1
|
Hou X, Ren C, Jin J, Chen Y, Lyu X, Bi K, Carrillo ND, Cryns VL, Anderson RA, Sun J, Chen M. Phosphoinositide signalling in cell motility and adhesion. Nat Cell Biol 2025:10.1038/s41556-025-01647-4. [PMID: 40169755 DOI: 10.1038/s41556-025-01647-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025]
Abstract
Cell motility and adhesion are fundamental components for diverse physiological functions, including embryonic development, immune responses and tissue repair. Dysregulation of these processes can lead to a range of diseases, including cancer. Cell motility and adhesion are complex and often require regulation by an intricate network of signalling pathways, with phosphatidylinositol phosphates (PIPs) having a central role. PIPs are derived from phosphatidylinositol phosphorylation and are instrumental in mediating membrane dynamics, intracellular trafficking, cytoskeletal organization and signal transduction, all of which are crucial for cellular responses to environmental stimuli. Here we discuss the mechanisms through which PIPs modulate cell motility and adhesion by examining their roles at focal adhesions, within the cytoskeleton, at protein scaffolds and in the nucleus. By providing a comprehensive overview of PIP signalling, this Review underscores their significance in maintaining cellular homeostasis and highlights their potential as therapeutic targets in diseases characterized by aberrant cell motility and adhesion.
Collapse
Affiliation(s)
- Xiaoting Hou
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chang Ren
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jing Jin
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Critical Care Medicine, Shenzhen People's Hospital (The Second Clinical Medical College at Jinan University and The First Affiliated Hospital at the Southern University of Science and Technology), Shenzhen, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China
| | - Yu Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xinyu Lyu
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Kangle Bi
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Noah D Carrillo
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Vincent L Cryns
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jichao Sun
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- Department of Critical Care Medicine, Shenzhen People's Hospital (The Second Clinical Medical College at Jinan University and The First Affiliated Hospital at the Southern University of Science and Technology), Shenzhen, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, Shenzhen, China.
| | - Mo Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
2
|
He B, Wood KH, Li ZJ, Ermer JA, Li J, Bastow ER, Sakaram S, Darcy PK, Spalding LJ, Redfern CT, Canes J, Oliveira M, Prat A, Cortes J, Thompson EW, Littlefield BA, Redfern A, Ganss R. Selective tubulin-binding drugs induce pericyte phenotype switching and anti-cancer immunity. EMBO Mol Med 2025:10.1038/s44321-025-00222-6. [PMID: 40140727 DOI: 10.1038/s44321-025-00222-6] [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: 10/10/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
The intratumoral immune milieu is crucial for the success of anti-cancer immunotherapy. We show here that stromal modulation by the tubulin-binding anti-cancer drugs combretastatin A4 (CA-4) and eribulin improved tumor perfusion and anti-tumor immunity. This was achieved by reverting highly proliferative, angiogenic pericytes into a quiescent, contractile state which durably normalized the vascular bed and reduced hypoxia in mouse models of pancreatic neuroendocrine cancer, breast cancer and melanoma. The crucial event in pericyte phenotype switching was RhoA kinase activation, which distinguished CA-4 and eribulin effects from other anti-mitotic drugs such as paclitaxel and vinorelbine. Importantly, eribulin pre-treatment sensitized tumors for adoptive T cell therapy or checkpoint inhibition resulting in effector cell infiltration and better survival outcomes in mice. In breast cancer patients, eribulin neoadjuvant treatment induced pericyte maturity and RhoA kinase activity indicating similar vessel remodeling effects as seen in mice. Moreover, a contractile pericyte signature was associated with overall better survival outcome in two independent breast cancer cohorts. This underscores the potential of re-purposing specific anti-cancer drugs to enable synergistic complementation with emerging immunotherapies.
Collapse
Affiliation(s)
- Bo He
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Kira H Wood
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Zhi-Jie Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Guangdong, P. R. China
| | - Judith A Ermer
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Ji Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Edward R Bastow
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | | | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa J Spalding
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Cameron T Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Jordi Canes
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Mafalda Oliveira
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Aleix Prat
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Javier Cortes
- SOLTI Cancer Research Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Jersey City, NJ, USA
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Sao Paulo, Brazil
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Erik W Thompson
- School of Biomedical Sciences and Centre for Genomics and Personalised Health, Faculty of Health, Queensland University of Technology (QUT) and Translational Research Institute, Brisbane, Australia
| | | | - Andrew Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Fiona Stanley Hospital, Perth, WA, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
3
|
Yu M, Yu H, Wang H, Xu X, Sun Z, Chen W, Yu M, Liu C, Jiang M, Zhang X. Tumor‑associated macrophages activated in the tumor environment of hepatocellular carcinoma: Characterization and treatment (Review). Int J Oncol 2024; 65:100. [PMID: 39239752 PMCID: PMC11387121 DOI: 10.3892/ijo.2024.5688] [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: 05/09/2024] [Accepted: 08/08/2024] [Indexed: 09/07/2024] Open
Abstract
Hepatocellular carcinoma (HCC) tissue is rich in dendritic cells, T cells, B cells, macrophages, natural killer cells and cellular stroma. Together they form the tumor microenvironment (TME), which is also rich in numerous cytokines. Tumor‑associated macrophages (TAMs) are involved in the regulation of tumor development. TAMs in HCC receive stimuli in different directions, polarize in different directions and release different cytokines to regulate the development of HCC. TAMs are mostly divided into two cell phenotypes: M1 and M2. M1 TAMs secrete pro‑inflammatory mediators, and M2 TAMs secrete a variety of anti‑inflammatory and pro‑tumorigenic substances. The TAM polarization in HCC tumors is M2. Both direct and indirect methods for TAMs to regulate the development of HCC are discussed. TAMs indirectly support HCC development by promoting peripheral angiogenesis and regulating the immune microenvironment of the TME. In terms of the direct regulation between TAMs and HCC cells, the present review mainly focuses on the molecular mechanism. TAMs are involved in both the proliferation and apoptosis of HCC cells to regulate the quantitative changes of HCC, and stimulate the related invasive migratory ability and cell stemness of HCC cells. The present review aims to identify immunotherapeutic options based on the mechanisms of TAMs in the TME of HCC.
Collapse
Affiliation(s)
- Mingkai Yu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Haixia Yu
- Pharmacy College, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiaoya Xu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Zhaoqing Sun
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Wenshuai Chen
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Miaomiao Yu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Chunhua Liu
- Department of Physiology and Neurobiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Mingchun Jiang
- Department of Physiology and Neurobiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, Shandong 250000, P.R. China
| | - Xiaowei Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| |
Collapse
|
4
|
Roshan MK, Afshari AR, Mirzavi F, Mousavi SH, Soukhtanloo M. Combretastatin A-4 suppresses the invasive and metastatic behavior of glioma cells and induces apoptosis in them: in-vitro study. Med Oncol 2023; 40:331. [PMID: 37838642 DOI: 10.1007/s12032-023-02197-1] [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: 08/10/2023] [Accepted: 09/19/2023] [Indexed: 10/16/2023]
Abstract
The most common primary brain malignancy, glioblastoma multiforme, is tremendously resistant to conventional treatments due to its potency for metastasis to surrounding brain tissue. Temozolomide is a chemotherapeutic agent that currently is administrated during the treatment procedure. Studies have attempted to investigate new agents with higher effectiveness and fewer side effects. Combretastatin A-4 (CA-4), a natural compound derived from Combretum caffrum, has been recently considered for its potent antitumor activities in a wide variety of preclinical solid tumor models. Our findings have shown that CA-4 exerts potent anti-proliferative and apoptotic effects on glioma cells, and ROS generation may be involved in these cellular events. CA-4 has imposed G2 arrest in U-87 cells. We also observed that CA-4 significantly reduced the migration and invasion capability of U-87 cells. Furthermore, the gene expression and enzyme activity of MMP-2 and MMP-9 were significantly inhibited in the presence of CA-4. We also observed a considerable decrease in PI3K and Akt protein expression following treatment with CA-4. In conclusion, our findings showed significant apoptogenic and anti-metastatic effects of CA-4 on glioma cells and also suggested that the PI3K/Akt/MMP-2/-9 and also ROS pathway might play roles in these cellular events.
Collapse
Affiliation(s)
- Mostafa Karimi Roshan
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Hadi Mousavi
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Deng S, Leong HC, Datta A, Gopal V, Kumar AP, Yap CT. PI3K/AKT Signaling Tips the Balance of Cytoskeletal Forces for Cancer Progression. Cancers (Basel) 2022; 14:1652. [PMID: 35406424 PMCID: PMC8997157 DOI: 10.3390/cancers14071652] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
The PI3K/AKT signaling pathway plays essential roles in multiple cellular processes, which include cell growth, survival, metabolism, and motility. In response to internal and external stimuli, the PI3K/AKT signaling pathway co-opts other signaling pathways, cellular components, and cytoskeletal proteins to reshape individual cells. The cytoskeletal network comprises three main components, which are namely the microfilaments, microtubules, and intermediate filaments. Collectively, they are essential for many fundamental structures and cellular processes. In cancer, aberrant activation of the PI3K/AKT signaling cascade and alteration of cytoskeletal structures have been observed to be highly prevalent, and eventually contribute to many cancer hallmarks. Due to their critical roles in tumor progression, pharmacological agents targeting PI3K/AKT, along with cytoskeletal components, have been developed for better intervention strategies against cancer. In our review, we first discuss existing evidence in-depth and then build on recent advances to propose new directions for therapeutic intervention.
Collapse
Affiliation(s)
- Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Hin Chong Leong
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Arpita Datta
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
6
|
Post-translational modifications of tubulin: their role in cancers and the regulation of signaling molecules. Cancer Gene Ther 2021; 30:521-528. [PMID: 34671113 DOI: 10.1038/s41417-021-00396-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/10/2021] [Accepted: 09/28/2021] [Indexed: 11/09/2022]
Abstract
Microtubules play an important role in regulating several vital cellular activities, including cell division and tissue organization, through their dynamic protofilament network. In addition to forming the cytoskeleton, microtubules regulate the intracellular trafficking of cytoplasmic components and various signaling molecules, depending on the presence of post-transitional modifications (PTMs) and binding proteins. Accumulating evidence indicates the significant role of microtubule PTMs on cancer behavior. The PTMs that frequently occur on microtubules include acetylation, detyrosination, tyrosination, polyglutamylation, and polyglycylation. Alterations in these PTMs cause global effects on intracellular signal transduction, strongly linked to cancer pathogenesis. This review provides an update on the role of microtubule PTMs in cancer aggressiveness, particularly regarding cell death, sensitivity to chemotherapy, cell migration, and invasion. Additionally, it provides a mechanistic explanation of the molecular signaling pathways involved. This information might prove useful for predictive or therapeutic purposes.
Collapse
|
7
|
Özdemİr A, Ark M. A novel ROCK inhibitor: off-target effects of metformin. ACTA ACUST UNITED AC 2021; 45:35-45. [PMID: 33597820 PMCID: PMC7877715 DOI: 10.3906/biy-2004-12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022]
Abstract
In drug discovery, most small molecules cannot cross many stages, only a few can become drug candidates. Once the drug molecule is approved and marketed, nontarget effects that are not easily distinguishable from the actual target of the drugs might be evaluated. This situation restricts the treatment. Thus, the discovery of new drugs is a very long and expensive process. In recent years, without developing new drugs, the approach of using different and new target molecules in new indications apart from the indications of licensed drug molecules has gained importance.In this study, using the connectivity map program, it was determined that metformin and tolbutamide used in the treatment of type II diabetes had the potential to inhibit Rho kinase. In the experimental results to confirm this data, it has been shown that metformin and tolbutamide decrease the cell area within 24 h and metformin inhibits the activation of Rho kinase in MCF-7 cells.These results indicate that metformin, which is used in the treatment of type II diabetes, acts as a ROCK inhibitor. Metformin has potential in the treatment of various pathological conditions in which Rho kinase has a role.
Collapse
Affiliation(s)
- Aysun Özdemİr
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara Turkey
| | - Mustafa Ark
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Ankara Turkey
| |
Collapse
|
8
|
Gao S, Wu X, Wang L, Bu T, Perrotta A, Guaglianone G, Silvestrini B, Sun F, Cheng CY. Signaling Proteins That Regulate Spermatogenesis Are the Emerging Target of Toxicant-Induced Male Reproductive Dysfunction. Front Endocrinol (Lausanne) 2021; 12:800327. [PMID: 35002976 PMCID: PMC8739942 DOI: 10.3389/fendo.2021.800327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/22/2021] [Indexed: 12/05/2022] Open
Abstract
There is emerging evidence that environmental toxicants, in particular endocrine disrupting chemicals (EDCs) such as cadmium and perfluorooctanesulfonate (PFOS), induce Sertoli cell and testis injury, thereby perturbing spermatogenesis in humans, rodents and also widelife. Recent studies have shown that cadmium (e.g., cadmium chloride, CdCl2) and PFOS exert their disruptive effects through putative signaling proteins and signaling cascade similar to other pharmaceuticals, such as the non-hormonal male contraceptive drug adjudin. More important, these signaling proteins were also shown to be involved in modulating testis function based on studies in rodents. Collectively, these findings suggest that toxicants are using similar mechanisms that used to support spermatogenesis under physiological conditions to perturb Sertoli and testis function. These observations are physiologically significant, since a manipulation on the expression of these signaling proteins can possibly be used to manage the toxicant-induced male reproductive dysfunction. In this review, we highlight some of these findings and critically evaluate the possibility of using this approach to manage toxicant-induced defects in spermatrogenesis based on recent studies in animal models.
Collapse
Affiliation(s)
- Sheng Gao
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, China
| | - Lingling Wang
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, China
| | - Adolfo Perrotta
- Department of Translational & Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Guaglianone
- Department of Hospital Pharmacy, “Azienda Sanitaria Locale (ASL) Roma 4”, Civitavecchia, Italy
| | - Bruno Silvestrini
- Institute of Pharmacology and Pharmacognosy, Sapienza University of Rome, Rome, Italy
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: C. Yan Cheng, ; Fei Sun,
| | - C. Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, China
- *Correspondence: C. Yan Cheng, ; Fei Sun,
| |
Collapse
|
9
|
The dynactin subunit DCTN1 controls osteoclastogenesis via the Cdc42/PAK2 pathway. Exp Mol Med 2020; 52:514-528. [PMID: 32210358 PMCID: PMC7156411 DOI: 10.1038/s12276-020-0406-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/04/2020] [Accepted: 02/12/2020] [Indexed: 01/11/2023] Open
Abstract
Osteoclasts (OCs), cells specialized for bone resorption, are generated from monocyte/macrophage precursors by a differentiation process governed by RANKL. Here, we show that DCTN1, a key component of the dynactin complex, plays important roles in OC differentiation. The expression of DCTN1 was upregulated by RANKL. The inhibition of DCTN1 expression by gene knockdown suppressed OC formation, bone resorption, and the induction of NFATc1 and c-Fos, critical transcription factors for osteoclastogenesis. More importantly, the activation of Cdc42 by RANKL was inhibited upon DCTN1 silencing. The forced expression of constitutively active Cdc42 restored the OC differentiation of precursors with DCTN1 deletion. In addition, PAK2 was found to be activated by RANKL and to function downstream of Cdc42. The DCTN1-Cdc42 axis also inhibited apoptosis and caspase-3 activation. Furthermore, the anti-osteoclastogenic effect of DCTN1 knockdown was verified in an animal model of bone erosion. Intriguingly, DCTN1 overexpression was also detrimental to OC differentiation, suggesting that DCTN1 should be regulated at the appropriate level for effective osteoclastogenesis. Collectively, our results reveal that DCTN1 participates in the activation of Cdc42/PAK2 signaling and the inhibition of apoptosis during osteoclastogenesis. A critical mechanism for maintaining bone health uncovered by scientists in South Korea could provide insights into bone disease development. Bone remodeling is a lifetime process of bone generation that ensures bones remain healthy. Osteoclasts (OC), cells that break down bone, differentiate from white blood cell populations. Disruption to OC formation and function plays a critical role in bone diseases, yet the regulatory mechanisms in OC generation are unclear. Hong-Hee Kim at Seoul National University and co-workers investigated the role of a protein called DCTN1, which is involved in skeletal assembly processes. The team found that inhibiting DCTN1 suppressed the expression of key proteins needed for OC formation in cell cultures and mouse models. Overexpressing DCTN1 was equally damaging, suggesting the protein plays a key regulatory role.
Collapse
|
10
|
The Cytoskeleton as Regulator of Cell Signaling Pathways. Trends Biochem Sci 2019; 45:96-107. [PMID: 31812462 DOI: 10.1016/j.tibs.2019.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
During interphase, filamentous actin, microtubules, and intermediate filaments regulate cell shape, motility, transport, and interactions with the environment. These activities rely on signaling events that control cytoskeleton properties. Recent studies uncovered mechanisms that go far beyond this one-directional flow of information. Thus, the three branches of the cytoskeleton impinge on signaling pathways to determine their activities. We propose that this regulatory role of the cytoskeleton provides sophisticated mechanisms to control the spatiotemporal output and the intensity of signaling events. Specific examples emphasize these emerging contributions of the cytoskeleton to cell physiology. In our opinion, further exploration of these pathways will uncover new concepts of cellular communication that originate from the cytoskeleton.
Collapse
|
11
|
Wen W, Marcinkowski E, Luyimbazi D, Luu T, Xing Q, Yan J, Wang Y, Wu J, Guo Y, Tully D, Han ES, Yost SE, Yuan Y, Yim JH. Eribulin Synergistically Increases Anti-Tumor Activity of an mTOR Inhibitor by Inhibiting pAKT/pS6K/pS6 in Triple Negative Breast Cancer. Cells 2019; 8:cells8091010. [PMID: 31480338 PMCID: PMC6770784 DOI: 10.3390/cells8091010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
Unlike other breast cancer subtypes, patients with triple negative breast cancer (TNBC) have poor outcomes and no effective targeted therapies, leaving an unmet need for therapeutic targets. Efforts to profile these tumors have revealed the PI3K/AKT/mTOR pathway as a potential target. Activation of this pathway also contributes to resistance to anti-cancer agents, including microtubule-targeting agents. Eribulin is one such microtubule-targeting agent that is beneficial in treating taxane and anthracycline refractory breast cancer. In this study, we compared the effect of eribulin on the PI3K/AKT/mTOR pathway with other microtubule-targeting agents in TNBC. We found that the phosphorylation of AKT was suppressed by eribulin, a microtubule depolymerizing agent, but activated by paclitaxel, a microtubule stabilizing agent. The combination of eribulin and everolimus, an mTOR inhibitor, resulted in an increased reduction of p-S6K1 and p-S6, a synergistic inhibition of cell survival in vitro, and an enhanced suppression of tumor growth in two orthotopic mouse models. These findings provide a preclinical foundation for targeting both the microtubule cytoskeleton and the PI3K/AKT/mTOR pathway in the treatment of refractory TNBC.
Collapse
Affiliation(s)
- Wei Wen
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Emily Marcinkowski
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - David Luyimbazi
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Thehang Luu
- Department of Medical Oncology and Molecular Therapy, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Quanhua Xing
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Jin Yan
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Yujun Wang
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Jun Wu
- Department of Comparative Medicine, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Yuming Guo
- Department of Comparative Medicine, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Dylan Tully
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Ernest S Han
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Susan E Yost
- Department of Medical Oncology and Molecular Therapy, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - Yuan Yuan
- Department of Medical Oncology and Molecular Therapy, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA
| | - John H Yim
- Division of Surgery, Beckman Research Institute, City of Hope National Medical Center, 1500 East Duarte Rd., Duarte, CA 91010, USA.
| |
Collapse
|
12
|
Sugiyama MG, Fairn GD, Antonescu CN. Akt-ing Up Just About Everywhere: Compartment-Specific Akt Activation and Function in Receptor Tyrosine Kinase Signaling. Front Cell Dev Biol 2019; 7:70. [PMID: 31131274 PMCID: PMC6509475 DOI: 10.3389/fcell.2019.00070] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
The serine/threonine kinase Akt is a master regulator of many diverse cellular functions, including survival, growth, metabolism, migration, and differentiation. Receptor tyrosine kinases are critical regulators of Akt, as a result of activation of phosphatidylinositol-3-kinase (PI3K) signaling leading to Akt activation upon receptor stimulation. The signaling axis formed by receptor tyrosine kinases, PI3K and Akt, as well as the vast range of downstream substrates is thus central to control of cell physiology in many different contexts and tissues. This axis must be tightly regulated, as disruption of PI3K-Akt signaling underlies the pathology of many diseases such as cancer and diabetes. This sophisticated regulation of PI3K-Akt signaling is due in part to the spatial and temporal compartmentalization of Akt activation and function, including in specific nanoscale domains of the plasma membrane as well as in specific intracellular membrane compartments. Here, we review the evidence for localized activation of PI3K-Akt signaling by receptor tyrosine kinases in various specific cellular compartments, as well as that of compartment-specific functions of Akt leading to control of several fundamental cellular processes. This spatial and temporal control of Akt activation and function occurs by a large number of parallel molecular mechanisms that are central to regulation of cell physiology.
Collapse
Affiliation(s)
- Michael G. Sugiyama
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Gregory D. Fairn
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Costin N. Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
13
|
Pongrakhananon V, Wattanathamsan O, Takeichi M, Chetprayoon P, Chanvorachote P. Loss of CAMSAP3 promotes EMT via the modification of microtubule-Akt machinery. J Cell Sci 2018; 131:jcs.216168. [PMID: 30282632 DOI: 10.1242/jcs.216168] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) plays pivotal roles in a variety of biological processes, including cancer invasion. Although EMT involves alterations of cytoskeletal proteins such as microtubules, the role of microtubules in EMT is not fully understood. Microtubule dynamics are regulated by microtubule-binding proteins, and one such protein is CAMSAP3, which binds the minus-end of microtubules. Here, we show that CAMSAP3 is important to preserve the epithelial phenotypes in lung carcinoma cells. Deletion of CAMSAP3 in human lung carcinoma-derived cell lines showed that CAMSAP3-deficient cells acquired increased mesenchymal features, mostly at the transcriptional level. Analysis of the mechanisms underlying these changes demonstrated that tubulin acetylation was dramatically increased following CAMSAP3 removal, leading to the upregulation of Akt proteins (also known as protein kinase B proteins, hereafter Akt) activity, which is known to promote EMT. These findings suggest that CAMSAP3 functions to protect lung carcinoma cells against EMT by suppressing Akt activity via microtubule regulation and that CAMSAP3 loss promotes EMT in these cells.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Varisa Pongrakhananon
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok 10330, Thailand .,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Onsurang Wattanathamsan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.,Inter-department Program of Pharmacology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Masatoshi Takeichi
- Laboratory for Cell adhesion and Tissue Patterning, RIKEN Center for Developmental Biology and RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Paninee Chetprayoon
- Nano Safety and Risk Assessment Laboratory, National Nanotechnology Center, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Pithi Chanvorachote
- Cell-Based Drug and Health Product Development Research Unit, Chulalongkorn University, Bangkok 10330, Thailand.,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
14
|
Rozic G, Paukov L, Jakubikova J, Ben-Shushan D, Duek A, Leiba A, Avigdor A, Nagler A, Leiba M. The novel compound STK405759 is a microtubule-targeting agent with potent and selective cytotoxicity against multiple myeloma in vitro and in vivo. Oncotarget 2018; 7:62572-62584. [PMID: 27613836 PMCID: PMC5308747 DOI: 10.18632/oncotarget.11539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/09/2016] [Indexed: 12/31/2022] Open
Abstract
Despite advances in treatment, multiple myeloma (MM) remains incurable. Here we propose the use of STK405759, a novel microtubule targeting agent (MTA) and member of the furan metotica family for MM therapy. STK405759 inhibited tubulin polymerization in a cell-free system and in myeloma cells. This molecule had potent cytotoxic activity against several MM cell lines and patient-derived MM cells. Moreover, STK405759 demonstrated cytotoxicity against drug-resistant myeloma cells that overexpressed the P-glycoprotein drug-efflux pump. STK405759 was not cytotoxic to peripheral blood mononuclear cells, including activated B and T lymphocytes. This compound caused mitotic arrest and apoptosis of myeloma cells characterized by cleavage of poly (ADP-ribose) polymerase-1 and caspase-8, as well as decreased protein expression of mcl-1. The combination of STK405759 with bortezomib, lenalidomide or dexamethasone had synergistic cytotoxic activity. In in vivo studies, STK405759-treated mice had significantly decreased MM tumor burden and prolonged survival compared to vehicle treated- mice. These results provide a rationale for further evaluation of STK405759 as monotherapy or part of combination therapy for treating patients with MM.
Collapse
Affiliation(s)
- Gabriela Rozic
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Lena Paukov
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Jana Jakubikova
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dikla Ben-Shushan
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Adrian Duek
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Adi Leiba
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Medical Education, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Abraham Avigdor
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Merav Leiba
- Division of Hematology and BMT, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
15
|
Itoh Y. A balancing Akt: How to fine-tune neuronal migration speed. NEUROGENESIS 2016; 3:e1256854. [PMID: 28405587 DOI: 10.1080/23262133.2016.1256854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/01/2016] [Indexed: 01/17/2023]
Abstract
In the developing mammalian neocortex, newborn neurons produced deep in the brain from neural stem/progenitor cells set out for a long journey to reach their final destination at the brain surface. This process called radial neuronal migration is prerequisite for the formation of appropriate layers and networks in the cortex, and its dysregulation has been implicated in cortical malformation and neurological diseases. Considering a fine correlation between temporal order of cortical neuronal cell types and their spatial distribution, migration speed needs to be tightly controlled to achieve correct neocortical layering, although the underlying molecular mechanisms remain not fully understood. Recently, we discovered that the kinase Akt and its activator PDK1 regulate the migration speed of mouse neocortical neurons through the cortical plate. We further found that the PDK1-Akt pathway controls coordinated movement of the nucleus and the centrosome during migration. Our data also suggested that control of neuronal migration by the PDK1-Akt pathway is mediated at the level of microtubules, possibly through regulation of the cytoplasmic dynein/dynactin complex. Our findings thus identified a signaling pathway controlling neuronal migration speed as well as a novel link between Akt signaling and cytoplasmic dynein/dynactin complex.
Collapse
Affiliation(s)
- Yasuhiro Itoh
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University , Cambridge, MA, USA
| |
Collapse
|
16
|
Li N, Mruk DD, Tang EI, Lee WM, Wong CKC, Cheng CY. Formin 1 Regulates Microtubule and F-Actin Organization to Support Spermatid Transport During Spermatogenesis in the Rat Testis. Endocrinology 2016; 157:2894-908. [PMID: 27145014 PMCID: PMC4929546 DOI: 10.1210/en.2016-1133] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Formin 1 confers actin nucleation by generating long stretches of actin microfilaments to support cell movement, cell shape, and intracellular protein trafficking. Formin 1 is likely involved in microtubule (MT) dynamics due to the presence of a MT binding domain near its N terminus. Here, formin 1 was shown to structurally interact with α-tubulin, the building block of MT, and also end-binding protein 1 (a MT plus [+]-end-binding protein that stabilizes MT) in the testis. Knockdown of formin 1 in Sertoli cells with an established tight junction barrier was found to induce down-regulation of detyrosinated MT (a stabilized form of MT), and disorganization of MTs, in which MTs were retracted from the cell cortical zone, mediated through a loss of MT polymerization and down-regulation of Akt1/2 signaling kinase. An efficient knockdown of formin 1 in the testis reduced the number of track-like structures conferred by MTs and F-actin considerably, causing defects in spermatid and phagosome transport across the seminiferous epithelium. In summary, formin1 maintains MT and F-actin track-like structures to support spermatid and phagosome transport across the seminiferous epithelium during spermatogenesis.
Collapse
Affiliation(s)
- Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (N.L., D.D.M., E.I.T., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065-6307; College of Life Sciences and Oceanography (N.L.), Shenzhen University, Shenzhen 518060, China; School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China; and Department of Biology (C.K.C.W.), Hong Kong Baptist University, Hong Kong, China
| | - Dolores D Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (N.L., D.D.M., E.I.T., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065-6307; College of Life Sciences and Oceanography (N.L.), Shenzhen University, Shenzhen 518060, China; School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China; and Department of Biology (C.K.C.W.), Hong Kong Baptist University, Hong Kong, China
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (N.L., D.D.M., E.I.T., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065-6307; College of Life Sciences and Oceanography (N.L.), Shenzhen University, Shenzhen 518060, China; School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China; and Department of Biology (C.K.C.W.), Hong Kong Baptist University, Hong Kong, China
| | - Will M Lee
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (N.L., D.D.M., E.I.T., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065-6307; College of Life Sciences and Oceanography (N.L.), Shenzhen University, Shenzhen 518060, China; School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China; and Department of Biology (C.K.C.W.), Hong Kong Baptist University, Hong Kong, China
| | - Chris K C Wong
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (N.L., D.D.M., E.I.T., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065-6307; College of Life Sciences and Oceanography (N.L.), Shenzhen University, Shenzhen 518060, China; School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China; and Department of Biology (C.K.C.W.), Hong Kong Baptist University, Hong Kong, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (N.L., D.D.M., E.I.T., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065-6307; College of Life Sciences and Oceanography (N.L.), Shenzhen University, Shenzhen 518060, China; School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China; and Department of Biology (C.K.C.W.), Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
17
|
Mao J, Wang D, Wang Z, Tian W, Li X, Duan J, Wang Y, Yang H, You L, Cheng Y, Bian J, Chen Z, Yang Y. Combretastatin A-1 phosphate, a microtubule inhibitor, acts on both hepatocellular carcinoma cells and tumor-associated macrophages by inhibiting the Wnt/β-catenin pathway. Cancer Lett 2016; 380:134-43. [PMID: 27349166 DOI: 10.1016/j.canlet.2016.06.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023]
Abstract
Combretastatin A-1 phosphate (CA1P) is a microtubule polymerization inhibitor that binds to the colchicine-binding site of tubulin. We demonstrated that CA1P has outstanding anti-cancer activity against hepatocellular carcinoma (HCC) in vitro and in vivo. As determined by fluorescence staining and western blots (WBs), CA1P induced reactive oxygen species (ROS) accumulation and apoptosis in HepG2 cells with a down-regulation of Mcl-1. Additional studies indicated that CA1P induced microtubule depolymerization-mediated AKT inactivation, which resulted in GSK-3β activation, Wnt/β-Catenin pathway inhibition, and Mcl-1 down-regulation. The induction of HepG2 cell apoptosis by CA1P was prevented by a GSK-3β-specific inhibitor. Furthermore, immunohistochemistry studies on hepatocellular carcinoma mouse models showed that CA1P had activity against tumor-associated macrophages (TAMs). CA1P induced TAM apoptosis in vitro through the same mechanism observed with HepG2 cells, and it eliminated TAMs in the tumor microenvironment (TME) in vivo. In TME, the expression of TGF-β and TNF-α was also altered. The adoptive transfer of macrophages partly rescued the growth of tumor inhibited by CA1P. These findings indicate that CA1P has great potential to impact both cancer cells and the microenvironment, and our results should accelerate the application of CA1P for HCC therapy in clinic.
Collapse
Affiliation(s)
- Jie Mao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Duowei Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Zhuo Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China; School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Xianjing Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jingjing Duan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yun Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Hongbao Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Linjun You
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yan Cheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jinsong Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Zhen Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| | - Yong Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
18
|
Chauhan S, Ahmed Z, Bradfute SB, Arko-Mensah J, Mandell MA, Won Choi S, Kimura T, Blanchet F, Waller A, Mudd MH, Jiang S, Sklar L, Timmins GS, Maphis N, Bhaskar K, Piguet V, Deretic V. Pharmaceutical screen identifies novel target processes for activation of autophagy with a broad translational potential. Nat Commun 2015; 6:8620. [PMID: 26503418 PMCID: PMC4624223 DOI: 10.1038/ncomms9620] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a conserved homeostatic process active in all human cells and affecting a spectrum of diseases. Here we use a pharmaceutical screen to discover new mechanisms for activation of autophagy. We identify a subset of pharmaceuticals inducing autophagic flux with effects in diverse cellular systems modelling specific stages of several human diseases such as HIV transmission and hyperphosphorylated tau accumulation in Alzheimer's disease. One drug, flubendazole, is a potent inducer of autophagy initiation and flux by affecting acetylated and dynamic microtubules in a reciprocal way. Disruption of dynamic microtubules by flubendazole results in mTOR deactivation and dissociation from lysosomes leading to TFEB (transcription factor EB) nuclear translocation and activation of autophagy. By inducing microtubule acetylation, flubendazole activates JNK1 leading to Bcl-2 phosphorylation, causing release of Beclin1 from Bcl-2-Beclin1 complexes for autophagy induction, thus uncovering a new approach to inducing autophagic flux that may be applicable in disease treatment. Autophagy is a homeostatic process that could be a potential drug target in the treatment of disease. Here the authors identify in a pharmaceutical screen flubendazole as an inducer of autophagy initiation and flux by affecting microtubules, mTOR, TFEB and Beclin 1 activity.
Collapse
Affiliation(s)
- Santosh Chauhan
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Zahra Ahmed
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Steven B Bradfute
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - John Arko-Mensah
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Michael A Mandell
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Seong Won Choi
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Tomonori Kimura
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Fabien Blanchet
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Anna Waller
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Michal H Mudd
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Shanya Jiang
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Larry Sklar
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Graham S Timmins
- College of Pharmacy, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Nicole Maphis
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA.,Department of Neurology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Vincent Piguet
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA.,Department of Neurology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| |
Collapse
|