment of multiple diseases.
Collapse
Affiliation(s)
- Meike Vogler
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Yannick Braun
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Victoria M Smith
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Raquel S Pereira
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Nadja M Pieper
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Marius Anders
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Manon Callens
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Tim Vervliet
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Maha Abbas
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Ralf Schmid
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Institute for Structural and Chemical Biology, University of Leicester, Leicester, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Martin Js Dyer
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
9
|
Hanne N, Hu D, Vidal-García M, Allen C, Shakir MB, Liu W, Hallgrímsson B, Marcucio R. Downstream branches of receptor tyrosine kinase signaling act interdependently to shape the face. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627829. [PMID: 39713427 PMCID: PMC11661274 DOI: 10.1101/2024.12.10.627829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Background – Previously we found that increasing fibroblast growth factor (FGF) signaling in the neural crest cells within the frontonasal process (FNP) of the chicken embryo caused dysmorphology that was correlated with reduced proliferation, disrupted cellular orientation, and lower MAPK activation but no change in PLCy and PI3K activation. This suggests RTK signaling may drive craniofacial morphogenesis through specific downstream effectors that affect cellular activities. In this study we inhibited three downstream branches of RTK signaling to determine their role in regulating cellular activities and how these changes affect morphogenesis of the FNP. Results – Small molecule inhibitors of MEK1/2, PI3K, and PLCy were delivered individually and in tandem to the right FNP of chicken embryos. All treatments caused asymmetric proximodistal truncation on the treated side and a mild expansion on the untreated side compared to DMSO control treated FNPs. Inhibiting each pathway caused similar decreased proliferation and disrupted cellular orientation, but did not affect apoptosis. Conclusions – Since RTK signaling is a ubiquitous and tightly regulated biochemical system we conclude that the downstream pathways are robust to developmental perturbation through redundant signaling systems.
Collapse
Affiliation(s)
- Nicholas Hanne
- Department of Orthopaedic Surgery, University of California - San Francisco, San Francisco, California, USA
| | - Diane Hu
- Department of Orthopaedic Surgery, University of California - San Francisco, San Francisco, California, USA
| | - Marta Vidal-García
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Charlie Allen
- Cardiovascular Research Institute, University of California - San Francisco, San Francisco, California, USA
| | - M Bilal Shakir
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Wei Liu
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Benedikt Hallgrímsson
- Deptartment of Cell Biology & Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, University of California - San Francisco, San Francisco, California, USA
| |
Collapse
|
10
|
Lu Y, Zhou J, Wang H, Gao H, Ning E, Shao Z, Hao Y, Yang X. Endoplasmic reticulum stress-mediated apoptosis and autophagy in osteoarthritis: From molecular mechanisms to therapeutic applications. Cell Stress Chaperones 2024; 29:805-830. [PMID: 39571722 DOI: 10.1016/j.cstres.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 12/09/2024] Open
Abstract
Osteoarthritis (OA) is characterized primarily by the degeneration of articular cartilage, with a high prevalence and disability rate. The functional phenotype of chondrocytes, as the sole cell type within cartilage, is vital for OA progression. Due to the avascular nature of cartilage and its limited regenerative capacity, repair following injury poses significant challenges. Various cellular stressors, including hypoxia, nutrient deprivation, oxidative stress, and collagen mutations, can lead to the accumulation of misfolded proteins in the endoplasmic reticulum (ER), resulting in ER stress (ERS). In response to restore ER homeostasis as well as cellular vitality and function, a series of adaptive mechanisms are triggered, including the unfolded protein response, ER-associated degradation, and ER-phagy. Prolonged or severe ERS may exceed the adaptive capacity of cells, leading to dysregulation in apoptosis and autophagy-key pathogenic factors contributing to chondrocyte damage and OA progression. This review examines the relationship between ERS in OA chondrocytes and both apoptosis and autophagy in order to identify potential therapeutic targets and strategies for prevention and treatment of OA.
Collapse
Affiliation(s)
- Yifan Lu
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China
| | - Hong Wang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China
| | - Hua Gao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China
| | - Eryu Ning
- Gusu School, Nanjing Medical University, Suzhou, PR China; Department of Sports Rehabilitation, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China
| | - Zhiqiang Shao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China.
| | - Xing Yang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, PR China; Gusu School, Nanjing Medical University, Suzhou, PR China.
| |
Collapse
|
11
|
Seyrek K, Espe J, Reiss E, Lavrik IN. The Crosstalk of Apoptotic and Non-Apoptotic Signaling in CD95 System. Cells 2024; 13:1814. [PMID: 39513921 PMCID: PMC11545656 DOI: 10.3390/cells13211814] [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: 09/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanisms of CD95 (Fas/APO-1)-mediated extrinsic apoptotic pathway in cancer cells have been extensively studied. The majority of human cells express CD95, but not all these cells can induce extrinsic apoptosis. Accumulating evidence has shown that CD95 is a multifunctional protein, and its stimulation can also elicit non-apoptotic or even survival signals. It has become clear that under certain cellular contexts, due to the various checkpoints, CD95 activation can trigger both apoptotic and non-apoptotic signals. The crosstalk of death and survival signals may occur at different levels of signal transduction. The strength of the CD95 stimulation, initial levels of anti-apoptotic proteins, and posttranslational modifications of the core DISC components have been proposed to be the most important factors in the life/death decisions at CD95. Successful therapeutic targeting of CD95 signaling pathways will require a better understanding of the crosstalk between CD95-induced apoptotic and cell survival pathways. In this review, in order to gain a systematic understanding of the crosstalk between CD95-mediated apoptosis and non-apoptotic signaling, we will discuss these issues in a step-by-step way.
Collapse
Affiliation(s)
| | | | | | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.S.); (J.E.); (E.R.)
| |
Collapse
|
12
|
Zhang S, Kim J, Lee G, Ahn HR, Kim YE, Kim HJ, Yu JS, Park M, Kang KW, Kim H, Jung BH, Kwon SW, Jang DS, Yang HO. Phytotherapeutic BS012 and Its Active Component Ameliorate Allergic Asthma via Inhibition of Th2-Mediated Immune Response and Apoptosis. Biomol Ther (Seoul) 2024; 32:744-758. [PMID: 39370723 PMCID: PMC11535288 DOI: 10.4062/biomolther.2024.058] [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: 04/12/2024] [Revised: 06/03/2024] [Accepted: 06/26/2024] [Indexed: 10/08/2024] Open
Abstract
Asthma is a chronic inflammatory disorder of the lungs that results in airway inflammation and narrowing. BS012 is an herbal remedy containing Asarum sieboldii, Platycodon grandiflorum, and Cinnamomum cassia extracts. To elucidate the anti-asthma effect of BS012, this study analyzed the immune response, respiratory protection, and changes in metabolic mechanisms in an ovalbumin-induced allergic asthma mouse model. Female BALB/c mice were exposed to ovalbumin to induce allergic asthma. Bronchoalveolar lavage fluid and plasma were analyzed for interleukin and immunoglobulin E levels. Histological analyses of the lungs were performed to measure morphological changes. Apoptosis-related mediators were assayed by western blotting. Plasma and lung tissue metabolomic analyses were performed to investigate the metabolic changes. A T-helper-2-like differentiated cell model was used to identify the active components of BS012. BS012 treatment improved inflammatory cell infiltration, mucus production, and goblet cell hyperplasia in lung tissues. BS012 also significantly downregulated ovalbumin-specific immunoglobulin E in plasma and T-helper-2-specific cytokines, interleukin-4 and -5, in bronchoalveolar lavage fluid. The lungs of ovalbumin-inhaled mice exhibited nerve growth factor-mediated apoptotic protein expression, which was significantly attenuated by BS012 treatment. Ovalbumin-induced abnormalities in amino acid and lipid metabolism were improved by BS012 in correlation with its anti-inflammatory properties and normalization of energy metabolism. Additionally, the differentiated cell model revealed that N-isobutyl-dodecatetraenamide is an active component that contributes to the anti-allergic properties of BS012. The current findings demonstrate the anti-allergic and respiratory protective functions of BS012 against allergic asthma, which can be considered a therapeutic candidate.
Collapse
Affiliation(s)
- Siqi Zhang
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
- KIST-School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Joonki Kim
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
- KIST-School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Gakyung Lee
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| | - Hong Ryul Ahn
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Yeo Eun Kim
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| | - Hee Ju Kim
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| | - Jae Sik Yu
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| | - Miso Park
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hocheol Kim
- Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Byung Hwa Jung
- KIST-School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sung Won Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae Sik Jang
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Ok Yang
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
13
|
Venkatachalam A, Correia C, Peterson KL, Hou X, Schneider PA, Strathman AR, Flatten KS, Sine CC, Balczewski EA, McGehee CD, Larson MC, Duffield LN, Meng XW, Vincelette ND, Ding H, Oberg AL, Couch FJ, Swisher EM, Li H, Weroha SJ, Kaufmann SH. Proapoptotic activity of JNK-sensitive BH3-only proteins underpins ovarian cancer response to replication checkpoint inhibitors. Mol Cancer 2024; 23:224. [PMID: 39375715 PMCID: PMC11457406 DOI: 10.1186/s12943-024-02125-5] [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: 05/23/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
Recent studies indicate that replication checkpoint modulators (RCMs) such as inhibitors of CHK1, ATR, and WEE1 have promising monotherapy activity in solid tumors, including platinum-resistant high grade serous ovarian cancer (HGSOC). However, clinical response rates are generally below 30%. While RCM-induced DNA damage has been extensively examined in preclinical and clinical studies, the link between replication checkpoint interruption and tumor shrinkage remains incompletely understood. Here we utilized HGSOC cell lines and patient-derived xenografts (PDXs) to study events leading from RCM treatment to ovarian cancer cell death. These studies show that RCMs increase CDC25A levels and CDK2 signaling in vitro, leading to dysregulated cell cycle progression and increased replication stress in HGSOC cell lines independent of homologous recombination status. These events lead to sequential activation of JNK and multiple BH3-only proteins, including BCL2L11/BIM, BBC3/PUMA and the BMF, all of which are required to fully initiate RCM-induced apoptosis. Activation of the same signaling pathway occurs in HGSOC PDXs that are resistant to poly(ADP-ribose) polymerase inhibitors but respond to RCMs ex vivo with a decrease in cell number in 3-dimensional culture and in vivo with xenograft shrinkage or a significantly diminished growth rate. These findings identify key cell death-initiating events that link replication checkpoint inhibition to antitumor response in ovarian cancer.
Collapse
Affiliation(s)
- Annapoorna Venkatachalam
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Cristina Correia
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Kevin L Peterson
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Xianon Hou
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Paula A Schneider
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Annabella R Strathman
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Karen S Flatten
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Chance C Sine
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Emily A Balczewski
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Present Address: Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Cordelia D McGehee
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Melissa C Larson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Laura N Duffield
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - X Wei Meng
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Nicole D Vincelette
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Present Address: H. Lee Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Husheng Ding
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Fergus J Couch
- Division of Experimental Pathology, Department of Laboratory Medicine, and Pathology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Elizabeth M Swisher
- Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Hu Li
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - S John Weroha
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA.
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA.
- Division of Hematology, Department of Medicine, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
14
|
Li X, Ruan T, Wang S, Sun X, Liu C, Peng Y, Tao Y. Mitochondria at the Crossroads of Cholestatic Liver Injury: Targeting Novel Therapeutic Avenues. J Clin Transl Hepatol 2024; 12:792-801. [PMID: 39280065 PMCID: PMC11393838 DOI: 10.14218/jcth.2024.00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 09/18/2024] Open
Abstract
Bile acids are byproducts of cholesterol metabolism in the liver and constitute the primary components of bile. Disruption of bile flow leads to cholestasis, characterized by the accumulation of hydrophobic bile acids in the liver and bloodstream. Such accumulation can exacerbate liver impairment. This review discussed recent developments in understanding how bile acids contribute to liver damage, including disturbances in mitochondrial function, endoplasmic reticulum stress, inflammation, and autophagy dysfunction. Mitochondria play a pivotal role in cholestatic liver injury by influencing hepatocyte apoptosis and inflammation. Recent findings linking bile acids to liver damage highlight new potential treatment targets for cholestatic liver injury.
Collapse
Affiliation(s)
- Xutao Li
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianyin Ruan
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Siyuan Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Sun
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai, China
| | - Yuan Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanyan Tao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai, China
| |
Collapse
|
15
|
Roshdy M, Zaky DA, Abbas SS, Abdallah DM. Niacin, an innovative protein kinase-C-dependent endoplasmic reticulum stress reticence in murine Parkinson's disease. Life Sci 2024; 351:122865. [PMID: 38914304 DOI: 10.1016/j.lfs.2024.122865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/08/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
AIMS Niacin (NIA) supplementation showed effectiveness against Parkinson's disease (PD) in clinical trials. The depletion of NAD and endoplasmic reticulum stress response (ERSR) are implicated in the pathogenesis of PD, but the potential role for NAD precursors on ERSR is not yet established. This study was undertaken to decipher NIA molecular mechanisms against PD-accompanied ERSR, especially in relation to PKC. METHODS Alternate-day-low-dose-21 day-subcutaneous exposure to rotenone (ROT) in rats induced PD. Following the 5th ROT injection, rats received daily doses of either NIA alone or preceded by the PKC inhibitor tamoxifen (TAM). Extent of disease progression was assessed by behavioral, striatal biochemical and striatal/nigral histopathological/immunohistochemical analysis. KEY FINDINGS Via activating PKC/LKB1/AMPK stream, NIA post-treatment attenuated the ERSR reflected by the decline in ATF4, ATF6 and XBP1s to downregulate the apoptotic markers, CHOP/GADD153, p-JNK and active caspase-3. Such amendments congregated in motor activity/coordination improvements in open field and rotarod tasks, enhanced grid test latency and reduced overall PD scores, while boosting nigral/striatal tyrosine hydroxylase immunoreactivity and increasing intact neurons (Nissl stain) in both SNpc and striatum that showed less neurodegeneration (H&E stain). To different extents, TAM reverted all the NIA-related actions to prove PKC as a fulcrum in conveying the drug neurotherapeutic potential. SIGNIFICANCE PKC activation is a pioneer mechanism in the drug ERSR inhibitory anti-apoptotic modality to clarify NIA promising clinical and potent preclinical anti-PD efficacy. This kinase can be tagged as a druggable target for future add-on treatments that can assist dopaminergic neuronal aptitude against this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Merna Roshdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Ahmed Orabi District, Cairo 44971, Egypt
| | - Doaa A Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| | - Samah S Abbas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr International University, Ahmed Orabi District, Cairo 44971, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt
| |
Collapse
|
16
|
Kim SH, Kang DW, Kwon D, Jung YS. Critical role of endoplasmic reticulum stress on bisphenol A-induced cytotoxicity in human keratinocyte HaCaT cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:4091-4104. [PMID: 38629620 DOI: 10.1002/tox.24290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/04/2024] [Accepted: 03/31/2024] [Indexed: 07/14/2024]
Abstract
Bisphenol A (BPA) is widely used in plastic and paper products, and its exposure can occur through skin contact or oral ingestion. The hazardous effects of BPA absorbed through the skin may be more severe; however, few studies have investigated the skin toxicity of BPA. This study investigated the effects of BPA on human epidermal keratinocyte cell lines, which is relevant for skin exposure. BPA treatment reduced cell viability in a time- and concentration-dependent manner and elevated oxidative and endoplasmic reticulum (ER) stress. N-acetylcysteine (NAC), an oxidative stress inhibitor, reduced BPA-induced reactive oxygen species (ROS) levels. However, only 10% of the decreased cell viability was restored at the highest NAC concentration. Treatment with tauroursodeoxycholic acid (TUDCA), which is an ER stress inhibitor, effectively countered the increase in ER stress-related proteins induced by BPA. Moreover, TUDCA treatment led to a reduction in oxidative stress, as demonstrated by the decrease in ROS levels, maintenance of mitochondrial membrane potential, and modulation of stress signaling proteins. Consequently, TUDCA significantly improved BPA-induced cytotoxicity in a concentration-dependent manner. Notably, combined treatment using TUDCA and NAC further reduced the BPA-induced ROS levels; however, no significant difference in cell viability was observed compared with that for TUDCA treatment alone. These findings indicated that the oxidative stress observed following BPA exposure was exacerbated by ER stress. Moreover, the principal factor driving BPA-induced cytotoxicity was indeed ER stress, which has potential implications for developing therapeutic strategies for diseases associated with similar stress responses.
Collapse
Affiliation(s)
- Sou Hyun Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Dong Wan Kang
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Doyoung Kwon
- College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, Republic of Korea
| | - Young-Suk Jung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
17
|
Chen JH, Wei CM, Lin QY, Wang Z, Zhang FM, Shi MN, Lan WJ, Sun CG, Lin WJ, Ma WZ. Notopterygium Incisum Extract Promotes Apoptosis by Preventing the Degradation of BIM in Colorectal Cancer. Curr Med Sci 2024; 44:833-840. [PMID: 38967889 DOI: 10.1007/s11596-024-2883-1] [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: 01/24/2024] [Accepted: 04/09/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Colorectal cancer (CRC), a prevalent malignancy worldwide, has prompted extensive research into anticancer drugs. Traditional Chinese medicinal materials offer promising avenues for cancer management due to their diverse pharmacological activities. This study investigated the effects of Notopterygium incisum, a traditional Chinese medicine named Qianghuo (QH), on CRC cells and the underlying mechanism. METHODS The sulforhodamine B assay and colony formation assay were employed to assess the effect of QH extract on the proliferation of CRC cell lines HCT116 and Caco-2. Propidium iodide (PI) staining was utilized to detect cell cycle progression, and PE Annexin V staining to detect apoptosis. Western blotting was conducted to examine the levels of apoptotic proteins, including B-cell lymphoma 2-interacting mediator of cell death (BIM), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (BAX) and cleaved caspase-3, as well as BIM stability after treatment with the protein synthesis inhibitor cycloheximide. The expression of BAX was suppressed using lentivirus-mediated shRNA to validate the involvement of the BIM/BAX axis in QH-induced apoptosis. The in vivo effects of QH extract on tumor growth were observed using a xenograft model. Lastly, APCMin+ mice were used to study the effects of QH extract on primary intestinal tumors. RESULTS QH extract exhibited significant in vitro anti-CRC activities evidenced by the inhibition of cell proliferation, perturbation of cell cycle progression, and induction of apoptosis. Mechanistically, QH extract significantly increased the stability of BIM proteins, which undergo rapid degradation under unstressed conditions. Knockdown of BAX, the downstream effector of BIM, significantly rescued QH-induced apoptosis. Furthermore, the in vitro effect of QH extract was recapitulated in vivo. QH extract significantly inhibited the tumor growth of HCT116 xenografts in nude mice and decreased the number of intestinal polyps in the APCMin+ mice. CONCLUSION QH extract promotes the apoptosis of CRC cells by preventing the degradation of BIM.
Collapse
Affiliation(s)
- Jun-He Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Cheng-Ming Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Qian-Yu Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Zi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Fu-Ming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Mei-Na Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chang-Gang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261053, China
| | - Wan-Jun Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Wen-Zhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| |
Collapse
|
18
|
Choi MJ, Kang SJ, Lee YK, Choi KC, Lee DH, Jeong HY, Kim MW, Kim KS, Park YS. Novel Lipid Nanocomplex Co-Carrying Bcl2 siRNA and Quantum Dots for EGF Receptor-Targeted Anti-Cancer Theranosis. Int J Mol Sci 2024; 25:6246. [PMID: 38892434 PMCID: PMC11172456 DOI: 10.3390/ijms25116246] [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: 05/02/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Many different types of nanoparticles have been suggested for tumor-targeted theranosis. However, most systems were prepared through a series of complicated processes and could not even overcome the blood-immune barriers. For the accurate diagnosis and effective treatment of cancers, herein we suggested the lipid micellar structure capturing quantum dot (QD) for cancer theranosis. The QD/lipid micelles (QDMs) were prepared using a simple self-assembly procedure and then conjugated with anti-epidermal growth factor receptor (EGFR) antibodies for tumor targeting. As a therapeutic agent, Bcl2 siRNA-cholesterol conjugates were loaded on the surface of QDMs. The EGFR-directed QDMs containing Bcl2 siRNA, so-called immuno-QDM/siBcl2 (iQDM/siBcl2), exhibited the more effective delivery of QDs and siBcl2 to target human colorectal cancer cells in cultures as well as in mouse xenografts. The effective in vivo targeting of iQDM/siBcl2 resulted in a more enhanced therapeutic efficacy of siBcl2 to the target cancer in mice. Based on the results, anti-EGFR QDM capturing therapeutic siRNA could be suggested as an alternative modality for tumor-targeted theranosis.
Collapse
Affiliation(s)
- Moon Jung Choi
- Department of Medicine, Brown University, Providence, RI 02903, USA;
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
| | - Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
| | - Yeon Kyung Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
| | - Kang Chan Choi
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
| | - Do Hyun Lee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
| | - Hwa Yeon Jeong
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Min Woo Kim
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Keun Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea;
| | - Yong Serk Park
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26496, Republic of Korea; (S.J.K.); (Y.K.L.); (K.C.C.); (D.H.L.); (H.Y.J.); (M.W.K.)
| |
Collapse
|
19
|
Balhara M, Neikirk K, Marshall A, Hinton A, Kirabo A. Endoplasmic Reticulum Stress in Hypertension and Salt Sensitivity of Blood Pressure. Curr Hypertens Rep 2024; 26:273-290. [PMID: 38602583 PMCID: PMC11166838 DOI: 10.1007/s11906-024-01300-9] [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] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Hypertension is a principal risk factor for cardiovascular morbidity and mortality, with its severity exacerbated by high sodium intake, particularly in individuals with salt-sensitive blood pressure. However, the mechanisms underlying hypertension and salt sensitivity are only partly understood. Herein, we review potential interactions in hypertension pathophysiology involving the immune system, endoplasmic reticulum (ER) stress, the unfolded protein response (UPR), and proteostasis pathways; identify knowledge gaps; and discuss future directions. RECENT FINDINGS Recent advancements by our research group and others reveal interactions within and between adaptive and innate immune responses in hypertension pathophysiology. The salt-immune-hypertension axis is further supported by the discovery of the role of dendritic cells in hypertension, marked by isolevuglandin (IsoLG) formation. Alongside these broadened understandings of immune-mediated salt sensitivity, the contributions of T cells to hypertension have been recently challenged by groups whose findings did not support increased resistance of Rag-1-deficient mice to Ang II infusion. Hypertension has also been linked to ER stress and the UPR. Notably, a holistic approach is needed because the UPR engages in crosstalk with autophagy, the ubiquitin proteasome, and other proteostasis pathways, that may all involve hypertension. There is a critical need for studies to establish cause and effect relationships between ER stress and the UPR in hypertension pathophysiology in humans and to determine whether the immune system and ER stress function mainly to exacerbate or initiate hypertension and target organ injury. This review of recent studies proposes new avenues for future research for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maria Balhara
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA.
- Vanderbilt Center for Immunobiology, Nashville, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, USA.
- Vanderbilt Institute for Global Health, Nashville, USA.
| |
Collapse
|
20
|
Zavvarian MM, Modi AD, Sadat S, Hong J, Fehlings MG. Translational Relevance of Secondary Intracellular Signaling Cascades Following Traumatic Spinal Cord Injury. Int J Mol Sci 2024; 25:5708. [PMID: 38891894 PMCID: PMC11172219 DOI: 10.3390/ijms25115708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a life-threatening and life-altering condition that results in debilitating sensorimotor and autonomic impairments. Despite significant advances in the clinical management of traumatic SCI, many patients continue to suffer due to a lack of effective therapies. The initial mechanical injury to the spinal cord results in a series of secondary molecular processes and intracellular signaling cascades in immune, vascular, glial, and neuronal cell populations, which further damage the injured spinal cord. These intracellular cascades present promising translationally relevant targets for therapeutic intervention due to their high ubiquity and conservation across eukaryotic evolution. To date, many therapeutics have shown either direct or indirect involvement of these pathways in improving recovery after SCI. However, the complex, multifaceted, and heterogeneous nature of traumatic SCI requires better elucidation of the underlying secondary intracellular signaling cascades to minimize off-target effects and maximize effectiveness. Recent advances in transcriptional and molecular neuroscience provide a closer characterization of these pathways in the injured spinal cord. This narrative review article aims to survey the MAPK, PI3K-AKT-mTOR, Rho-ROCK, NF-κB, and JAK-STAT signaling cascades, in addition to providing a comprehensive overview of the involvement and therapeutic potential of these secondary intracellular pathways following traumatic SCI.
Collapse
Affiliation(s)
- Mohammad-Masoud Zavvarian
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Akshat D. Modi
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Department of Biological Sciences, University of Toronto, Scarborough, ON M1C 1A4, Canada
- Department of Human Biology, University of Toronto, Toronto, ON M5S 3J6, Canada
| | - Sarah Sadat
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - James Hong
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
| | - Michael G. Fehlings
- Division of Genetics and Development, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada; (M.-M.Z.); (A.D.M.); (S.S.); (J.H.)
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
21
|
Zhou ZY, Wu L, Liu YF, Tang MY, Tang JY, Deng YQ, Liu L, Nie BB, Zou ZK, Huang L. IRE1α: from the function to the potential therapeutic target in atherosclerosis. Mol Cell Biochem 2024; 479:1079-1092. [PMID: 37310588 DOI: 10.1007/s11010-023-04780-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/03/2023] [Indexed: 06/14/2023]
Abstract
Inositol requiring enzyme 1 (IRE1) is generally thought to control the most conserved pathway in the unfolded protein response (UPR). Two isoforms of IRE1, IRE1α and IRE1β, have been reported in mammals. IRE1α is a ubiquitously expressed protein whose knockout shows marked lethality. In contrast, the expression of IRE1β is exclusively restricted in the epithelial cells of the respiratory and gastrointestinal tracts, and IRE1β-knockout mice are phenotypically normal. As research continues to deepen, IRE1α was showed to be tightly linked to inflammation, lipid metabolism regulation, cell death and so on. Growing evidence also suggests an important role for IRE1α in promoting atherosclerosis (AS) progression and acute cardiovascular events through disrupting lipid metabolism balance, facilitating cells apoptosis, accelerating inflammatory responses and promoting foam cell formation. In addition, IRE1α was recognized as novel potential therapeutic target in AS prevention. This review provides some clues about the relationship between IRE1α and AS, hoping to contribute to further understanding roles of IRE1α in atherogenesis and to be helpful for the design of novel efficacious therapeutics agents targeting IRE1α-related pathways.
Collapse
Affiliation(s)
- Zheng-Yang Zhou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Li Wu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yi-Fan Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Mu-Yao Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Jing-Yi Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Anaesthesiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Ya-Qian Deng
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Lei Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bin-Bin Nie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zi-Kai Zou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Liang Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
22
|
Skalka GL, Tsakovska M, Murphy DJ. Kinase signalling adaptation supports dysfunctional mitochondria in disease. Front Mol Biosci 2024; 11:1354682. [PMID: 38434478 PMCID: PMC10906720 DOI: 10.3389/fmolb.2024.1354682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 03/05/2024] Open
Abstract
Mitochondria form a critical control nexus which are essential for maintaining correct tissue homeostasis. An increasing number of studies have identified dysregulation of mitochondria as a driver in cancer. However, which pathways support and promote this adapted mitochondrial function? A key hallmark of cancer is perturbation of kinase signalling pathways. These pathways include mitogen activated protein kinases (MAPK), lipid secondary messenger networks, cyclic-AMP-activated (cAMP)/AMP-activated kinases (AMPK), and Ca2+/calmodulin-dependent protein kinase (CaMK) networks. These signalling pathways have multiple substrates which support initiation and persistence of cancer. Many of these are involved in the regulation of mitochondrial morphology, mitochondrial apoptosis, mitochondrial calcium homeostasis, mitochondrial associated membranes (MAMs), and retrograde ROS signalling. This review will aim to both explore how kinase signalling integrates with these critical mitochondrial pathways and highlight how these systems can be usurped to support the development of disease. In addition, we will identify areas which require further investigation to fully understand the complexities of these regulatory interactions. Overall, this review will emphasize how studying the interaction between kinase signalling and mitochondria improves our understanding of mitochondrial homeostasis and can yield novel therapeutic targets to treat disease.
Collapse
Affiliation(s)
- George L. Skalka
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mina Tsakovska
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Daniel J. Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
- CRUK Scotland Institute, Glasgow, United Kingdom
| |
Collapse
|
23
|
Rowland MB, Moore PE, Correll RN. Regulation of cardiac fibroblast cell death by unfolded protein response signaling. Front Physiol 2024; 14:1304669. [PMID: 38283278 PMCID: PMC10811265 DOI: 10.3389/fphys.2023.1304669] [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: 09/29/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024] Open
Abstract
The endoplasmic reticulum (ER) is a tightly regulated organelle that requires specific environmental properties to efficiently carry out its function as a major site of protein synthesis and folding. Embedded in the ER membrane, ER stress sensors inositol-requiring enzyme 1 (IRE1), protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6) serve as a sensitive quality control system collectively known as the unfolded protein response (UPR). In response to an accumulation of misfolded proteins, the UPR signals for protective mechanisms to cope with the cellular stress. Under prolonged unstable conditions and an inability to regain homeostasis, the UPR can shift from its original adaptive response to mechanisms leading to UPR-induced apoptosis. These UPR signaling pathways have been implicated as an important feature in the development of cardiac fibrosis, but identifying effective treatments has been difficult. Therefore, the apoptotic mechanisms of UPR signaling in cardiac fibroblasts (CFs) are important to our understanding of chronic fibrosis in the heart. Here, we summarize the maladaptive side of the UPR, activated downstream pathways associated with cell death, and agents that have been used to modify UPR-induced apoptosis in CFs.
Collapse
Affiliation(s)
- Mary B. Rowland
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Patrick E. Moore
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Robert N. Correll
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- Center for Convergent Bioscience and Medicine, University of Alabama, Tuscaloosa, AL, United States
| |
Collapse
|
24
|
Zhou Y, Fang C, Yuan L, Guo M, Xu X, Shao A, Zhang A, Zhou D. Redox homeostasis dysregulation in noise-induced hearing loss: oxidative stress and antioxidant treatment. J Otolaryngol Head Neck Surg 2023; 52:78. [PMID: 38082455 PMCID: PMC10714662 DOI: 10.1186/s40463-023-00686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
Noise exposure is an important cause of acquired hearing loss. Studies have found that noise exposure causes dysregulated redox homeostasis in cochlear tissue, which has been recognized as a signature feature of hearing loss. Oxidative stress plays a pivotal role in many diseases via very complex and diverse mechanisms and targets. Reactive oxygen species are products of oxidative stress that exert toxic effects on a variety of physiological activities and are considered significant in noise-induced hearing loss (NIHL). Endogenous cellular antioxidants can directly or indirectly counteract oxidative stress and regulate intracellular redox homeostasis, and exogenous antioxidants can complement and enhance this effect. Therefore, antioxidant therapy is considered a promising direction for NIHL treatment. However, drug experiments have been limited to animal models of NIHL, and these experiments and related observations are difficult to translate in humans; therefore, the mechanisms and true effects of these drugs need to be further analyzed. This review outlines the effects of oxidative stress in NIHL and discusses the main mechanisms and strategies of antioxidant treatment for NIHL.
Collapse
Affiliation(s)
- Yuhang Zhou
- Health Management Center, Tongde Hospital of Zhejiang Province, Hangzhou, China
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengchen Guo
- Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Xu
- School of Medicine, Ningbo University, Ningbo, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Danyang Zhou
- Health Management Center, Tongde Hospital of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
25
|
Lyu Y, Guan X, Xu X, Wang P, Li Q, Panigrahi M, Zhang J, Chen N, Huang B, Lei C. A whole genome scan reveals distinct features of selection in Zhaotong cattle of Yunnan province. Anim Genet 2023; 54:731-742. [PMID: 37796667 DOI: 10.1111/age.13363] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 10/07/2023]
Abstract
Over the years, indigenous cattle have not only played an essential role in securing primary food sources but have also been utilized for labor by humans, making them invaluable genetic resources. The Zhaotong cattle, a native Chinese breed from the Yunnan province, possess excellent meat quality and resistance to heat and humidity. Here we used whole genome sequencing data of 104 animals to delve into the population structure, genomic diversity and potential positive selection signals in Zhaotong cattle. The findings of this study demonstrate that the genetic composition of Zhaotong cattle was primarily derived from Chinese indicine cattle and East Asian cattle. The nucleotide diversity of Zhaotong cattle was only lower than that of Chinese indicine cattle, which was much higher than that of other taurine cattle. Genome-wide selection scans detected a series of positive candidate regions containing multiple key genes related to bone development and metabolism (CA10, GABRG3, GLDN and NOTUM), meat quality traits (ALG8, LINGO2, MYO5B, PRKG1 and GABRB1), immune response (ADA2, BMF, LEF1 and PAK6) and heat resistance (EIF2AK4 and LEF1). In summary, this study supplies essential genetic insights into the genome diversity within Zhaotong cattle and provides a foundational framework for comprehending the genetic basis of indigenous cattle breeds.
Collapse
Affiliation(s)
- Yang Lyu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Xiwen Guan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinglong Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Pengfei Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qiaoxian Li
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Manjit Panigrahi
- Division of Animal Genetics, Indian Veterinary Research Institute, Bareilly, UP, India
| | - Jicai Zhang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
26
|
Hai S, Zhao J, Chen C, Wang C, Ma L, Rahman SU, Zhao C, Feng S, Wu J, Wang X. Zearalenone promotes porcine ESCs apoptosis by enhancing Drp1-mediated mitochondrial fragmentation and activating the JNK pathway. Food Chem Toxicol 2023; 182:114110. [PMID: 37879531 DOI: 10.1016/j.fct.2023.114110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
Zearalenone (ZEA) is widely present in food and feed, and pigs are susceptible to its effects. This study explored the underlying function of ZEA-induced apoptosis in porcine endometrial stromal cells (ESCs) through activation of the JNK signaling pathway and mitochondrial division. This study utilized ESCs to explore the impact of exposure to ZEA. A mitochondrial division inhibitor (Mdivi) was also included as a reference. The results indicated a gradual decrease in cell viability with increasing ZEA concentration. In addition, ZEA can modify the growth status of porcine ESCs, disrupt their ultrastructure, and lead to apoptosis of porcine ESCs via the mitochondrial division pathway and JNK signaling pathway. In summary, our study found the critical targets of ZEA infected with pig ESCs, which provided a conceptual foundation to prevent and control ZEA.
Collapse
Affiliation(s)
- Sirao Hai
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Jie Zhao
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Chuangjiang Chen
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Chenlong Wang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Li Ma
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Sajid Ur Rahman
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chang Zhao
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, Hefei, 230036, China.
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Engineering Laboratory for Animal Food Quality and Bio-safety, Hefei, 230036, China.
| |
Collapse
|
27
|
Shi L, Zha H, Pan Z, Wang J, Xia Y, Li H, Huang H, Yue R, Song Z, Zhu J. DUSP1 protects against ischemic acute kidney injury through stabilizing mtDNA via interaction with JNK. Cell Death Dis 2023; 14:724. [PMID: 37935658 PMCID: PMC10630453 DOI: 10.1038/s41419-023-06247-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/23/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023]
Abstract
The mechanism underlying acute kidney injury (AKI) and AKI-to-Chronic kidney disease (CKD) transition remains unclear, but mitochondrial dysfunction may be a key driving factor. Literature reports suggest that dual-specificity phosphatase 1 (DUSP1) plays a critical role in maintaining mitochondrial function and structural integrity. In this study, ischemic Acute Kidney Injury (AKI) and post-ischemic fibrosis models were established by clamping the renal pedicle with different reperfusion times. To investigate the role of DUSP1, constitutional Dusp1 knockout mice and tubular-specific Sting knockout mice were used. Mitochondrial damage was assessed through electron microscopy observation, measurements of mitochondrial membrane potential, mtDNA release, and BAX translocation. We found that Dusp1 expression was significantly upregulated in human transplant kidney tissue and mouse AKI tissue. Dusp1 gene deletion exacerbated acute ischemic injury, post-ischemic renal fibrosis, and tubular mitochondrial dysfunction in mice. Mechanistically, DUSP1 could directly bind to JNK, and DUSP1 deficiency could lead to aberrant phosphorylation of JNK and BAX mitochondria translocation. BAX translocation promoted mitochondrial DNA (mtDNA) leakage and activated the cGAS-STING pathway. Inhibition of JNK or BAX could inhibit mtDNA leakage. Furthermore, STING knockout or JNK inhibition could significantly mitigate the adverse effects of DUSP1 deficiency in ischemic AKI model. Collectively, our findings suggest that DUSP1 is a regulator for the protective response during AKI. DUSP1 protects against AKI by preventing BAX-induced mtDNA leakage and blocking excessive activation of the cGAS-STING signaling axis through JNK dephosphorylation.
Collapse
Affiliation(s)
- Lang Shi
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei, 443000, China
| | - Zhou Pan
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiayi Wang
- Department of Anesthesiology, the Xiangya Second Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Yao Xia
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei, 443000, China
| | - Huimin Li
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei, 443000, China
| | - Hua Huang
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei, 443000, China
| | - Ruchi Yue
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei, 443000, China
| | - Zhixia Song
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei, 443000, China
| | - Jiefu Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
28
|
Zhao H, Liu R, Wang L, Tang F, Chen W, Liu YN. Artificial Macrophage with Hierarchical Nanostructure for Biomimetic Reconstruction of Antitumor Immunity. NANO-MICRO LETTERS 2023; 15:216. [PMID: 37737506 PMCID: PMC10516848 DOI: 10.1007/s40820-023-01193-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/27/2023] [Indexed: 09/23/2023]
Abstract
Artificial cells are constructed from synthetic materials to imitate the biological functions of natural cells. By virtue of nanoengineering techniques, artificial cells with designed biomimetic functions provide alternatives to natural cells, showing vast potential for biomedical applications. Especially in cancer treatment, the deficiency of immunoactive macrophages results in tumor progression and immune resistance. To overcome the limitation, a BaSO4@ZIF-8/transferrin (TRF) nanomacrophage (NMΦ) is herein constructed as an alternative to immunoactive macrophages. Alike to natural immunoactive macrophages, NMΦ is stably retained in tumors through the specific affinity of TRF to tumor cells. Zn2+ as an "artificial cytokine" is then released from the ZIF-8 layer of NMΦ under tumor microenvironment. Similar as proinflammatory cytokines, Zn2+ can trigger cell anoikis to expose tumor antigens, which are selectively captured by the BaSO4 cavities. Therefore, the hierarchical nanostructure of NMΦs allows them to mediate immunogenic death of tumor cells and subsequent antigen capture for T cell activation to fabricate long-term antitumor immunity. As a proof-of-concept, the NMΦ mimics the biological functions of macrophage, including tumor residence, cytokine release, antigen capture and immune activation, which is hopeful to provide a paradigm for the design and biomedical applications of artificial cells.
Collapse
Affiliation(s)
- Henan Zhao
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, People's Republic of China
| | - Renyu Liu
- Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Liqiang Wang
- Henan Province Industrial Technology Research Institute of Resources and Materials, School of Material Science and Engineering, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Feiying Tang
- College of Chemical Engineering, Xiangtan University, Xiangtan, 411105, Hunan, People's Republic of China
| | - Wansong Chen
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, People's Republic of China.
| | - You-Nian Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan, People's Republic of China.
| |
Collapse
|
29
|
Merati A, Kotian S, Acton A, Placzek W, Smithberger E, Shelton AK, Miller CR, Stern JL. Glioma Stem Cells Are Sensitized to BCL-2 Family Inhibition by Compromising Histone Deacetylases. Int J Mol Sci 2023; 24:13688. [PMID: 37761989 PMCID: PMC10530722 DOI: 10.3390/ijms241813688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastoma (GBM) remains an incurable disease with an extremely high five-year recurrence rate. We studied apoptosis in glioma stem cells (GSCs) in response to HDAC inhibition (HDACi) combined with MEK1/2 inhibition (MEKi) or BCL-2 family inhibitors. MEKi effectively combined with HDACi to suppress growth, induce cell cycle defects, and apoptosis, as well as to rescue the expression of the pro-apoptotic BH3-only proteins BIM and BMF. A RNAseq analysis of GSCs revealed that HDACi repressed the pro-survival BCL-2 family genes MCL1 and BCL-XL. We therefore replaced MEKi with BCL-2 family inhibitors and observed enhanced apoptosis. Conversely, a ligand for the cancer stem cell receptor CD44 led to reductions in BMF, BIM, and apoptosis. Our data strongly support further testing of HDACi in combination with MEKi or BCL-2 family inhibitors in glioma.
Collapse
Affiliation(s)
- Aran Merati
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Spandana Kotian
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alexus Acton
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - William Placzek
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Erin Smithberger
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Abigail K. Shelton
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - C. Ryan Miller
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Josh L. Stern
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, Birmingham, AL 35294, USA
| |
Collapse
|
30
|
Nadel G, Maik-Rachline G, Seger R. JNK Cascade-Induced Apoptosis-A Unique Role in GqPCR Signaling. Int J Mol Sci 2023; 24:13527. [PMID: 37686335 PMCID: PMC10487481 DOI: 10.3390/ijms241713527] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The response of cells to extracellular signals is mediated by a variety of intracellular signaling pathways that determine stimulus-dependent cell fates. One such pathway is the cJun-N-terminal Kinase (JNK) cascade, which is mainly involved in stress-related processes. The cascade transmits its signals via a sequential activation of protein kinases, organized into three to five tiers. Proper regulation is essential for securing a proper cell fate after stimulation, and the mechanisms that regulate this cascade may involve the following: (1) Activatory or inhibitory phosphorylations, which induce or abolish signal transmission. (2) Regulatory dephosphorylation by various phosphatases. (3) Scaffold proteins that bring distinct components of the cascade in close proximity to each other. (4) Dynamic change of subcellular localization of the cascade's components. (5) Degradation of some of the components. In this review, we cover these regulatory mechanisms and emphasize the mechanism by which the JNK cascade transmits apoptotic signals. We also describe the newly discovered PP2A switch, which is an important mechanism for JNK activation that induces apoptosis downstream of the Gq protein coupled receptors. Since the JNK cascade is involved in many cellular processes that determine cell fate, addressing its regulatory mechanisms might reveal new ways to treat JNK-dependent pathologies.
Collapse
Affiliation(s)
| | | | - Rony Seger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (G.N.); (G.M.-R.)
| |
Collapse
|
31
|
Netterfield TS, Ostheimer GJ, Tentner AR, Joughin BA, Dakoyannis AM, Sharma CD, Sorger PK, Janes KA, Lauffenburger DA, Yaffe MB. Biphasic JNK-Erk signaling separates the induction and maintenance of cell senescence after DNA damage induced by topoisomerase II inhibition. Cell Syst 2023; 14:582-604.e10. [PMID: 37473730 PMCID: PMC10627503 DOI: 10.1016/j.cels.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 03/24/2023] [Accepted: 06/13/2023] [Indexed: 07/22/2023]
Abstract
Genotoxic stress in mammalian cells, including those caused by anti-cancer chemotherapy, can induce temporary cell-cycle arrest, DNA damage-induced senescence (DDIS), or apoptotic cell death. Despite obvious clinical importance, it is unclear how the signals emerging from DNA damage are integrated together with other cellular signaling pathways monitoring the cell's environment and/or internal state to control different cell fates. Using single-cell-based signaling measurements combined with tensor partial least square regression (t-PLSR)/principal component analysis (PCA) analysis, we show that JNK and Erk MAPK signaling regulates the initiation of cell senescence through the transcription factor AP-1 at early times after doxorubicin-induced DNA damage and the senescence-associated secretory phenotype (SASP) at late times after damage. These results identify temporally distinct roles for signaling pathways beyond the classic DNA damage response (DDR) that control the cell senescence decision and modulate the tumor microenvironment and reveal fundamental similarities between signaling pathways responsible for oncogene-induced senescence (OIS) and senescence caused by topoisomerase II inhibition. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tatiana S Netterfield
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gerard J Ostheimer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrea R Tentner
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brian A Joughin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexandra M Dakoyannis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charvi D Sharma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Computer Science and Molecular Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin A Janes
- Department of Biomedical Engineering and Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael B Yaffe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Acute Care Surgery, Trauma, and Surgical Critical Care, and Division of Surgical Oncology, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
32
|
Dong L, Xu M, Li Y, Xu W, Wu C, Zheng H, Xiao Z, Sun G, Ding L, Li X, Li W, Zhou L, Xia Q. SMURF1 attenuates endoplasmic reticulum stress by promoting the degradation of KEAP1 to activate NRF2 antioxidant pathway. Cell Death Dis 2023; 14:361. [PMID: 37316499 PMCID: PMC10267134 DOI: 10.1038/s41419-023-05873-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
Cancer cells consistently utilize the unfolded protein response (UPR) to encounter the abnormal endoplasmic reticulum (ER) stress induced by the accumulation of misfolded proteins. Extreme activation of the UPR could also provoke maladaptive cell death. Previous reports have shown that NRF2 antioxidant signaling is activated by UPR and serves as noncanonical pathway to defense and reduce excessive ROS levels during ER stress. However, the mechanisms of regulating NRF2 signaling upon ER stress in glioblastoma have not been fully elucidated. Here we identify that SMURF1 protects against ER stress and facilitates glioblastoma cell survival by rewiring KEAP1-NRF2 pathway. We show that ER stress induces SMURF1 degradation. Knockdown of SMURF1 upregulates IRE1 and PERK signaling in the UPR pathway and prevents ER-associated protein degradation (ERAD) activity, leading to cell apoptosis. Importantly, SMURF1 overexpression activates NRF2 signaling to reduce ROS levels and alleviate UPR-mediated cell death. Mechanistically, SMURF1 interacts with and ubiquitinates KEAP1 for its degradation (NRF2 negative regulator), resulting in NRF2 nuclear import. Moreover, SMURF1 loss reduces glioblastoma cell proliferation and growth in subcutaneously implanted nude mice xenografts. Taken together, SMURF1 rewires KEAP1-NRF2 pathway to confer resistance to ER stress inducers and protect glioblastoma cell survival. ER stress and SMURF1 modulation may provide promising therapeutic targets for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Lei Dong
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Mengchuan Xu
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yang Li
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Wanting Xu
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Chengwei Wu
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Hanfei Zheng
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhenyu Xiao
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Guochen Sun
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lei Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaobo Li
- BeiJing Tide Pharmaceutical Co. LTD, BeiJing, 102600, China
| | - Wenming Li
- BeiJing Tide Pharmaceutical Co. LTD, BeiJing, 102600, China
| | - Liying Zhou
- BeiJing Tide Pharmaceutical Co. LTD, BeiJing, 102600, China
| | - Qin Xia
- Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
33
|
20(S)-Ginsenoside Rh1 inhibits cisplatin-induced hearing loss by inhibiting the MAPK signaling pathway and suppressing apoptosis in vitro. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119461. [PMID: 36931607 DOI: 10.1016/j.bbamcr.2023.119461] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
As an anticancer drug, cisplatin is widely used, but its clinical application is restricted due to its severe side effects of ototoxicity. Therefore, this study was dedicated to assessing the benefit of ginsenoside extract, 20(S)-Ginsenoside Rh1 (Rh1), on cisplatin-induced ototoxicity. HEI-OC1 cells and neonatal cochlear explants were cultured. Cleaved caspase-3, TUNEL, and MitoSOX Red were observed in vitro by immunofluorescence staining. CCK8 and LDH cytotoxicity assays were detected to measure cell viability and cytotoxicity. Our results showed that Rh1 significantly increased cell viability, reduced cytotoxicity, and alleviated cisplatin-induced apoptosis. In addition, Rh1 pretreatment decreased the excessive accumulation of intracellular reactive oxygen species. Mechanistic studies indicated that Rh1 pretreatment reversed the increase of apoptotic protein expression, accumulation of mitochondrial ROS, and activation of the MAPK signaling pathway. These results suggested that Rh1 can act as an antioxidant and anti-apoptotic agent against cisplatin-induced hearing loss by suppressing the excessive accumulation of mitochondrial ROS, activation of MAPK signaling pathway and apoptosis.
Collapse
|
34
|
JNK Activation Correlates with Cognitive Impairment and Alteration of the Post-Synaptic Element in the 5xFAD AD Mouse Model. Cells 2023; 12:cells12060904. [PMID: 36980245 PMCID: PMC10047857 DOI: 10.3390/cells12060904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs) are a family of proteins that, once activated by stress stimuli, can alter neuronal functions and survival. The JNK cascade plays a crucial role in the post-synaptic neuronal compartment by altering its structural organization and leading, at worst, to an overall impairment of neuronal communication. Increasing evidence suggests that synaptic impairment is the first neurodegenerative event in Alzheimer’s disease (AD). To better elucidate this mechanism, we longitudinally studied 5xFAD mice at three selected time points representative of human AD symptom progression. We tested the mice cognitive performance by using the radial arm water maze (RAWM) in parallel with biochemical evaluations of post-synaptic enriched protein fraction and total cortical parenchyma. We found that 5xFAD mice presented a strong JNK activation at 3.5 months of age in the post-synaptic enriched protein fraction. This JNK activation correlates with a structural alteration of the post-synaptic density area and with memory impairment at this early stage of the disease that progressively declines to cause cell death. These findings pave the way for future studies on JNK as a key player in early neurodegeneration and as an important therapeutic target for the development of new compounds able to tackle synaptic impairment in the early phase of AD pathology.
Collapse
|
35
|
Javadi A, Nikhbakht MR, Ghasemian Yadegari J, Rustamzadeh A, Mohammadi M, Shirazinejad A, Azadbakht S, Abdi Z. In-vivo and in vitro assessments of the radioprotective potential natural and chemical compounds: a review. Int J Radiat Biol 2023; 99:155-165. [PMID: 35549605 DOI: 10.1080/09553002.2022.2078007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE The study of the radioactive role of natural and chemical substances on human and animal studies has been the subject of research by some researchers. Therefore, the review of some of the past and current studies conducted in this field, can provide helpful information to elucidate of the importance of radioprotective components in reducing radiation exposure side effects. METHODS The authors search for keywords including In vitro, In vivo, Radioprotective, Ionizing radiation, and Vitamin in ScienceDirect, Scopus, Pubmed, and Google Scholar databases to access previously published articles and search for more reference articles on the role of radioprotective materials from natural and chemical compounds. RESULTS Radiation exposure can produce reactive oxygen species (ROS) in the body, however most of which are eliminated by the body's natural mechanisms, but when the body's antioxidant systems do not have enough ability to neutralize free radicals, oxidative stress occurs, which causes damage to DNA and body tissues. Therefore, it is necessary use of alternative substances that reduce and inhibit free radicals. CONCLUSION In general, recommended that antioxidant component(s) can be protect tissue damages in humans or animals, due to the their ability to scavenge free radicals generated by ionizing radiation.
Collapse
Affiliation(s)
- Anis Javadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Reza Nikhbakht
- Department of Physiology and Pharmacology, School of Medicine Medicinal Plants Research Center Yasuj, University of Medical Sciences, Yasuj, Iran
| | - Javad Ghasemian Yadegari
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Mohammadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran.,Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.,Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alireza Shirazinejad
- Department of Food Science and Technology, Sarvestan Branch, Islamic Azad University, Sarvestan, Iran
| | - Saleh Azadbakht
- Department of Internal Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Zahra Abdi
- Department of Medical Biotechnology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
36
|
Wen Z, He X, Wang J, Wang H, Li T, Wen S, Ren Z, Cai N, Yang J, Li M, Ai H, Lu Y, Zhu Y, Shi G, Chen Y. Hyperlipidemia induces proinflammatory responses by activating STING pathway through IRE1α-XBP1 in retinal endothelial cells. J Nutr Biochem 2023; 112:109213. [PMID: 36370931 DOI: 10.1016/j.jnutbio.2022.109213] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/02/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022]
Abstract
Diabetic retinopathy (DR) is one of the most prevalent microvascular complications caused by diabetes mellitus. Previous studies demonstrate that microvascular endothelial inflammation caused by chronic hyperglycemia and hyperlipidemia plays a key role in the pathogenesis of DR. However, the detailed mechanisms on how endothelial inflammation contributes to DR are not fully understood. The STING pathway is an important innate immune signaling pathway. Although STING has been implicated in multiple autoimmune and metabolic diseases, it is not clear whether STING is involved in the pathogenesis of DR. Thus, re-analysis of the public single cell RNA sequencing (sc-RNAseq) data demonstrated that STING was highly expressed in mouse retinal vessels. Moreover, our results demonstrated that STING and p-TBK1 protein levels in retinal endothelial cells are significantly increased in mice fed with high fat diet compared with chow diet. In vitro, palmitic acid treatment on HRVECs induced mitochondrial DNA leakage into the cytosol, and augmented p-TBK1 protein and IFN-β mRNA levels. As STING is localized to the ER, we analyzed the relation between STING activation and ER stress. In HRVECs, STING pathway was shown to be activated under chemical-induced ER stress, but attenuated when IRE1α was abolished by genetic deletion or pharmacological inhibition. Taken together, our findings revealed that STING signaling plays an important role in mediating lipotoxicity-induced endothelial inflammatory and injury, and IRE1α-XBP1 signaling potentiated STING signaling. Thus, targeting the IRE1α or STING pathways to alleviate endothelial inflammation provides candidate therapeutic target for treating DR as well as other microvascular complications.
Collapse
Affiliation(s)
- Zheyao Wen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xuemin He
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Multidisciplinary Team for Obesity, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jin Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Heting Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ting Li
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Siying Wen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhitao Ren
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Nan Cai
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jifeng Yang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mei Li
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Multidisciplinary Team for Obesity, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; VIP Medical Service Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Heying Ai
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Lu
- Multidisciplinary Team for Obesity, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanhua Zhu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Multidisciplinary Team for Obesity, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guojun Shi
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Multidisciplinary Team for Obesity, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Yanming Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; Multidisciplinary Team for Obesity, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
37
|
Abstract
Significance: Thioredoxin (Trx) is a powerful antioxidant that reduces protein disulfides to maintain redox stability in cells and is involved in regulating multiple redox-dependent signaling pathways. Recent Advance: The current accumulation of findings suggests that Trx participates in signaling pathways that interact with various proteins to manipulate their dynamic regulation of structure and function. These network pathways are critical for cancer pathogenesis and therapy. Promising clinical advances have been presented by most anticancer agents targeting such signaling pathways. Critical Issues: We herein link the signaling pathways regulated by the Trx system to potential cancer therapeutic opportunities, focusing on the coordination and strengths of the Trx signaling pathways in apoptosis, ferroptosis, immunomodulation, and drug resistance. We also provide a mechanistic network for the exploitation of therapeutic small molecules targeting the Trx signaling pathways. Future Directions: As research data accumulate, future complex networks of Trx-related signaling pathways will gain in detail. In-depth exploration and establishment of these signaling pathways, including Trx upstream and downstream regulatory proteins, will be critical to advancing novel cancer therapeutics. Antioxid. Redox Signal. 38, 403-424.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zhengjia Zhao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | | | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, China
| |
Collapse
|
38
|
Yang S, Wang L, Wu Y, Wu A, Huang F, Tang X, Kantawong F, Anuchapreeda S, Qin D, Mei Q, Chen J, Huang X, Zhang C, Wu J. Apoptosis in megakaryocytes: Safeguard and threat for thrombopoiesis. Front Immunol 2023; 13:1025945. [PMID: 36685543 PMCID: PMC9845629 DOI: 10.3389/fimmu.2022.1025945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023] Open
Abstract
Platelets, generated from precursor megakaryocytes (MKs), are central mediators of hemostasis and thrombosis. The process of thrombopoiesis is extremely complex, regulated by multiple factors, and related to many cellular events including apoptosis. However, the role of apoptosis in thrombopoiesis has been controversial for many years. Some researchers believe that apoptosis is an ally of thrombopoiesis and platelets production is apoptosis-dependent, while others have suggested that apoptosis is dispensable for thrombopoiesis, and is even inhibited during this process. In this review, we will focus on this conflict, discuss the relationship between megakaryocytopoiesis, thrombopoiesis and apoptosis. In addition, we also consider why such a vast number of studies draw opposite conclusions of the role of apoptosis in thrombopoiesis, and try to figure out the truth behind the mystery. This review provides more comprehensive insights into the relationship between megakaryocytopoiesis, thrombopoiesis, and apoptosis and finds some clues for the possible pathological mechanisms of platelet disorders caused by abnormal apoptosis.
Collapse
Affiliation(s)
- Shuo Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yuesong Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Xiaoqin Tang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Fahsai Kantawong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Dalian Qin
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qibing Mei
- School of Pharmacy, Southwest Medical University, Luzhou, China
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xinwu Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunxiang Zhang
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Luzhou, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
39
|
CircSETD3 mediates acquired resistance to gefitinib in non-small lung cancer cells by FXR1/ECT2 pathway. Int J Biochem Cell Biol 2023; 154:106344. [PMID: 36503048 DOI: 10.1016/j.biocel.2022.106344] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gefitinib is the first-line treatment for non-small cell lung cancer (NSCLC) harboring EGFR sensitive mutation. However, acquired resistance significantly limits its therapeutic efficacy. CircSETD3 has been reported to promote gefitinib resistance in NSCLC cells, however, its underlying mechanisms have not been fully clarified. METHODS The expression of circSETD3 were detected in NSCLC patients who received gefitinib as first-line treatment, including 20 gefitinib-sensitive patients and 20 acquired gefitinib-resistant patients. Cell viability were examined by CCK8 assay. The mRNA and protein levels were detected by qRT-PCR and western blot. Using RNA pull-down assay followed by mass spectrometry to identified proteins that interact with circSETD3. The interaction between circSETD3 and fragile X-related protein-1 (FXR1) were further validated by RNA immunoprecipitation (RIP) and pull-down analysis. Fuorescence in situ hybridization (FISH) and immunofluorescence (IF) assays was used for the identification of sub-location of circSETD3 and FXR1 in cells. The effect of circSETD3 overexpression and knockdown on NSCLC tumor growth to gefitinib sensitivity was detected using the mouse xenograft model. RESULTS CircSETD3 was significantly upregulated in gefitinib-resistant NSCLC cells, and decreased the gefitinib sensitivity in vitro and in vivo. Mechanically, circSETD3 facilitated FXR1 binding to its downstream mRNA target, epithelial cell-transforming sequence 2 (ECT2), promoting ECT2 mRNA decay, which further inhibited cellular apoptosis. CONCLUSION CircSETD3/FXR1/ECT2 axis plays a critical role in the acquired resistance to gefitinib in NSCLC. Our results highlight the potential of circSETD3 as a biomarker and therapeutic target for NSCLC patients with acquired gefitinib resistance.
Collapse
|
40
|
Kaloni D, Diepstraten ST, Strasser A, Kelly GL. BCL-2 protein family: attractive targets for cancer therapy. Apoptosis 2023; 28:20-38. [PMID: 36342579 PMCID: PMC9950219 DOI: 10.1007/s10495-022-01780-7] [Citation(s) in RCA: 195] [Impact Index Per Article: 97.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
Acquired resistance to cell death is a hallmark of cancer. The BCL-2 protein family members play important roles in controlling apoptotic cell death. Abnormal over-expression of pro-survival BCL-2 family members or abnormal reduction of pro-apoptotic BCL-2 family proteins, both resulting in the inhibition of apoptosis, are frequently detected in diverse malignancies. The critical role of the pro-survival and pro-apoptotic BCL-2 family proteins in the regulation of apoptosis makes them attractive targets for the development of agents for the treatment of cancer. This review describes the roles of the various pro-survival and pro-apoptotic members of the BCL-2 protein family in normal development and organismal function and how defects in the control of apoptosis promote the development and therapy resistance of cancer. Finally, we discuss the development of inhibitors of pro-survival BCL-2 proteins, termed BH3-mimetic drugs, as novel agents for cancer therapy.
Collapse
Affiliation(s)
- Deeksha Kaloni
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia ,Department of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Sarah T Diepstraten
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia
| | - Andreas Strasser
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC Australia ,Department of Medical Biology, University of Melbourne, Melbourne, VIC Australia
| | - Gemma L Kelly
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
41
|
Wang X, Zhang S, Han K, Wang L, Liu X. Induction of Apoptosis by Matrine Derivative ZS17 in Human Hepatocellular Carcinoma BEL-7402 and HepG2 Cells through ROS-JNK-P53 Signalling Pathway Activation. Int J Mol Sci 2022; 23:ijms232415991. [PMID: 36555631 PMCID: PMC9783520 DOI: 10.3390/ijms232415991] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies and ranks third among cancer-related deaths worldwide. Using matrine as a lead compound, 12 matrine derivatives were designed and synthesised, and their antiproliferative activities were evaluated in four cancer cell lines. Eight of the twelve compounds showed strong antiproliferative activity, with an IC50 of <10 μM. The compound ZS17 exhibited strong antiproliferative activity in hepatocellular carcinoma cell lines with IC50 values in the range of 3.014−3.388 μM, which was much lower than that of matrine. Furthermore, we explored the role of ZS17 in inducing apoptosis in HCC cells in vitro and in vivo, as well as possible mechanisms involved. ZS17 inhibited the proliferation of BEL-7402 and HepG2 cells in time- and dose-dependent manners. In addition, we found that ZS17 significantly induced apoptosis and ROS (reactive oxygen species) production, promoted JNK phosphorylation, activated p53, and activated the caspase signalling pathway. Furthermore, the antioxidant NAC, JNK inhibitor SP600125, and Si-JNK increased cell viability, re-established cell metastasis, and inhibited ZS17-induced apoptosis. An in vivo antitumour assay demonstrated that ZS17 significantly reduced the number of migrating HepG2 cells in zebrafish embryos and suppressed the growth of HepG2 xenografts in nude mice without any obvious side effects. Our study demonstrated that the ROS-JNK-P53 pathway plays an important role in the destruction of liver tumour cells by ZS17. Thus, ZS17 may represent a promising chemotherapeutic agent for the treatment of HCC patients.
Collapse
Affiliation(s)
| | | | | | | | - Xu Liu
- Correspondence: (L.W.); (X.L.)
| |
Collapse
|
42
|
Luo J, Jin W, Jin M, Pan W, Gao S, Zhao X, Lai X, Sun L, Piao C. Jiedutongluotiaogan formula restores pancreatic function by suppressing excessive autophagy and endoplasmic reticulum stress. PHARMACEUTICAL BIOLOGY 2022; 60:1542-1555. [PMID: 35944284 PMCID: PMC9367665 DOI: 10.1080/13880209.2022.2107019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Jiedutongluotiaogan formula (JTTF), a traditional Chinese medicine (TCM), could promote islet function. However, the potential effect of JTTF on endoplasmic reticulum stress (ERS) and autophagy have not been reported. OBJECTIVE This study explores the potential effect of JTTF on ERS and autophagy in the pancreas. MATERIALS AND METHODS The Zucker diabetic fatty (ZDF) rats were randomised into five groups, control, model, JTTF (1, 3, 5 g/kg/day for 12 weeks). LPS induced pancreatic β-cells were treated with JTTF (50, 100, 200 μg/mL). LPS was used to induce pancreatic β-cell injury, with cell viability and insulin secretion evaluated using MTT, glucose-stimulated insulin secretion (GSIS) assays, and PCR. Intracellular Ca2+ concentration was measured using flow cytometry, while ERS and autophagy levels were monitored via Western blotting and/or immunostaining. RESULTS Compared with the model group, body weight, FGB, HbA1c, IPGTT, FINs, and HOMA-IR in JTTF treatment groups were significantly reduced. In islets cells treated with JTTF, the pancreatic islet cells in the JTTF group were increased, lipid droplets were reduced, and there was a decrease in Ca2+ (16.67%). After JTTF intervention, PERK, p-PERK, IRE1α, p- IRE1α, ATF6, eIF2α, GRP78, p-ULK1, LC3 and p62 expression decreased, whereas Beclin1and p-mTOR expression increased. In addition, the expression of proteins related to apoptosis in the JTTF groups were lower than those in the control group. DISCUSSION AND CONCLUSIONS JTTF may alleviate pancreatic β-cell injury by inhibiting ER stress and excessive autophagy in diabetic rats. This provides a new direction for treating diabetes and restoring pancreatic dysfunction by TCM.
Collapse
Affiliation(s)
- Jinli Luo
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Wenqi Jin
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Meiying Jin
- The Third Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Weiwei Pan
- School of Clinical Medicine, Changchun Medical College, Changchun, China
| | - Shengnan Gao
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xiaohua Zhao
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xingrong Lai
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Chunli Piao
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| |
Collapse
|
43
|
Dudkevich R, Koh JH, Beaudoin-Chabot C, Celik C, Lebenthal-Loinger I, Karako-Lampert S, Ahmad-Albukhari S, Thibault G, Henis-Korenblit S. Neuronal IRE-1 coordinates an organism-wide cold stress response by regulating fat metabolism. Cell Rep 2022; 41:111739. [PMID: 36450261 DOI: 10.1016/j.celrep.2022.111739] [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: 04/20/2022] [Revised: 10/07/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
Cold affects many aspects of biology, medicine, agriculture, and industry. Here, we identify a conserved endoplasmic reticulum (ER) stress response, distinct from the canonical unfolded protein response, that maintains lipid homeostasis during extreme cold. We establish that the ER stress sensor IRE-1 is critical for resistance to extreme cold and activated by cold temperature. Specifically, neuronal IRE-1 signals through JNK-1 and neuropeptide signaling to regulate lipid composition within the animal. This cold-response pathway can be bypassed by dietary supplementation with unsaturated fatty acids. Altogether, our findings define an ER-centric conserved organism-wide cold stress response, consisting of molecular neuronal sensors, effectors, and signaling moieties, which control adaptation to cold conditions in the organism. Better understanding of the molecular basis of this stress response is crucial for the optimal use of cold conditions on live organisms and manipulation of lipid saturation homeostasis, which is perturbed in human pathologies.
Collapse
Affiliation(s)
- Reut Dudkevich
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Jhee Hong Koh
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | | | - Cenk Celik
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | | | - Sarit Karako-Lampert
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Syed Ahmad-Albukhari
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore
| | - Sivan Henis-Korenblit
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
44
|
GRP94 Inhabits the Immortalized Porcine Hepatic Stellate Cells Apoptosis under Endoplasmic Reticulum Stress through Modulating the Expression of IGF-1 and Ubiquitin. Int J Mol Sci 2022; 23:ijms232214059. [PMID: 36430538 PMCID: PMC9694842 DOI: 10.3390/ijms232214059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/15/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Endoplasmic reticulum stress (ERS) is closely related to the occurrence and progression of metabolic liver disease. The treatment targeting glucose-regulated protein 94 (GRP94) for liver disease has gotten much attention, but the specific effect of GRP94 on hepatocyte apoptosis is still unclear. So far, all the studies on GRP94 have been conducted in mice or rats, and little study has been reported on pigs, which share more similarities with humans. In this study, we used low-dose (LD) and high-dose (HD) tunicamycin (TM) to establish ERS models on piglet livers and immortalized porcine hepatic stellate cells (HSCs). On the piglet ERS model we found that ERS could significantly (p < 0.01) stimulate the secretion and synthesis of insulin-like growth factor (IGF-1), IGF-1 receptor (IGF-1R), and IGF-binding protein (IGFBP)-1 and IGFBP-3; however, with the increase in ERS degree, the effect of promoting secretion and synthesis significantly (p < 0.01) decreased. In addition, the ubiquitin protein and ubiquitination-related gene were significantly increased (p < 0.05) in the LD group compared with the vehicle group. The protein level of Active-caspase 3 was significantly increased (p < 0.01) in the HD group, however, the TUNEL staining showed there was no significant apoptosis in the piglet liver ERS model. To explore the biofunction of ER chaperone GRP94, we used shRNA to knock down the expression of GRP94 in porcine HSCs. Interestingly, on porcine HSCs, the knockdown of GRP94 significantly (p < 0.05) decreased the secretion of IGF-1, IGFBP-1 and IGFBP-3 under ERS, but had no significant effect on these under normal condition, and knockdown GRP94 had a significant (p < 0.01) effect on the UBE2E gene and ubiquitin protein from the analysis of two-way ANOVA. On porcine HSCs apoptosis, the knockdown of GRP94 increased the cell apoptosis in TUNEL staining, and the two-way ANOVA analysis shows that knockdown GRP94 had a significant (p < 0.01) effect on the protein levels of Bcl-2 and Caspase-3. For CCK-8 assay, ERS had a significant inhibitory(p < 0.05) effect on cell proliferation when treated with ERS for 24 h, and both knockdown GRP94 and ERS had a significant inhibitory(p < 0.05) effect on cell proliferation when treated with ERS for 36 h and 48 h. We concluded that GRP94 can protect the cell from ERS-induced apoptosis by promoting the IGF-1 system and ubiquitin. These results provide valuable information on the adaptive mechanisms of the liver under ERS, and could help identify vital functional genes to be applied as possible diagnostic biomarkers and treatments for diseases induced by ERS in the future.
Collapse
|
45
|
Yu Y, Yang A, Yu G, Wang H. Endoplasmic Reticulum Stress in Chronic Obstructive Pulmonary Disease: Mechanisms and Future Perspectives. Biomolecules 2022; 12:1637. [PMID: 36358987 PMCID: PMC9687722 DOI: 10.3390/biom12111637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 09/08/2024] Open
Abstract
The endoplasmic reticulum (ER) is an integral organelle for maintaining protein homeostasis. Multiple factors can disrupt protein folding in the lumen of the ER, triggering ER stress and activating the unfolded protein response (UPR), which interrelates with various damage mechanisms, such as inflammation, apoptosis, and autophagy. Numerous studies have linked ER stress and UPR to the progression of chronic obstructive pulmonary disease (COPD). This review focuses on the mechanisms of other cellular processes triggered by UPR and summarizes drug intervention strategies targeting the UPR pathway in COPD to explore new therapeutic approaches and preventive measures for COPD.
Collapse
Affiliation(s)
| | | | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
46
|
Zhao J, Hai S, Chen J, Ma L, Rahman SU, Zhao C, Feng S, Li Y, Wu J, Wang X. Zearalenone Induces Apoptosis in Porcine Endometrial Stromal Cells through JNK Signaling Pathway Based on Endoplasmic Reticulum Stress. Toxins (Basel) 2022; 14:toxins14110758. [PMID: 36356008 PMCID: PMC9694026 DOI: 10.3390/toxins14110758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Zearalenone (ZEA) is an estrogen-like mycotoxin characterized mainly by reproductive toxicity, to which pigs are particularly sensitive. The aim of this study was to investigate the molecular mechanism of ZEA-induced apoptosis in porcine endometrial stromal cells (ESCs) by activating the JNK signaling pathway through endoplasmic reticulum stress (ERS). In this study, ESCs were exposed to ZEA, with the ERS inhibitor sodium 4-Phenylbutyrate (4-PBA) as a reference. The results showed that ZEA could damage cell structures, induce endoplasmic reticulum swelling and fragmentation, and decreased the ratio of live cells to dead cells significantly. In addition, ZEA could increase reactive oxygen species and Ca2+ levels; upregulate the expression of GRP78, CHOP, PERK, ASK1 and JNK; activate JNK phosphorylation and its high expression in the nucleus; upregulate the expression Caspase 3 and Caspase 9; and increase the Bax/Bcl-2 ratio, resulting in increased apoptosis. After 3 h of 4-PBA-pretreatment, ZEA was added for mixed culture, which showed that the inhibition of ERS could reduce the cytotoxicity of ZEA toward ESCs. Compared with the ZEA group, ERS inhibition increased cell viability; downregulated the expression of GRP78, CHOP, PERK, ASK1 and JNK; and decreased the nuclear level of p-JNK. The Bax/Bcl-2 ratio and the expression of Caspase 3 and Caspase 9 were downregulated, significantly alleviating apoptosis. These results demonstrate that ZEA can alter the morphology of ESCs, destroy their ultrastructure, and activate the JNK signaling via the ERS pathway, leading to apoptosis.
Collapse
Affiliation(s)
- Jie Zhao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Sirao Hai
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jiawen Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Li Ma
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Sajid Ur Rahman
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chang Zhao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yu Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, Hefei 230036, China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, Hefei 230036, China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, Hefei 230036, China
- Correspondence:
| |
Collapse
|
47
|
Nisa A, Kipper FC, Panigrahy D, Tiwari S, Kupz A, Subbian S. Different modalities of host cell death and their impact on Mycobacterium tuberculosis infection. Am J Physiol Cell Physiol 2022; 323:C1444-C1474. [PMID: 36189975 PMCID: PMC9662802 DOI: 10.1152/ajpcell.00246.2022] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/22/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is the pathogen that causes tuberculosis (TB), a leading infectious disease of humans worldwide. One of the main histopathological hallmarks of TB is the formation of granulomas comprised of elaborately organized aggregates of immune cells containing the pathogen. Dissemination of Mtb from infected cells in the granulomas due to host and mycobacterial factors induces multiple cell death modalities in infected cells. Based on molecular mechanism, morphological characteristics, and signal dependency, there are two main categories of cell death: programmed and nonprogrammed. Programmed cell death (PCD), such as apoptosis and autophagy, is associated with a protective response to Mtb by keeping the bacteria encased within dead macrophages that can be readily phagocytosed by arriving in uninfected or neighboring cells. In contrast, non-PCD necrotic cell death favors the pathogen, resulting in bacterial release into the extracellular environment. Multiple types of cell death in the PCD category, including pyroptosis, necroptosis, ferroptosis, ETosis, parthanatos, and PANoptosis, may be involved in Mtb infection. Since PCD pathways are essential for host immunity to Mtb, therapeutic compounds targeting cell death signaling pathways have been experimentally tested for TB treatment. This review summarizes different modalities of Mtb-mediated host cell deaths, the molecular mechanisms underpinning host cell death during Mtb infection, and its potential implications for host immunity. In addition, targeting host cell death pathways as potential therapeutic and preventive approaches against Mtb infection is also discussed.
Collapse
Affiliation(s)
- Annuurun Nisa
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Franciele C Kipper
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sangeeta Tiwari
- Department of Biological Sciences, Border Biomedical Research Center (BBRC), University of Texas, El Paso, Texas
| | - Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Townsville, Queensland, Australia
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
48
|
Li S, Luo X, Liao Z, Xu H, Liang M, Mai K, Zhang Y. Additional supplementation of sulfur-containing amino acids in the diets improves the intestinal health of turbot fed high-lipid diets. FISH & SHELLFISH IMMUNOLOGY 2022; 130:368-379. [PMID: 36115604 DOI: 10.1016/j.fsi.2022.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/29/2022] [Accepted: 09/10/2022] [Indexed: 06/15/2023]
Abstract
An eight-week feeding trial was conducted to investigate the effects of diets supplemented with three sulfur-containing amino acids (SAA), namely, methionine, cysteine, and taurine, on the intestinal health status of juvenile turbot (Scophthalmus maximus) fed high-lipid diets. Four diets were formulated, namely, a high-lipid control diet (16% lipid, HL) and three SAA-supplemented diets, which were formulated by supplementing 1.5% methionine (HLM), 1.5% cysteine (HLC), and 1.5% taurine (HLT) into the HL control diet, respectively. Each diet was assigned to triplicate tanks, and each tank was stocked with 30 juvenile fish (appr. initial weight, 8 g). The histological and morphometric results showed that dietary SAA supplementation obviously improved the intestinal morphology and integrity, in particular as reflected by higher height of microvilli and mucosal folds. Dietary SAA supplementation, in particular cysteine, up-regulated the gene expression of mucin-2 and tight junction proteins (ZO-1, Tricellilun and JAM). Dietary SAA supplementation remarkably down-regulated the gene expression of apoptosis-related factors such as p38, JNK, and Bax, expression of pro-inflammatory factors (e.g., NF-κB, AP-1 IL-1β, IL-8, and TNF-α). SAA supplementation resulted in higher antioxidative abilities in the intestine. Additionally, dietary SAA supplementation largely altered the communities of intestinal microbiota. Compared with the HL group, higher relative abundance of potential beneficial bacteria, and lower relative abundance of opportunistic pathogens were observed in SAA-supplemented groups. Dietary taurine supplementation significantly increased the relative abundance of Ligilactobacillus (in particular Lactobacillus murinus) and Limosilactobacillus (especially Lactobacillus reuteri). In conclusion, dietary sulfur-containing amino acids supplementation have promising potential in ameliorating the intestinal inflammation of turbot fed high-lipid diets. Especially dietary cysteine and taurine supplementation have more positive effects on the communities of the intestinal microbiota of turbot.
Collapse
Affiliation(s)
- Sihui Li
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Xing Luo
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Zhangbin Liao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao, 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Yanjiao Zhang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| |
Collapse
|
49
|
Lu Z, Sun GF, Pan XA, Qu XH, Yang P, Chen ZP, Han XJ, Wang T. BCATc inhibitor 2 ameliorated mitochondrial dysfunction and apoptosis in oleic acid-induced non-alcoholic fatty liver disease model. Front Pharmacol 2022; 13:1025551. [PMID: 36386234 PMCID: PMC9650408 DOI: 10.3389/fphar.2022.1025551] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/18/2022] [Indexed: 09/14/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent hepatic disease in the world. Disorders of branched chain amino acid (BCAA) metabolism is involved in various diseases. In this study, we aim to explore the role of BCAA metabolism in the development of NAFLD and the protective effect of BCATc Inhibitor 2, an inhibitor of cytosolic branched chain amino acid transaminase, against NAFLD as well as its underlying mechanism. It was found that oleic acid induced lipid accumulation and apoptosis in HepG2 and LO2 cells. Supplementation of BCAAs further aggravated oleic acid-induced lipid accumulation and apoptosis. In contrast, treatment of BCATc Inhibitor 2 ameliorated oleic acid-induced lipid accumulation and apoptosis. Molecularly, supplementation of BCAAs or treatment of BCATc Inhibitor 2 up-regulated or down-regulated the expression of SREBP1 and lipogenesis-related genes without affecting lipolysis-related genes. BCATc Inhibitor 2 maintained mitochondrial function by ameliorating oleic acid-induced mitochondrial ROS generation and mitochondrial membrane potential disruption. In addition, BCATc Inhibitor 2 treatment alleviated oleic acid-induced activation of JNK and AKT signaling pathway and Bcl2/Bax/Caspase axis. In conclusion, our results indicate BCAA metabolism is involved in NAFLD and BCATc Inhibitor 2 protects against oleic acid-induced lipid accumulation and apoptosis. These findings suggest that BCATc Inhibitor 2 is a promising candidate drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Zhuo Lu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Feng Sun
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xiao-An Pan
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Xin-Hui Qu
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ping Yang
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhi-Ping Chen
- Department of Critical Care Medicine, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Tao Wang
- Institute of Geriatrics, Jiangxi Provincial People’s Hospital and The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
50
|
Xie X, Hu L, Mi B, Xue H, Hu Y, Panayi AC, Endo Y, Chen L, Yan C, Lin Z, Li H, Zhou W, Liu G. Metformin alleviates bone loss in ovariectomized mice through inhibition of autophagy of osteoclast precursors mediated by E2F1. Cell Commun Signal 2022; 20:165. [PMID: 36284303 PMCID: PMC9594975 DOI: 10.1186/s12964-022-00966-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background Postmenopausal bone loss, mainly caused by excessive bone resorption mediated by osteoclasts, has become a global public health burden. Metformin, a hypoglycemic drug, has been reported to have beneficial effects on maintaining bone health. However, the role and underlying mechanism of metformin in ovariectomized (OVX)-induced bone loss is still vague. Results In this study, we demonstrated for the first time that metformin administration alleviated bone loss in postmenopausal women and ovariectomized mice, based on reduced bone resorption markers, increased bone mineral density (BMD) and improvement of bone microstructure. Then, osteoclast precursors administered metformin in vitro and in vivo were collected to examine the differentiation potential and autophagical level. The mechanism was investigated by infection with lentivirus-mediated BNIP3 or E2F1 overexpression. We observed a dramatical inhibition of autophagosome synthesis and osteoclast formation and activity. Treatment with RAPA, an autophagy activator, abrogated the metformin-mediated autophagy downregulation and inhibition of osteoclastogenesis. Additionally, overexpression of E2F1 demonstrated that reduction of OVX-upregulated autophagy mediated by metformin was E2F1 dependent. Mechanistically, metformin-mediated downregulation of E2F1 in ovariectomized mice could downregulate BECN1 and BNIP3 levels, which subsequently perturbed the binding of BECN1 to BCL2. Furthermore, the disconnect between BECN1 and BCL2 was shown by BNIP3 overexpression. Conclusion In summary, we demonstrated the effect and underlying mechanism of metformin on OVX-induced bone loss, which could be, at least in part, ascribed to its role in downregulating autophagy during osteoclastogenesis via E2F1-dependent BECN1 and BCL2 downregulation, suggesting that metformin or E2F1 inhibitor is a potential agent against postmenopausal bone loss. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00966-5.
Collapse
Affiliation(s)
- Xudong Xie
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Liangcong Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Hang Xue
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yiqiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Yori Endo
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Chenchen Yan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|