1
|
Wu Q, Xu J, Tang X, Yu J, Li B, Yang J, Zhang X. SB218078 inhibits angiogenesis and epithelial-mesenchymal transition in breast cancer. Front Pharmacol 2025; 16:1552707. [PMID: 40160462 PMCID: PMC11949895 DOI: 10.3389/fphar.2025.1552707] [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: 12/29/2024] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose Small-molecule inhibitors of vascular endothelial growth factor receptor 2 (VEGFR2) face clinical limitations due to adverse effects. This study aimed to evaluate the novel compound SB218078 as a dual-targeting agent against both tumor angiogenesis and epithelial-mesenchymal transition (EMT) in breast cancer, while exploring its mechanisms of action. Methods The anti-angiogenic effects of SB218078 were investigated using in vitro models of endothelial cell migration, invasion, and tube formation, alongside in vivo zebrafish developmental angiogenesis assays. Breast cancer progression was assessed through cellular proliferation, migration, invasion tests, and mouse xenograft models. Mechanistic studies focused on the Chk1/ZEB1 signaling axis, validated through genetic interventions. Results SB218078 effectively suppressed angiogenesis by inhibiting endothelial cell functions and disrupting developmental vascular networks in zebrafish. It also impeded breast cancer cell aggressiveness and tumor growth in vivo. Mechanistically, SB218078 selectively targeted ZEB1-an EMT transcription factor-via Chk1 inhibition, with ZEB1 knockdown mimicking its anti-angiogenic effects, while ZEB1 overexpression reversed this activity. Conclusion SB218078 emerges as a promising dual-action therapeutic candidate for breast cancer, simultaneously blocking angiogenesis and EMT through the Chk1-ZEB1 axis. Its specificity for ZEB1, distinct from other EMT regulators, offers a novel strategy to overcome the limitations of traditional VEGFR2 inhibitors, warranting further preclinical development.
Collapse
Affiliation(s)
- Qianxue Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiawei Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Tang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Jin Yu
- Jiulongpo Center for Disease Control and Prevention of Chongqing, Chongqing, China
| | - Benhua Li
- Department of Clinical Laboratory, The Second People’s Hospital of Liangshan yi Autonomous Prefecture, Xichang, China
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Yang
- Department of Clinical Laboratory, The Second People’s Hospital of Liangshan yi Autonomous Prefecture, Xichang, China
| | - Xiang Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Purkerson MM, Amend SR, Pienta KJ. Bystanders or active players: the role of extra centrosomes as signaling hubs. Cancer Metastasis Rev 2024; 44:1. [PMID: 39570514 PMCID: PMC11582193 DOI: 10.1007/s10555-024-10224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
Centrosomes serve as microtubule-organizing organelles that function in spindle pole organization, cell cycle progression, and cilia formation. A non-canonical role of centrosomes that has gained traction in recent years is the ability to act as signal transduction centers. Centrosome amplification, which includes numerical and structural aberrations of centrosomes, is a candidate hallmark of cancer. The function of centrosomes as signaling centers in cancer cells with centrosome amplification is poorly understood. Establishing a model of how cancer cells utilize centrosomes as signaling platforms will help elucidate the role of extra centrosomes in cancer cell survival and tumorigenesis. Centrosomes act in a diverse array of cellular processes, including cell migration, cell cycle progression, and proteasomal degradation. Given that cancer cells with amplified centrosomes exhibit an increased number and larger area of these signaling platforms, extra centrosomes may be acting to promote tumor development by enhancing signaling kinetics in pathways that are essential for the formation and growth of cancer. In this review, we identify the processes centrosomes are involved in as signal transduction platforms and highlight ways in which cancer cells with centrosome amplification may be taking advantage of these mechanisms.
Collapse
Affiliation(s)
- Madison M Purkerson
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Pharmacology and Molecular Sciences Program, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Sarah R Amend
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kenneth J Pienta
- Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Cancer Ecology Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Pharmacology and Molecular Sciences Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Kinghorn K, Gill A, Marvin A, Li R, Quigley K, Singh S, Gore MT, le Noble F, Gabhann FM, Bautch VL. A defined clathrin-mediated trafficking pathway regulates sFLT1/VEGFR1 secretion from endothelial cells. Angiogenesis 2024; 27:67-89. [PMID: 37695358 PMCID: PMC10881643 DOI: 10.1007/s10456-023-09893-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023]
Abstract
FLT1/VEGFR1 negatively regulates VEGF-A signaling and is required for proper vessel morphogenesis during vascular development and vessel homeostasis. Although a soluble isoform, sFLT1, is often mis-regulated in disease and aging, how sFLT1 is trafficked and secreted from endothelial cells is not well understood. Here we define requirements for constitutive sFLT1 trafficking and secretion in endothelial cells from the Golgi to the plasma membrane, and we show that sFLT1 secretion requires clathrin at or near the Golgi. Perturbations that affect sFLT1 trafficking blunted endothelial cell secretion and promoted intracellular mis-localization in cells and zebrafish embryos. siRNA-mediated depletion of specific trafficking components revealed requirements for RAB27A, VAMP3, and STX3 for post-Golgi vesicle trafficking and sFLT1 secretion, while STX6, ARF1, and AP1 were required at the Golgi. Live-imaging of temporally controlled sFLT1 release from the endoplasmic reticulum showed clathrin-dependent sFLT1 trafficking at the Golgi into secretory vesicles that then trafficked to the plasma membrane. Depletion of STX6 altered vessel sprouting in 3D, suggesting that endothelial cell sFLT1 secretion influences proper vessel sprouting. Thus, specific trafficking components provide a secretory path from the Golgi to the plasma membrane for sFLT1 in endothelial cells that utilizes a specialized clathrin-dependent intermediate, suggesting novel therapeutic targets.
Collapse
Affiliation(s)
- Karina Kinghorn
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Amy Gill
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Allison Marvin
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Renee Li
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Kaitlyn Quigley
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Simcha Singh
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Michaelanthony T Gore
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA
| | - Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, CB No. 3280, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Prakash A, Paunikar S, Webber M, McDermott E, Vellanki SH, Thompson K, Dockery P, Jahns H, Brown JAL, Hopkins AM, Bourke E. Centrosome amplification promotes cell invasion via cell-cell contact disruption and Rap-1 activation. J Cell Sci 2023; 136:jcs261150. [PMID: 37772773 PMCID: PMC10629695 DOI: 10.1242/jcs.261150] [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: 03/08/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
Centrosome amplification (CA) is a prominent feature of human cancers linked to tumorigenesis in vivo. Here, we report mechanistic contributions of CA induction alone to tumour architecture and extracellular matrix (ECM) remodelling. CA induction in non-tumorigenic breast cells MCF10A causes cell migration and invasion, with underlying disruption of epithelial cell-cell junction integrity and dysregulation of expression and subcellular localisation of cell junction proteins. CA also elevates expression of integrin β-3, its binding partner fibronectin-1 and matrix metalloproteinase enzymes, promoting cell-ECM attachment, ECM degradation, and a migratory and invasive cell phenotype. Using a chicken embryo xenograft model for in vivo validation, we show that CA-induced (+CA) MCF10A cells invade into the chick mesodermal layer, with inflammatory cell infiltration and marked focal reactions between chorioallantoic membrane and cell graft. We also demonstrate a key role of small GTPase Rap-1 signalling through inhibition using GGTI-298, which blocked various CA-induced effects. These insights reveal that in normal cells, CA induction alone (without additional oncogenic alterations) is sufficient to confer early pro-tumorigenic changes within days, acting through Rap-1-dependent signalling to alter cell-cell contacts and ECM disruption.
Collapse
Affiliation(s)
- Anu Prakash
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Shishir Paunikar
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Mark Webber
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| | - Emma McDermott
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Sri H. Vellanki
- Department of Surgery, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin D09 DK19, Ireland
| | - Kerry Thompson
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Peter Dockery
- Centre for Microscopy and Imaging, Discipline of Anatomy, School of Medicine, University of Galway, Galway H91 W5P7, Ireland
| | - Hanne Jahns
- Pathobiology Section, School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - James A. L. Brown
- Department of Biological Sciences, University of Limerick, Limerick V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC) and Health Research Institute, University of Limerick, Limerick V94T9PX, Ireland
| | - Ann M. Hopkins
- Department of Surgery, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin D09 DK19, Ireland
| | - Emer Bourke
- Lambe Institute for Translational Research, Discipline of Pathology, Centre for Chromosome Biology, University of Galway, Galway H91 V4AY, Ireland
| |
Collapse
|
5
|
Shakhov AS, Churkina AS, Kotlobay AA, Alieva IB. The Endothelial Centrosome: Specific Features and Functional Significance for Endothelial Cell Activity and Barrier Maintenance. Int J Mol Sci 2023; 24:15392. [PMID: 37895072 PMCID: PMC10607758 DOI: 10.3390/ijms242015392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
This review summarizes information about the specific features that are characteristic of the centrosome and its relationship with the cell function of highly specialized cells, such as endotheliocytes. It is based on data from other researchers and our own long-term experience. The participation of the centrosome in the functional activity of these cells, including its involvement in the performance of the main barrier function of the endothelium, is discussed. According to modern concepts, the centrosome is a multifunctional complex and an integral element of a living cell; the functions of which are not limited only to the ability to polymerize microtubules. The location of the centrosome near the center of the interphase cell, the concentration of various regulatory proteins in it, the organization of the centrosome radial system of microtubules through which intracellular transport is carried out by motor proteins and the involvement of the centrosome in the process of the perception of the external signals and their transmission make this cellular structure a universal regulatory and distribution center, controlling the entire dynamic morphology of an animal cell. Drawing from modern data on the tissue-specific features of the centrosome's structure, we discuss the direct involvement of the centrosome in the performance of functions by specialized cells.
Collapse
Affiliation(s)
- Anton Sergeevich Shakhov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninskye Gory, 119992 Moscow, Russia
| | - Aleksandra Sergeevna Churkina
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninskye Gory, 119992 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1–73, Leninskye Gory, 119992 Moscow, Russia
| | - Anatoly Alekseevich Kotlobay
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Irina Borisovna Alieva
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninskye Gory, 119992 Moscow, Russia
| |
Collapse
|
6
|
Buglak DB, Bougaran P, Kulikauskas MR, Liu Z, Monaghan-Benson E, Gold AL, Marvin AP, Burciu A, Tanke NT, Oatley M, Ricketts SN, Kinghorn K, Johnson BN, Shiau CE, Rogers S, Guilluy C, Bautch VL. Nuclear SUN1 stabilizes endothelial cell junctions via microtubules to regulate blood vessel formation. eLife 2023; 12:83652. [PMID: 36989130 PMCID: PMC10059686 DOI: 10.7554/elife.83652] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Endothelial cells line all blood vessels, where they coordinate blood vessel formation and the blood-tissue barrier via regulation of cell-cell junctions. The nucleus also regulates endothelial cell behaviors, but it is unclear how the nucleus contributes to endothelial cell activities at the cell periphery. Here, we show that the nuclear-localized linker of the nucleoskeleton and cytoskeleton (LINC) complex protein SUN1 regulates vascular sprouting and endothelial cell-cell junction morphology and function. Loss of murine endothelial Sun1 impaired blood vessel formation and destabilized junctions, angiogenic sprouts formed but retracted in SUN1-depleted sprouts, and zebrafish vessels lacking Sun1b had aberrant junctions and defective cell-cell connections. At the cellular level, SUN1 stabilized endothelial cell-cell junctions, promoted junction function, and regulated contractility. Mechanistically, SUN1 depletion altered cell behaviors via the cytoskeleton without changing transcriptional profiles. Reduced peripheral microtubule density, fewer junction contacts, and increased catastrophes accompanied SUN1 loss, and microtubule depolymerization phenocopied effects on junctions. Depletion of GEF-H1, a microtubule-regulated Rho activator, or the LINC complex protein nesprin-1 rescued defective junctions of SUN1-depleted endothelial cells. Thus, endothelial SUN1 regulates peripheral cell-cell junctions from the nucleus via LINC complex-based microtubule interactions that affect peripheral microtubule dynamics and Rho-regulated contractility, and this long-range regulation is important for proper blood vessel sprouting and junction integrity.
Collapse
Affiliation(s)
- Danielle B Buglak
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Pauline Bougaran
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Molly R Kulikauskas
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Ziqing Liu
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Elizabeth Monaghan-Benson
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State UniversityRaleighUnited States
| | - Ariel L Gold
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Allison P Marvin
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Andrew Burciu
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Natalie T Tanke
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Morgan Oatley
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Shea N Ricketts
- Department of Pathology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Karina Kinghorn
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Bryan N Johnson
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Celia E Shiau
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Stephen Rogers
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Christophe Guilluy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State UniversityRaleighUnited States
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
- Department of Biology, The University of North Carolina at Chapel HillChapel HillUnited States
- McAllister Heart Institute, The University of North Carolina at Chapel HillChapel HillUnited States
| |
Collapse
|
7
|
Kinghorn K, Gill A, Marvin A, Li R, Quigley K, le Noble F, Mac Gabhann F, Bautch VL. A defined clathrin-mediated trafficking pathway regulates sFLT1/VEGFR1 secretion from endothelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.27.525517. [PMID: 36747809 PMCID: PMC9900880 DOI: 10.1101/2023.01.27.525517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
FLT1/VEGFR1 negatively regulates VEGF-A signaling and is required for proper vessel morphogenesis during vascular development and vessel homeostasis. Although a soluble isoform, sFLT1, is often mis-regulated in disease and aging, how sFLT1 is trafficked and secreted from endothelial cells is not well understood. Here we define requirements for constitutive sFLT1 trafficking and secretion in endothelial cells from the Golgi to the plasma membrane, and we show that sFLT1 secretion requires clathrin at or near the Golgi. Perturbations that affect sFLT1 trafficking blunted endothelial cell secretion and promoted intracellular mis-localization in cells and zebrafish embryos. siRNA-mediated depletion of specific trafficking components revealed requirements for RAB27A, VAMP3, and STX3 for post-Golgi vesicle trafficking and sFLT1 secretion, while STX6, ARF1, and AP1 were required at the Golgi. Depletion of STX6 altered vessel sprouting in a 3D angiogenesis model, indicating that endothelial cell sFLT1 secretion is important for proper vessel sprouting. Thus, specific trafficking components provide a secretory path from the Golgi to the plasma membrane for sFLT1 in endothelial cells that utilizes a specialized clathrin-dependent intermediate, suggesting novel therapeutic targets.
Collapse
Affiliation(s)
- Karina Kinghorn
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill NC USA
| | - Amy Gill
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore MD, USA
| | - Allison Marvin
- Department of Biology, University of North Carolina, Chapel Hill NC USA
| | - Renee Li
- Department of Biology, University of North Carolina, Chapel Hill NC USA
| | - Kaitlyn Quigley
- Department of Biology, University of North Carolina, Chapel Hill NC USA
| | - Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Feilim Mac Gabhann
- Institute for Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore MD, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, University of North Carolina, Chapel Hill NC USA
- Department of Biology, University of North Carolina, Chapel Hill NC USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill NC USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill NC USA
| |
Collapse
|
8
|
Wang C, Qu K, Wang J, Qin R, Li B, Qiu J, Wang G. Biomechanical regulation of planar cell polarity in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166495. [PMID: 35850177 DOI: 10.1016/j.bbadis.2022.166495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2023]
Abstract
Cell polarity refers to the uneven distribution of certain cytoplasmic components in a cell with a spatial order. The planar cell polarity (PCP), the cell aligns perpendicular to the polar plane, in endothelial cells (ECs) has become a research hot spot. The planar polarity of ECs has a positive significance on the regulation of cardiovascular dysfunction, pathological angiogenesis, and ischemic stroke. The endothelial polarity is stimulated and regulated by biomechanical force. Mechanical stimuli promote endothelial polarization and make ECs produce PCP to maintain the normal physiological and biochemical functions. Here, we overview recent advances in understanding the interplay and mechanism between PCP and ECs function involved in mechanical forces, with a focus on PCP signaling pathways and organelles in regulating the polarity of ECs. And then showed the related diseases caused by ECs polarity dysfunction. This study provides new ideas and therapeutic targets for the treatment of endothelial PCP-related diseases.
Collapse
Affiliation(s)
- Caihong Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Rui Qin
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Bingyi Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| |
Collapse
|
9
|
Wu D, Ji H, Du W, Ren L, Qian G. Mitophagy alleviates ischemia/reperfusion-induced microvascular damage through improving mitochondrial quality control. Bioengineered 2022; 13:3596-3607. [PMID: 35112987 PMCID: PMC8973896 DOI: 10.1080/21655979.2022.2027065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The coronary arteries mainly function to perfuse the myocardium. When coronary artery resistance increases, myocardial perfusion decreases and myocardial remodeling occurs. Mitochondrial damage has been regarded as the primary cause of microvascular dysfunction. In the present study, we explored the effects of mitophagy activation on microvascular damage. Hypoxia/reoxygenation injury induced mitochondrial oxidative stress, thereby promoting mitochondrial dysfunction in endothelial cells. Mitochondrial impairment induced apoptosis, reducing the viability and proliferation of endothelial cells. However, supplementation with the mitophagy inducer urolithin A (UA) preserved mitochondrial function by reducing mitochondrial oxidative stress and stabilizing the mitochondrial membrane potential in endothelial cells. UA also sustained the viability and improved the proliferative capacity of endothelial cells by suppressing apoptotic factors and upregulating cyclins D and E. In addition, UA inhibited mitochondrial fission and restored mitochondrial fusion, which reduced the proportion of fragmented mitochondria within endothelial cells. UA enhanced mitochondrial biogenesis in endothelial cells by upregulating sirtuin 3 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha. These results suggested that activation of mitophagy may reduce hypoxia/reoxygenation-induced cardiac microvascular damage by improving mitochondrial quality control and increasing cell viability and proliferation.
Collapse
Affiliation(s)
- Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Haizhe Ji
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Beijing, China
| | - Wenjuan Du
- Laboratory of Radiation Injury Treatment, Medical Innovation Research Division, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lina Ren
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
10
|
Ma X, Wang S, Cheng H, Ouyang H, Ma X. Melatonin Attenuates Ischemia/Reperfusion-Induced Oxidative Stress by Activating Mitochondrial Fusion in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7105181. [PMID: 35047108 PMCID: PMC8763517 DOI: 10.1155/2022/7105181] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 10/01/2021] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury can stimulate mitochondrial reactive oxygen species production. Optic atrophy 1- (OPA1-) induced mitochondrial fusion is an endogenous antioxidative mechanism that preserves the mitochondrial function. In our study, we investigated whether melatonin augments OPA1-dependent mitochondrial fusion and thus maintains redox balance during myocardial I/R injury. In hypoxia/reoxygenation- (H/R-) treated H9C2 cardiomyocytes, melatonin treatment upregulated OPA1 mRNA and protein expression, thereby enhancing mitochondrial fusion. Melatonin also suppressed apoptosis in H/R-treated cardiomyocytes, as evidenced by increased cell viability, diminished caspase-3 activity, and reduced Troponin T secretion; however, silencing OPA1 abolished these effects. H/R treatment augmented mitochondrial ROS production and repressed antioxidative molecule levels, while melatonin reversed these changes in an OPA1-dependent manner. Melatonin also inhibited mitochondrial permeability transition pore opening and maintained the mitochondrial membrane potential, but OPA1 silencing prevented these outcomes. These results illustrate that melatonin administration alleviates cardiomyocyte I/R injury by activating OPA1-induced mitochondrial fusion and inhibiting mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Xiaoling Ma
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Shengchi Wang
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Hui Cheng
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| | - Haichun Ouyang
- Department of Cardiology, The Seventh Affiliated Hospital, Southern Medical University, China
| | - Xiaoning Ma
- Department of Critical Care Medicine, Shijiazhuang People's Hospital, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
11
|
TMEM60 Promotes the Proliferation and Migration and Inhibits the Apoptosis of Glioma through Modulating AKT Signaling. JOURNAL OF ONCOLOGY 2022; 2022:9913700. [PMID: 35027926 PMCID: PMC8749377 DOI: 10.1155/2022/9913700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/16/2021] [Accepted: 10/26/2021] [Indexed: 02/03/2023]
Abstract
Glioma is a highly fatal malignancy with aggressive proliferation, migration, and invasion metastasis due to aberrant genetic regulation. This work aimed to determine the function of transmembrane protein 60 (TMEM60) during glioma development. The level of TMEM60 in glioma tissues and normal tissues and its correlation with glioma prognosis were checked in The Cancer Genome Atlas (TCGA) database. The levels of TMEM60 in glioma cell lines and normal astrocytes were determined by quantitative real-time PCR and western blotting assay. TMEM60 knockdown and overexpression were conducted, followed by detection of cell viability, migration, invasion, and apoptosis. CCK-8 and colony formation assay were adopted to detect cell viability proliferation. Transwell assay was performed to measure cell migration and invasion. Cell apoptosis was evaluated by flow cytometry. The alternation of key proteins in the PI3K/Akt signaling pathway was measured by western blotting. TMEM60 expression was significantly higher in glioma tissues than that in the healthy control and was correlated with poor overall survival of patients. The protein and mRNA levels of TMEM60 were both elevated in glioma cell lines in comparison with the normal cell lines. Elevated level of TMEM60 led to enhanced proliferation, migration, and invasion and suppressed cell apoptosis. TMEM60 promoted the activation of PI3K/Akt signaling. Our data suggested that TMEM60 plays an oncogenic role in glioma progression via activating the PI3K/Akt signaling pathway.
Collapse
|
12
|
FUNDC1 activates the mitochondrial unfolded protein response to preserve mitochondrial quality control in cardiac ischemia/reperfusion injury. Cell Signal 2022; 92:110249. [DOI: 10.1016/j.cellsig.2022.110249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 12/28/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022]
|
13
|
Shi C, Zhang S, Guo C, Tie J. Yap-Hippo Signaling Activates Mitochondrial Protection and Sustains Breast Cancer Viability under Hypoxic Stress. JOURNAL OF ONCOLOGY 2021; 2021:5212721. [PMID: 34567116 PMCID: PMC8463197 DOI: 10.1155/2021/5212721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 11/17/2022]
Abstract
Yes-associated protein (Yap) is a transcriptional regulator that upregulates oncogenes and downregulates tumor repressor genes. In this study, we analyzed protein expression, RNA transcription, and signaling pathways to determine the function and mechanism of Yap in breast cancer survival during hypoxic stress. Yap transcription was drastically upregulated by hypoxia in a time-dependent manner. siRNA-mediated Yap knockdown attenuated breast cancer viability and impaired cell proliferation under hypoxic conditions. Yap knockdown induced mitochondrial stress, including mitochondrial membrane potential reduction, mitochondrial oxidative stress, and ATP exhaustion after exposure to hypoxia. It also repressed mitochondrial protective systems, including mitophagy and mitochondrial fusion upon exposure to hypoxia. Finally, our data showed that Yap knockdown suppresses MCF-7 cell migration by inhibiting F-actin transcription and promoting lamellipodium degradation under hypoxic stress. Taken together, Yap maintenance of mitochondrial function and activation of F-actin/lamellipodium signaling is required for breast cancer survival, migration, and proliferation under hypoxic stress.
Collapse
Affiliation(s)
- Chen Shi
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Siyuan Zhang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Changkuo Guo
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jian Tie
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
14
|
Zhang H, Qu X, Han L, Di X. Mst2 Overexpression Inhibits Thyroid Carcinoma Growth and Metastasis by Disrupting Mitochondrial Fitness and Endoplasmic Reticulum Homeostasis. JOURNAL OF ONCOLOGY 2021; 2021:1262291. [PMID: 34557228 PMCID: PMC8455210 DOI: 10.1155/2021/1262291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/27/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022]
Abstract
Although the incidence of thyroid carcinoma has increased over the past several decades, it has an excellent prognosis and overall 5-year survival, with a stable mortality rate, except in cases with advanced stages or rare malignant tumor types. Biomarkers have emerged as effective targets of molecular therapy against thyroid carcinoma due to their rapid and convenient detection; however, there has been little clinical application. Macrophage stimulating 2 (Mst2) is a proapoptotic protein with implications in carcinogenesis and metastasis. We found that Mst2 overexpression-induced endoplasmic reticulum (ER) stress in MDA-T32 thyroid carcinoma cells, accompanied by elevated caspase-12 activity, increased apoptotic rate, and reduced cell viability. In addition, Mst2 overexpression contributed to mitochondrial damage, as evidenced by increased mitochondrial oxidative stress and activated the mitochondrial apoptotic pathway. Inhibition of the JNK pathway abolished these effects. These results show Mst2 to be a novel tumor suppressor that induces mitochondrial dysfunction and ER stress via the JNK pathway. Thus, Mst2 could potentially serve as a biomarker for developing targeted therapy against thyroid carcinoma.
Collapse
Affiliation(s)
- Haichao Zhang
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Xin Qu
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Lu Han
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Xu Di
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| |
Collapse
|
15
|
Hou X, Li L, Chen S, Ge C, Shen M, Fu Z. MKP-1 Overexpression Reduces Postischemic Myocardial Damage through Attenuation of ER Stress and Mitochondrial Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8905578. [PMID: 34512872 PMCID: PMC8433005 DOI: 10.1155/2021/8905578] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022]
Abstract
Mitochondrial dysfunction and endoplasmic reticulum (ER) stress contribute to postischemic myocardial damage, but the upstream regulatory mechanisms have not been identified. In this study, we analyzed the role of mitogen-activated protein kinase (MAPK) phosphatase 1 (MKP-1) in the regulation of mitochondrial function and ER stress in hypoxic cardiomyocytes. Our results show that MKP-1 overexpression sustains viability and reduces hypoxia-induced apoptosis among H9C2 cardiomyocytes. MKP-1 overexpression attenuates ER stress and expression of ER stress genes and improves mitochondrial function in hypoxia-treated H9C2 cells. MKP-1 overexpression also increases ATP production and mitochondrial respiration and attenuates mitochondrial oxidative damage in hypoxic cardiomyocytes. Moreover, our results demonstrate that ERK and JNK are the downstream signaling targets of MKP-1 and that MKP-1 overexpression activates ERK, while it inhibits JNK. Inhibition of ERK reduces the ability of MKP-1 to preserve mitochondrial function and ER homeostasis in hypoxic cardiomyocytes. These results show that MKP-1 plays an essential role in the regulation of mitochondrial function and ER stress in hypoxic H9C2 cardiomyocytes through normalization of the ERK pathway and suggest that MKP-1 may serve as a novel target for the treatment of postischemic myocardial injury.
Collapse
Affiliation(s)
- Xiaoling Hou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Lijun Li
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Si Chen
- Department of Cardiology, The First Medical Center of People's Liberation Army General Hospital, China
| | - Cheng Ge
- Department of Cardiology, The First Medical Center of People's Liberation Army General Hospital, China
| | - Mingzhi Shen
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan 572013, China
| | - Zhenhong Fu
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
16
|
LATS2 Deletion Attenuates Myocardial Ischemia-Reperfusion Injury by Promoting Mitochondrial Biogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1058872. [PMID: 34457109 PMCID: PMC8390173 DOI: 10.1155/2021/1058872] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/25/2021] [Accepted: 08/07/2021] [Indexed: 02/03/2023]
Abstract
Reperfusion therapy is the most effective treatment for acute myocardial infarction, but it can damage cardiomyocytes through a mechanism known as myocardial ischemia/reperfusion injury (MIRI). In this study, we investigated whether the large tumor suppressor kinase 2 (LATS2) contributes to the development of myocardial MIRI by disrupting mitochondrial biogenesis. Our in vitro data demonstrate that cardiomyocyte viability was reduced and apoptosis was increased in response to hypoxia/reoxygenation (H/R) injury. However, suppression of LATS2 by shRNA sustained cardiomyocyte viability by maintaining mitochondrial function. Compared to H/R-treated control cardiomyocytes, cardiomyocytes transfected with LATS2 shRNA exhibited increased mitochondrial respiration, improved mitochondrial ATP generation, and more stable mitochondrial membrane potential. LATS2 suppression increased cardiomyocyte viability and mitochondrial biogenesis in a manner dependent on PGC1α, a key regulator of mitochondrial metabolism. These results identify LATS2 as a new inducer of mitochondrial damage and myocardial MIRI and suggest that approaches targeting LATS2 or mitochondrial biogenesis may be beneficial in the clinical management of cardiac MIRI.
Collapse
|
17
|
Protective Effect of Optic Atrophy 1 on Cardiomyocyte Oxidative Stress: Roles of Mitophagy, Mitochondrial Fission, and MAPK/ERK Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3726885. [PMID: 34211623 PMCID: PMC8205577 DOI: 10.1155/2021/3726885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is associated with oxidative stress and mitochondrial damage. However, the regulatory mechanisms underlying cardiomyocyte oxidative stress during myocardial infarction are not fully understood. In the present study, we explored the cardioprotective action of optic atrophy 1- (Opa1-) mediated mitochondrial autophagy (mitophagy) in oxidative stress-challenged cardiomyocytes, with a focus on mitochondrial homeostasis and the MAPK/ERK pathway. Our results demonstrated that overexpression of Opa1 in cultured rat H9C2 cardiomyocytes, a procedure that stimulates mitophagy, attenuates oxidative stress and increases cellular antioxidant capacity. Activation of Opa1-mediated mitophagy suppressed cardiomyocyte apoptosis by downregulating Bax, caspase-9, and caspase-12 and upregulating Bcl-2 and c-IAP. Using mitochondrial tracker staining and a reactive oxygen species indicator, our assays showed that Opa1-mediated mitophagy attenuated mitochondrial fission and reduced ROS production in cardiomyocytes. In addition, we found that inhibition of the MAPK/ERK pathway abolished the antioxidant action of Opa1-mediated mitophagy in these cells. Taken together, our data demonstrate that Opa1-mediated mitophagy protects cardiomyocytes against oxidative stress damage through inhibition of mitochondrial fission and activation of MAPK/ERK signaling. These findings reveal a critical role for Opa1 in the modulation of cardiomyocyte redox balance and suggest a potential target for the treatment of myocardial infarction.
Collapse
|
18
|
Qiu J, Fu Y, Chen Z, Zhang L, Li L, Liang D, Wei F, Wen Z, Wang Y, Liang S. BTK Promotes Atherosclerosis by Regulating Oxidative Stress, Mitochondrial Injury, and ER Stress of Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9972413. [PMID: 34136067 PMCID: PMC8175170 DOI: 10.1155/2021/9972413] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022]
Abstract
Atherosclerosis (AS) is a chronic metabolic disease in arterial walls, characterized by lipid deposition and persistent aseptic inflammation. AS is regarded as the basis of a variety of cardiovascular and cerebrovascular diseases. It is widely acknowledged that macrophages would become foam cells after internalizing lipoprotein particles, which is an initial factor in atherogenesis. Here, we showed the influences of Bruton's tyrosine kinase (BTK) in macrophage-mediated AS and how BTK regulates the inflammatory responses of macrophages in AS. Our bioinformatic results suggested that BTK was a potential hub gene, which is closely related to oxidative stress, ER stress, and inflammation in macrophage-induced AS. Moreover, we found that BTK knockdown could restrain ox-LDL-induced NK-κB signaling activation in macrophages and repressed M1 polarization. The mechanistic studies revealed that oxidative stress, mitochondrial injury, and ER stress in macrophages were also suppressed by BTK knockdown. Furthermore, we found that sh-BTK adenovirus injection could alleviate the severity of AS in ApoE-/- mice induced by a high-fat diet in vivo. Our study suggested that BTK promoted ox-LDL-induced ER stress, oxidative stress, and inflammatory responses in macrophages, and it may be a potential therapeutic target in AS.
Collapse
Affiliation(s)
- Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Yuan Fu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Zhiteng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Lisui Zhang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Ling Li
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Diefei Liang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Feng Wei
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Zhuzhi Wen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Yajing Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| | - Shi Liang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China 510120
| |
Collapse
|
19
|
Ricolo D, Castro-Ribera J, Araújo SJ. Cytoskeletal players in single-cell branching morphogenesis. Dev Biol 2021; 477:22-34. [PMID: 34004181 DOI: 10.1016/j.ydbio.2021.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022]
Abstract
Branching networks are a very common feature of multicellular animals and underlie the formation and function of numerous organs including the nervous system, the respiratory system, the vasculature and many internal glands. These networks range from subcellular structures such as dendritic trees to large multicellular tissues such as the lungs. The production of branched structures by single cells, so called subcellular branching, which has been better described in neurons and in cells of the respiratory and vascular systems, involves complex cytoskeletal remodelling events. In Drosophila, tracheal system terminal cells (TCs) and nervous system dendritic arborisation (da) neurons are good model systems for these subcellular branching processes. During development, the generation of subcellular branches by single-cells is characterized by extensive remodelling of the microtubule (MT) network and actin cytoskeleton, followed by vesicular transport and membrane dynamics. In this review, we describe the current knowledge on cytoskeletal regulation of subcellular branching, based on the terminal cells of the Drosophila tracheal system, but drawing parallels with dendritic branching and vertebrate vascular subcellular branching.
Collapse
Affiliation(s)
- Delia Ricolo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Judith Castro-Ribera
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Sofia J Araújo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
20
|
Bevacizumab-Induced Mitochondrial Dysfunction, Endoplasmic Reticulum Stress, and ERK Inactivation Contribute to Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5548130. [PMID: 33859777 PMCID: PMC8009725 DOI: 10.1155/2021/5548130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 12/26/2022]
Abstract
The molecular mechanisms underlying the cardiotoxicity associated with bevacizumab, a first-line immunotherapeutic agent used to treat lung cancer, are not fully understood. Here, we examined intracellular signal transduction in cardiomyocytes after exposure to different doses of bevacizumab in vitro. Our results demonstrated that bevacizumab significantly and dose-dependently reduces cardiomyocyte viability and increases cell apoptosis. Bevacizumab treatment also led to mitochondrial dysfunction in cardiomyocytes, as evidenced by the decreased ATP production, increased ROS production, attenuated antioxidative enzyme levels, and reduced respiratory complex function. In addition, bevacizumab induced intracellular calcium overload, ER stress, and caspase-12 activation. Finally, bevacizumab treatment inhibited the ERK signaling pathway, which, in turn, significantly reduced cardiomyocyte viability and contributed to mitochondrial dysfunction. Together, our results demonstrate that bevacizumab-mediated cardiotoxicity is associated with mitochondrial dysfunction, ER stress, and ERK pathway inactivation. These findings may provide potential treatment targets to attenuate myocardial injury during lung cancer immunotherapy.
Collapse
|
21
|
Melatonin Attenuates ox-LDL-Induced Endothelial Dysfunction by Reducing ER Stress and Inhibiting JNK/Mff Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5589612. [PMID: 33763168 PMCID: PMC7952160 DOI: 10.1155/2021/5589612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Endothelial dysfunction, which is characterized by damage to the endoplasmic reticulum (ER) and mitochondria, is involved in a variety of cardiovascular disorders. Here, we explored whether mitochondrial damage and ER stress are associated with endothelial dysfunction. We also examined whether and how melatonin protects against oxidized low-density lipoprotein- (ox-LDL-) induced damage in endothelial cells. We found that CHOP, GRP78, and PERK expressions, which are indicative of ER stress, increased significantly in response to ox-LDL treatment. ox-LDL also induced mitochondrial dysfunction as evidenced by decreased mitochondrial membrane potential, increased mitochondrial ROS levels, and downregulation of mitochondrial protective factors. In addition, ox-LDL inhibited antioxidative processes, as evidenced by decreased antioxidative enzyme activity and reduced Nrf2/HO-1 expression. Melatonin clearly reduced ER stress and promoted mitochondrial function and antioxidative processes in the presence of ox-LDL. Molecular investigation revealed that ox-LDL activated the JNK/Mff signaling pathway, and melatonin blocked this effect. These results demonstrate that ox-LDL induces ER stress and mitochondrial dysfunction and activates the JNK/Mff signaling pathway, thereby contributing to endothelial dysfunction. Moreover, melatonin inhibited JNK/Mff signaling and sustained ER homeostasis and mitochondrial function, thereby protecting endothelial cells against ox-LDL-induced damage.
Collapse
|