1
|
Aquino A, Franzese O. Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches. Cancers (Basel) 2025; 17:1547. [PMID: 40361472 PMCID: PMC12072109 DOI: 10.3390/cancers17091547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Dysregulated cell movement is a hallmark of cancer progression and metastasis, the leading cause of cancer-related mortality. The metastatic cascade involves tumour cell migration, invasion, intravasation, dissemination, and colonisation of distant organs. These processes are influenced by reciprocal interactions between cancer cells and the tumour microenvironment (TME), including immune cells, stromal components, and extracellular matrix proteins. The epithelial-mesenchymal transition (EMT) plays a crucial role in providing cancer cells with invasive and stem-like properties, promoting dissemination and resistance to apoptosis. Conversely, the mesenchymal-epithelial transition (MET) facilitates metastatic colonisation and tumour re-initiation. Immune cells within the TME contribute to either anti-tumour response or immune evasion. These cells secrete cytokines, chemokines, and growth factors that shape the immune landscape and influence responses to immunotherapy. Notably, immune checkpoint blockade (ICB) has transformed cancer treatment, yet its efficacy is often dictated by the immune composition of the tumour site. Elucidating the molecular cross-talk between immune and cancer cells, identifying predictive biomarkers for ICB response, and developing strategies to convert cold tumours into immune-active environments is critical to overcoming resistance to immunotherapy and improving patient survival.
Collapse
Affiliation(s)
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
2
|
Yu RY, Jiang WG, Martin TA. The WASP/WAVE Protein Family in Breast Cancer and Their Role in the Metastatic Cascade. Cancer Genomics Proteomics 2025; 22:166-187. [PMID: 39993807 PMCID: PMC11880927 DOI: 10.21873/cgp.20495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 02/26/2025] Open
Abstract
The Wiskott-Aldrich syndrome protein (WASP) and the WASP family verprolin-homologous protein (WAVE) family are essential molecules that connect GTPases to the actin cytoskeleton, thereby controlling actin polymerisation through the actin-related protein 2/3 complex. This control is crucial for forming actin-based membrane protrusions necessary for cell migration and invasion. The elevated expression of WASP/WAVE proteins in invasive breast cancer cells highlights their significant role in promoting cell motility and invasion. This review summarises the discovery, structural properties, and activation mechanisms of WASP/WAVE proteins, focuses on the contribution of the WASP/WAVE family to breast cancer invasion and migration, particularly synthesises the results of nearly a decade of research in this field since 2015. By exploring promising therapeutic strategies for breast cancer, including small molecule inhibitors and biological agents, this review stresses the potential for developing anticancer drugs that target the WASP/WAVE family and associated pathways, intending to improve the prognosis for patients with metastatic breast cancer.
Collapse
Affiliation(s)
- Rhiannon Yannan Yu
- Cardiff-China Medical Research Collaborative, School of Medicine, Cardiff University, Cardiff, U.K
| | - Wen G Jiang
- Cardiff-China Medical Research Collaborative, School of Medicine, Cardiff University, Cardiff, U.K
| | - Tracey A Martin
- Cardiff-China Medical Research Collaborative, School of Medicine, Cardiff University, Cardiff, U.K.
| |
Collapse
|
3
|
Tocci P, Caprara V, Roman C, Sestito R, Rosanò L, Bagnato A. YAP signaling orchestrates the endothelin-1-guided invadopodia formation in high-grade serous ovarian cancer. Biosci Rep 2024; 44:BSR20241320. [PMID: 39495612 DOI: 10.1042/bsr20241320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/06/2024] Open
Abstract
The high-grade serous ovarian cancer (HG-SOC) is a notoriously challenging disease, characterized by a rapid peritoneal dissemination. HG-SOC cells leverage actin-rich membrane protrusions, known as invadopodia, to degrade the surrounding extracellular matrix (ECM) and invade, initiating the metastatic cascade. In HG-SOC, the endothelin-1 (ET-1)/endothelin A receptor (ETAR)-driven signaling coordinates invadopodia activity, however how this axis integrates pro-oncogenic signaling routes, as YAP-driven one, impacting on the invadopodia-mediated ECM degradation and metastatic progression, deserves a deeper investigation. Herein, we observed that downstream of the ET-1/ET-1R axis, the RhoC and Rac1 GTPases, acting as signaling intermediaries, promote the de-phosphorylation and nuclear accumulation of YAP. Conversely, the treatment with the dual ETA/ETB receptor antagonist, macitentan, inhibits the ET-1-driven YAP activity. Similarly, RhoC silencing, or cell transfection with a dominant inactive form of Rac1, restores YAP phosphorylation. Mechanistically, the ET-1R/YAP signal alliance coordinates invadopodia maturation into ECM-degrading structures, indicating how such ET-1R-guided protein network represents a route able to enhance the HG-SOC invasive potential. At functional level, we found that the interconnection between the ET-1R/RhoC and YAP signals is required for MMP-2 and MMP-9 proteolytic functions, cell invasion, and cytoskeleton architecture changes, supporting the HG-SOC metastatic strength. In HG-SOC patient-derived xenografts (PDX) macitentan, turning-off the invadopodia regulators RhoC/YAP, halts the metastatic colonization. ET-1R targeting, hindering the YAP activity, weakens the invadopodia machinery, embodying a promising therapeutic avenue to prevent peritoneal dissemination in HG-SOC.
Collapse
Affiliation(s)
- Piera Tocci
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Valentina Caprara
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Celia Roman
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Rosanna Sestito
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Rosanò
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), Rome 00185, Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
4
|
Zhou Y, Jin Y, Wu T, Wang Y, Dong Y, Chen P, Hu C, Pan N, Ye C, Shen L, Lin M, Fang T, Wu R. New insights on mitochondrial heteroplasmy observed in ovarian diseases. J Adv Res 2024; 65:211-226. [PMID: 38061426 PMCID: PMC11519015 DOI: 10.1016/j.jare.2023.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/26/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024] Open
Abstract
BACKGROUND The reportedly high mutation rate of mitochondrial DNA (mtDNA) may be attributed to the absence of histone protection and complete repair mechanisms. Mitochondrial heteroplasmy refers to the coexistence of wild-type and mutant mtDNA. Most healthy individuals carry a low point mutation load (<1 %) in their mtDNA, typically without any discernible phenotypic effects. However, as it exceeds a certain threshold, it may cause the onset of various diseases. Since the ovary is a highly energy-intensive organ, it relies heavily on mitochondrial function. Mitochondrial heteroplasmy can potentially contribute to a variety of significant ovarian disorders. AIM OF REVIEW In this review, we have elucidated the close relationship between mtDNA heteroplasmy and ovarian diseases, and summarized novel avenues and strategies for the potential treatment of these ovarian diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Mitochondrial heteroplasmy can potentially contribute to a variety of significant ovarian disorders, including polycystic ovary syndrome, premature ovarian insufficiency, and endometriosis. Current strategies related to mitochondrial heteroplasmy are untargeted and have low bioavailability. Nanoparticle delivery systems loaded with mitochondrial modulators, mitochondrial replacement/transplantation therapy, and mitochondria-targeted gene editing therapy may offer promising paths towards potentially more effective treatments for these diseases, despite ongoing challenges.
Collapse
Affiliation(s)
- Yong Zhou
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China; Women's Reproductive Health Key Laboratory of Zhejiang Province, People's Republic of China
| | - Yang Jin
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Tianyu Wu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Yinfeng Wang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Yuanhang Dong
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Pei Chen
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Changchang Hu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Ningping Pan
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Chaoshuang Ye
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Li Shen
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Mengyan Lin
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Tao Fang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China
| | - Ruijin Wu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, Zhejiang 310006, People's Republic of China; Women's Reproductive Health Key Laboratory of Zhejiang Province, People's Republic of China; Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, People's Republic of China.
| |
Collapse
|
5
|
Lu Y, Walji T, Ravaux B, Pandey P, Yang C, Li B, Luvsanjav D, Lam KH, Zhang R, Luo Z, Zhou C, Habela CW, Snapper SB, Li R, Goldhamer DJ, Schmidtke DW, Pan D, Svitkina TM, Chen EH. Spatiotemporal coordination of actin regulators generates invasive protrusions in cell-cell fusion. Nat Cell Biol 2024; 26:1860-1877. [PMID: 39487253 DOI: 10.1038/s41556-024-01541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/11/2024] [Indexed: 11/04/2024]
Abstract
Invasive membrane protrusions play a central role in a variety of cellular processes. Unlike filopodia, invasive protrusions are mechanically stiff and propelled by branched actin polymerization. However, how branched actin filaments are organized to create finger-like invasive protrusions is unclear. Here, by examining the mammalian fusogenic synapse, where invasive protrusions are generated to promote cell membrane juxtaposition and fusion, we have uncovered the mechanism underlying invasive protrusion formation. We show that two nucleation-promoting factors for the Arp2/3 complex, WAVE and N-WASP, exhibit different localization patterns in the protrusions. Whereas WAVE is closely associated with the plasma membrane at the leading edge of the protrusive structures, N-WASP is enriched with WIP along the actin bundles in the shafts of the protrusions. During protrusion initiation and growth, the Arp2/3 complex nucleates branched actin filaments to generate low-density actin clouds in which the large GTPase dynamin organizes the new branched actin filaments into bundles, followed by actin-bundle stabilization by WIP, the latter functioning as an actin-bundling protein. Disruption of any of these components results in defective protrusions and failed myoblast fusion in cultured cells and mouse embryos. Together, our study has revealed the intricate spatiotemporal coordination between two nucleation-promoting factors and two actin-bundling proteins in building invasive protrusions at the mammalian fusogenic synapse and has general implications in understanding invasive protrusion formation in cellular processes beyond cell-cell fusion.
Collapse
Affiliation(s)
- Yue Lu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tezin Walji
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin Ravaux
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pratima Pandey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Changsong Yang
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Bing Li
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Delgermaa Luvsanjav
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin H Lam
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Ruihui Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhou Luo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chuanli Zhou
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christa W Habela
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scott B Snapper
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - David J Goldhamer
- Department of Molecular and Cell Biology, University of Connecticut Stem Cell Institute, Storrs, CT, USA
| | - David W Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
6
|
Friedman-DeLuca M, Karagiannis GS, Condeelis JS, Oktay MH, Entenberg D. Macrophages in tumor cell migration and metastasis. Front Immunol 2024; 15:1494462. [PMID: 39555068 PMCID: PMC11563815 DOI: 10.3389/fimmu.2024.1494462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a phenotypically diverse, highly plastic population of cells in the tumor microenvironment (TME) that have long been known to promote cancer progression. In this review, we summarize TAM ontogeny and polarization, and then explore how TAMs enhance tumor cell migration through the TME, thus facilitating metastasis. We also discuss how chemotherapy and host factors including diet, obesity, and race, impact TAM phenotype and cancer progression. In brief, TAMs induce epithelial-mesenchymal transition (EMT) in tumor cells, giving them a migratory phenotype. They promote extracellular matrix (ECM) remodeling, allowing tumor cells to migrate more easily. TAMs also provide chemotactic signals that promote tumor cell directional migration towards blood vessels, and then participate in the signaling cascade at the blood vessel that allows tumor cells to intravasate and disseminate throughout the body. Furthermore, while chemotherapy can repolarize TAMs to induce an anti-tumor response, these cytotoxic drugs can also lead to macrophage-mediated tumor relapse and metastasis. Patient response to chemotherapy may be dependent on patient-specific factors such as diet, obesity, and race, as these factors have been shown to alter macrophage phenotype and affect cancer-related outcomes. More research on how chemotherapy and patient-specific factors impact TAMs and cancer progression is needed to refine treatment strategies for cancer patients.
Collapse
Affiliation(s)
- Madeline Friedman-DeLuca
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - George S. Karagiannis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Marilyn and Stanley M. Katz Institute for Immunotherapy of Cancer and Inflammatory Disorders, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - John S. Condeelis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Maja H. Oktay
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Cancer Dormancy Institute, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| |
Collapse
|
7
|
Miki T, Hashimoto M, Takahashi H, Shimizu M, Nakayama S, Furuta T, Mihara H. De novo designed YK peptides forming reversible amyloid for synthetic protein condensates in mammalian cells. Nat Commun 2024; 15:8503. [PMID: 39424799 PMCID: PMC11489810 DOI: 10.1038/s41467-024-52708-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/18/2024] [Indexed: 10/21/2024] Open
Abstract
In mammalian cells, protein condensates underlie diverse cell functions. Intensive synthetic biological research has been devoted to fabricating liquid droplets using de novo peptides/proteins designed from scratch in test tubes or bacterial cells. However, the development of de novo sequences for synthetic droplets forming in eukaryotes is challenging. Here, we report YK peptides, comprising 9-15 residues of alternating repeats of tyrosine and lysine, which form reversible amyloid-like fibrils accompanied by binding with poly-anion species such as ATP. By genetically tagging the YK peptide, superfolder GFPs assemble into artificial liquid-like droplets in living cells. Rational design of the YK system allows fine-tuning of the fluidity and construction of multi-component droplets. The YK system not only facilitates intracellular reconstitution of simplified models for natural protein condensates, but it also provides a toolbox for the systematic creation of droplets with different dynamics and composition for in situ evaluation.
Collapse
Affiliation(s)
- Takayuki Miki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan.
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Masahiro Hashimoto
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Hiroki Takahashi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Masatoshi Shimizu
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Sae Nakayama
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Tadaomi Furuta
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| | - Hisakazu Mihara
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8501, Japan
| |
Collapse
|
8
|
Li T, Song Y, Wei L, Song X, Duan R. Disulfidptosis: a novel cell death modality induced by actin cytoskeleton collapse and a promising target for cancer therapeutics. Cell Commun Signal 2024; 22:491. [PMID: 39394612 PMCID: PMC11470700 DOI: 10.1186/s12964-024-01871-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Disulfidptosis is a novel discovered form of programmed cell death (PCD) that diverges from apoptosis, necroptosis, ferroptosis, and cuproptosis, stemming from disulfide stress-induced cytoskeletal collapse. In cancer cells exhibiting heightened expression of the solute carrier family 7 member 11 (SLC7A11), excessive cystine importation and reduction will deplete nicotinamide adenine dinucleotide phosphate (NADPH) under glucose deprivation, followed by an increase in intracellular disulfide stress and aberrant disulfide bond formation within actin networks, ultimately culminating in cytoskeletal collapse and disulfidptosis. Disulfidptosis involves crucial physiological processes in eukaryotic cells, such as cystine and glucose uptake, NADPH metabolism, and actin dynamics. The Rac1-WRC pathway-mediated actin polymerization is also implicated in this cell death due to its contribution to disulfide bond formation. However, the precise mechanisms underlying disulfidptosis and its role in tumors are not well understood. This is probably due to the multifaceted functionalities of SLC7A11 within cells and the complexities of the downstream pathways driving disulfidptosis. This review describes the critical roles of SLC7A11 in cells and summarizes recent research advancements in the potential pathways of disulfidptosis. Moreover, the less-studied aspects of this newly discovered cell death process are highlighted to stimulate further investigations in this field.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Xiangyi Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China.
| |
Collapse
|
9
|
Morishita H, Kawai K, Egami Y, Honda K, Araki N. Live-cell imaging and CLEM reveal the existence of ACTN4-dependent ruffle-edge lamellipodia acting as a novel mode of cell migration. Exp Cell Res 2024; 442:114232. [PMID: 39222868 DOI: 10.1016/j.yexcr.2024.114232] [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: 05/21/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
α-Actinin-4 (ACTN4) expression levels are correlated with the invasive and metastatic potential of cancer cells; however, the underlying mechanism remains unclear. Here, we identified ACTN4-localized ruffle-edge lamellipodia using live-cell imaging and correlative light and electron microscopy (CLEM). BSC-1 cells expressing EGFP-ACTN4 showed that ACTN4 was most abundant in the leading edges of lamellipodia, although it was also present in stress fibers and focal adhesions. ACTN4 localization in lamellipodia was markedly diminished by phosphoinositide 3-kinase inhibition, whereas its localization in stress fibers and focal adhesions remained. Furthermore, overexpression of ACTN4, but not ACTN1, promoted lamellipodial formation. Live-cell analysis demonstrated that ACTN4-enriched lamellipodia are highly dynamic and associated with cell migration. CLEM revealed that ACTN4-enriched lamellipodia exhibit a characteristic morphology of multilayered ruffle-edges that differs from canonical flat lamellipodia. Similar ruffle-edge lamellipodia were observed in A549 and MDA-MB-231 invasive cancer cells. ACTN4 knockdown suppressed the formation of ruffle-edge lamellipodia and cell migration during wound healing in A549 monolayer cultures. Additionally, membrane-type 1 matrix metalloproteinase was observed in the membrane ruffles, suggesting that ruffle-edge lamellipodia have the ability to degrade the extracellular matrix and may contribute to active cell migration/invasion in certain cancer cell types.
Collapse
Affiliation(s)
- Haruka Morishita
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Katsuhisa Kawai
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Youhei Egami
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan
| | - Kazufumi Honda
- Department of Bioregulation, Graduate of Medicine, Nippon Medical School, Sendagi, Bunkyo-ku, 113-8602, Tokyo, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Kagawa, 761-0793, Miki, Japan.
| |
Collapse
|
10
|
Hsu CY, Li JY, Yang EY, Liao TL, Wen HW, Tsai PC, Ju TC, Lye LF, Nielsen BL, Liu HJ. The Oncolytic Avian Reovirus p17 Protein Inhibits Invadopodia Formation in Murine Melanoma Cancer Cells by Suppressing the FAK/Src Pathway and the Formation of theTKs5/NCK1 Complex. Viruses 2024; 16:1153. [PMID: 39066315 PMCID: PMC11281681 DOI: 10.3390/v16071153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/05/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
To explore whether the p17 protein of oncolytic avian reovirus (ARV) mediates cell migration and invadopodia formation, we applied several molecular biological approaches for studying the involved cellular factors and signal pathways. We found that ARV p17 activates the p53/phosphatase and tensin homolog (PTEN) pathway to suppress the focal adhesion kinase (FAK)/Src signaling and downstream signal molecules, thus inhibiting cell migration and the formation of invadopodia in murine melanoma cancer cell line (B16-F10). Importantly, p17-induced formation of invadopodia could be reversed in cells transfected with the mutant PTENC124A. p17 protein was found to significantly reduce the expression levels of tyrosine kinase substrate 5 (TKs5), Rab40b, non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), and matrix metalloproteinases (MMP9), suggesting that TKs5 and Rab40b were transcriptionally downregulated by p17. Furthermore, we found that p17 suppresses the formation of the TKs5/NCK1 complex. Coexpression of TKs5 and Rab40b in B16-F10 cancer cells reversed p17-modulated suppression of the formation of invadopodia. This work provides new insights into p17-modulated suppression of invadopodia formation by activating the p53/PTEN pathway, suppressing the FAK/Src pathway, and inhibiting the formation of the TKs5/NCK1 complex.
Collapse
Affiliation(s)
- Chao-Yu Hsu
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan;
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (T.-L.L.); (P.-C.T.)
| | - Jyun-Yi Li
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan (T.-C.J.)
| | - En-Ying Yang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan (T.-C.J.)
| | - Tsai-Ling Liao
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (T.-L.L.); (P.-C.T.)
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Hsiao-Wei Wen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan;
| | - Pei-Chien Tsai
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (T.-L.L.); (P.-C.T.)
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Tz-Chuen Ju
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan (T.-C.J.)
| | - Lon-Fye Lye
- Department of Medical Research, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan;
| | - Brent L. Nielsen
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA;
| | - Hung-Jen Liu
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan; (T.-L.L.); (P.-C.T.)
- Institute of Molecular Biology, National Chung Hsing University, Taichung 402, Taiwan (T.-C.J.)
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
11
|
Zhu T, Alves SM, Adamo A, Wen X, Corn KC, Shostak A, Johnson S, Shaub ND, Martello SE, Hacker BC, D'Amore A, Bardhan R, Rafat M. Mammary tissue-derived extracellular matrix hydrogels reveal the role of irradiation in driving a pro-tumor and immunosuppressive microenvironment. Biomaterials 2024; 308:122531. [PMID: 38531198 PMCID: PMC11065579 DOI: 10.1016/j.biomaterials.2024.122531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Radiation therapy (RT) is essential for triple negative breast cancer (TNBC) treatment. However, patients with TNBC continue to experience recurrence after RT. The role of the extracellular matrix (ECM) of irradiated breast tissue in tumor recurrence is still unknown. In this study, we evaluated the structure, molecular composition, and mechanical properties of irradiated murine mammary fat pads (MFPs) and developed ECM hydrogels from decellularized tissues (dECM) to assess the effects of RT-induced ECM changes on breast cancer cell behavior. Irradiated MFPs were characterized by increased ECM deposition and fiber density compared to unirradiated controls, which may provide a platform for cell invasion and proliferation. ECM component changes in collagens I, IV, and VI, and fibronectin were observed following irradiation in both MFPs and dECM hydrogels. Encapsulated TNBC cell proliferation and invasive capacity was enhanced in irradiated dECM hydrogels. In addition, TNBC cells co-cultured with macrophages in irradiated dECM hydrogels induced M2 macrophage polarization and exhibited further increases in proliferation. Our study establishes that the ECM in radiation-damaged sites promotes TNBC invasion and proliferation as well as an immunosuppressive microenvironment. This work represents an important step toward elucidating how changes in the ECM after RT contribute to breast cancer recurrence.
Collapse
Affiliation(s)
- Tian Zhu
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Steven M Alves
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Arianna Adamo
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kevin C Corn
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Anastasia Shostak
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Nicholas D Shaub
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shannon E Martello
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Benjamin C Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Antonio D'Amore
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
12
|
Gamblin C, Chavrier P. [Formation, organization and function of invadosomes in cell motility and tumor invasion]. Med Sci (Paris) 2024; 40:515-524. [PMID: 38986096 DOI: 10.1051/medsci/2024080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Abstract
Invadosome is an umbrella term used to describe a family of cellular structures including podosomes and invadopodia. They serve as contact zones between the cell plasma membrane and extracellular matrix, contributing to matrix remodeling by locally enriched proteolytic enzymes. Invadosomes, which are actin-dependent, are implicated in cellular processes promoting adhesion, migration, and invasion. Invadosomes, which exist in various cell types, play crucial roles in physiological phenomena such as vascularization and bone resorption. Invadosomes are also implicated in pathological processes such as matrix tissue remodeling during metastatic tumor cell invasion. This review summarizes basic information and recent advances about mechanisms underlying podosome and invadopodia formation, their organization and function.
Collapse
Affiliation(s)
- Cécile Gamblin
- Institut Curie, CNRS UMR 144, PSL Research University, Paris, France - Sorbonne Université, Paris, France
| | - Philippe Chavrier
- Institut Curie, CNRS UMR 144, PSL Research University, Paris, France
| |
Collapse
|
13
|
S A, Parida N, Patnaik S. SOX4 induces cytoskeleton remodeling and promotes cell motility via N-wasp/ARP2/3 pathway in colorectal cancer cells. Exp Cell Res 2024; 439:114059. [PMID: 38705228 DOI: 10.1016/j.yexcr.2024.114059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
Filopodia are thin, actin-rich projection from the plasma membrane that promote cancer cell invasion and migration. Sex-determining region Y-related high-mobility group-box 4 (SOX4) is a crucial transcription factor that plays a role in the development and metastasis of colorectal cancer (CRC). However, the involvement of SOX4 in cytoskeleton remodeling in CRC remains unknown. For the first time, we demonstrate that SOX4 is a potent regulator of filopodia formation in CRC cells. Overexpression of SOX4 protein enhances both migration and invasion ability of HCT116, and CACO2 cells, which is relevant to the metastasis. Furthermore, through phalloidin staining, cytoskeleton re-assembly was observed in SOX4-modified cell lines. Enhanced expression of SOX4 increased the number and length of filopodia on cell surface. In contrast, silencing SOX4 in SW620 cells with higher endogenous expression of SOX4, impeded the filopodia formation. Moreover, SOX4 was found to be positively regulating the expression of central regulators of actin cytoskeleton - N-Wiskott-Aldrich syndrome protein (N-WASP); WAVE2; Actin related proteins, ARP2 and ARP3. Inhibiting the N-WASP/ARP2/3 pathway diminishes the filopodia formation and the migration of CRC cells. These results indicate the crucial role of SOX4 in the regulation of filopodia formation mediated by N-WASP/ARP2/3 pathway in CRC cells.
Collapse
Affiliation(s)
- Anupriya S
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| | - Nandita Parida
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| | - Srinivas Patnaik
- School of Biotechnology, KIIT University, Campus-XI, Bhubaneswar, 751024, India.
| |
Collapse
|
14
|
Baro L, Almhassneh RA, Islam A, Juanes MA. Tumor invasiveness is regulated by the concerted function of APC, formins, and Arp2/3 complex. iScience 2024; 27:109687. [PMID: 38680662 PMCID: PMC11053316 DOI: 10.1016/j.isci.2024.109687] [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: 10/19/2023] [Revised: 02/23/2024] [Accepted: 04/05/2024] [Indexed: 05/01/2024] Open
Abstract
Tumor cell invasion is the initial step in metastasis, the leading cause of death from cancer. Invasion requires protrusive cellular structures that steer the migration of leader cells emanating from the tumor mass toward neighboring tissues. Actin is central to these processes and is therefore the prime target of drugs known as migrastatics. However, the broad effects of general actin inhibitors limit their therapeutic use. Here, we delineate the roles of specific actin nucleators in tuning actin-rich invasive protrusions and pinpoint potential pharmacological targets. We subject colorectal cancer spheroids embedded in collagen matrix-a preclinical model mirroring solid tumor invasiveness-to pharmacologic and/or genetic treatment of specific actin arrays to assess their roles in invasiveness. Our data reveal coordinated yet distinct involvement of actin networks nucleated by adenomatous polyposis coli, formins, and actin-related protein 2/3 complex in the biogenesis and maintenance of invasive protrusions. These findings may open avenues for better targeted therapies.
Collapse
Affiliation(s)
- Lautaro Baro
- Cytoskeletal Dynamics in Cell Migration and Cancer Invasion Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Rabeah A. Almhassneh
- Cytoskeletal Dynamics in Cell Migration and Cancer Invasion Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Asifa Islam
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - M. Angeles Juanes
- Cytoskeletal Dynamics in Cell Migration and Cancer Invasion Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| |
Collapse
|
15
|
Arnold L, Yap M, Jackson L, Barry M, Ly T, Morrison A, Gomez JP, Washburn MP, Standing D, Yellapu NK, Li L, Umar S, Anant S, Thomas SM. DCLK1-Mediated Regulation of Invadopodia Dynamics and Matrix Metalloproteinase Trafficking Drives Invasive Progression in Head and Neck Squamous Cell Carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588339. [PMID: 38645056 PMCID: PMC11030349 DOI: 10.1101/2024.04.06.588339] [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: 04/23/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a major health concern due to its high mortality from poor treatment responses and locoregional tumor invasion into life sustaining structures in the head and neck. A deeper comprehension of HNSCC invasion mechanisms holds the potential to inform targeted therapies that may enhance patient survival. We previously reported that doublecortin like kinase 1 (DCLK1) regulates invasion of HNSCC cells. Here, we tested the hypothesis that DCLK1 regulates proteins within invadopodia to facilitate HNSCC invasion. Invadopodia are specialized subcellular protrusions secreting matrix metalloproteinases that degrade the extracellular matrix (ECM). Through a comprehensive proteome analysis comparing DCLK1 control and shDCLK1 conditions, our findings reveal that DCLK1 plays a pivotal role in regulating proteins that orchestrate cytoskeletal and ECM remodeling, contributing to cell invasion. Further, we demonstrate in TCGA datasets that DCLK1 levels correlate with increasing histological grade and lymph node metastasis. We identified higher expression of DCLK1 in the leading edge of HNSCC tissue. Knockdown of DCLK1 in HNSCC reduced the number of invadopodia, cell adhesion and colony formation. Using super resolution microscopy, we demonstrate localization of DCLK1 in invadopodia and colocalization with mature invadopodia markers TKS4, TKS5, cortactin and MT1-MMP. We carried out phosphoproteomics and validated using immunofluorescence and proximity ligation assays, the interaction between DCLK1 and motor protein KIF16B. Pharmacological inhibition or knockdown of DCLK1 reduced interaction with KIF16B, secretion of MMPs, and cell invasion. This research unveils a novel function of DCLK1 within invadopodia to regulate the trafficking of matrix degrading cargo. The work highlights the impact of targeting DCLK1 to inhibit locoregional invasion, a life-threatening attribute of HNSCC.
Collapse
Affiliation(s)
- Levi Arnold
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Marion Yap
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Laura Jackson
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Michael Barry
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Thuc Ly
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Austin Morrison
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Juan P. Gomez
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Michael P. Washburn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Nanda Kumar Yellapu
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Linheng Li
- Stowers Institute, Kansas City, Kansas, USA
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Sufi Mary Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
16
|
Martínez‐López A, García‐Casas A, Infante G, González‐Fernández M, Salvador N, Lorente M, Mendiburu‐Eliçabe M, Gonzalez‐Moreno S, Villarejo‐Campos P, Velasco G, Malliri A, Castillo‐Lluva S. POTEE promotes breast cancer cell malignancy by inducing invadopodia formation through the activation of SUMOylated Rac1. Mol Oncol 2024; 18:620-640. [PMID: 38098337 PMCID: PMC10920093 DOI: 10.1002/1878-0261.13568] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/23/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023] Open
Abstract
The small GTPase Rac1 (Ras-related C3 botulinum toxin substrate 1) has been implicated in cancer progression and in the poor prognosis of various types of tumors. Rac1 SUMOylation occurs during epithelial-mesenchymal transition (EMT), and it is required for tumor cell migration and invasion. Here we identify POTEE (POTE Ankyrin domain family member E) as a novel Rac1-SUMO1 effector involved in breast cancer malignancy that controls invadopodium formation through the activation of Rac1-SUMO1. POTEE activates Rac1 in the invadopodium by recruiting TRIO-GEF (triple functional domain protein), and it induces tumor cell proliferation and metastasis in vitro and in vivo. We found that the co-localization of POTEE with Rac1 is correlated with more aggressive breast cancer subtypes. Given its role in tumor dissemination, the leading cause of cancer-related deaths, POTEE could represent a potential therapeutic target for these types of cancer.
Collapse
Affiliation(s)
- Angélica Martínez‐López
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Ana García‐Casas
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Guiomar Infante
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Mónica González‐Fernández
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Nélida Salvador
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Mar Lorente
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Marina Mendiburu‐Eliçabe
- Departamento de Estadística e Investigación Operativa, Facultad de Ciencias MatemáticasUniversidad Complutense de MadridSpain
| | | | | | - Guillermo Velasco
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Angeliki Malliri
- Cancer Research UK Manchester InstituteThe University of ManchesterUK
| | - Sonia Castillo‐Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias QuímicasUniversidad Complutense de MadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| |
Collapse
|
17
|
Hashimoto M, Miki T, Niwa T, Mihara H. Proximity labeling and identification of endogenous client proteins recruited to Y15-based artificial granules tethering a bait protein. J Pept Sci 2024; 30:e3536. [PMID: 37580979 DOI: 10.1002/psc.3536] [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: 04/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/16/2023]
Abstract
Protein clustering is a ubiquitous event in diverse cellular processes. Self-association of proteins triggers recruitment of downstream proteins to regulate cellular signaling. To investigate the interactions in detail, chemical biology tools to identify proteins recruited to defined assemblies are required. Here, we exploit an identification of proteins recruited in artificial granules (IPRAG) platform that combines intracellular Y15-based supramolecule construction with a proximity labeling method. We validated the IPRAG tool using Nck1 as a target bait protein. We constructed Nck1-tethering granules, labeled the recruited proteins with biotin, and analyzed them by LC-MS/MS. As a result, we successfully identified proteins that directly or indirectly interact with Nck1.
Collapse
Affiliation(s)
- Masahiro Hashimoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Takayuki Miki
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Department of Chemistry and Biotechnology, School of Engineering, University of Tokyo, Tokyo, Japan
| | - Tatsuya Niwa
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Hisakazu Mihara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
18
|
Zhang X, Zhao Y, Li M, Wang M, Qian J, Wang Z, Wang Y, Wang F, Guo K, Gao D, Zhao Y, Chen R, Ren Z, Song H, Cui J. A synergistic regulation works in matrix stiffness-driven invadopodia formation in HCC. Cancer Lett 2024; 582:216597. [PMID: 38145655 DOI: 10.1016/j.canlet.2023.216597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/23/2023] [Accepted: 12/08/2023] [Indexed: 12/27/2023]
Abstract
Growing evidence has suggested that increased matrix stiffness can significantly strengthen the malignant characteristics of hepatocellular carcinoma (HCC) cells. However, whether and how increased matrix stiffness regulates the formation of invadopodia in HCC cells remain largely unknown. In the study, we developed different experimental systems in vitro and in vivo to explore the effects of matrix stiffness on the formation of invadopodia and its relevant molecular mechanism. Our results demonstrated that increased matrix stiffness remarkably augmented the migration and invasion abilities of HCC cells, upregulated the expressions of invadopodia-associated genes and enhanced the number of invadopodia. Two regulatory pathways contribute to matrix stiffness-driven invadopodia formation together in HCC cells, including direct triggering invadopodia formation through activating integrin β1 or Piezo1/ FAK/Src/Arg/cortactin pathway, and indirect stimulating invadopodia formation through improving EGF production to activate EGFR/Src/Arg/cortactin pathway. Src was identified as the common hub molecule of two synergistic regulatory pathways. Simultaneously, activation of integrin β1/RhoA/ROCK1/MLC2 and Piezo1/Ca2+/MLCK/MLC2 pathways mediate matrix stiffness-reinforced cell migration. This study uncovers a new mechanism by which mechanosensory pathway and biochemical signal pathway synergistically regulate the formation of invadopodia in HCC cells.
Collapse
Affiliation(s)
- Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Mimi Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Jiali Qian
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China
| | - Yaohui Wang
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, 200032, PR China
| | - Fan Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China
| | - Haiyan Song
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China.
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, 180 Feng Lin Road, Shanghai, 200032, PR China.
| |
Collapse
|
19
|
Monteiro P, Remy D, Lemerle E, Routet F, Macé AS, Guedj C, Ladoux B, Vassilopoulos S, Lamaze C, Chavrier P. A mechanosensitive caveolae-invadosome interplay drives matrix remodelling for cancer cell invasion. Nat Cell Biol 2023; 25:1787-1803. [PMID: 37903910 PMCID: PMC10709148 DOI: 10.1038/s41556-023-01272-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 09/22/2023] [Indexed: 11/01/2023]
Abstract
Invadosomes and caveolae are mechanosensitive structures that are implicated in metastasis. Here, we describe a unique juxtaposition of caveola clusters and matrix degradative invadosomes at contact sites between the plasma membrane of cancer cells and constricting fibrils both in 2D and 3D type I collagen matrix environments. Preferential association between caveolae and straight segments of the fibrils, and between invadosomes and bent segments of the fibrils, was observed along with matrix remodelling. Caveola recruitment precedes and is required for invadosome formation and activity. Reciprocally, invadosome disruption results in the accumulation of fibril-associated caveolae. Moreover, caveolae and the collagen receptor β1 integrin co-localize at contact sites with the fibrils, and integrins control caveola recruitment to fibrils. In turn, caveolae mediate the clearance of β1 integrin and collagen uptake in an invadosome-dependent and collagen-cleavage-dependent mechanism. Our data reveal a reciprocal interplay between caveolae and invadosomes that coordinates adhesion to and proteolytic remodelling of confining fibrils to support tumour cell dissemination.
Collapse
Affiliation(s)
- Pedro Monteiro
- Actin and Membrane Dynamics Laboratory, Institut Curie-Research Center, CNRS UMR144, PSL Research University, Paris, France.
- Membrane Mechanics and Dynamics of Intracellular Signalling Laboratory, Institut Curie-Research Center, CNRS UMR3666, INSERM U1143, PSL Research University, Paris, France.
| | - David Remy
- Actin and Membrane Dynamics Laboratory, Institut Curie-Research Center, CNRS UMR144, PSL Research University, Paris, France
| | - Eline Lemerle
- Institute of Myology, Sorbonne Université, INSERM UMRS 974, Paris, France
| | - Fiona Routet
- Actin and Membrane Dynamics Laboratory, Institut Curie-Research Center, CNRS UMR144, PSL Research University, Paris, France
| | - Anne-Sophie Macé
- Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, PSL Research University, Paris, France
| | - Chloé Guedj
- Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, PSL Research University, Paris, France
| | - Benoit Ladoux
- Institut Jacques Monod, Université de Paris, CNRS UMR 7592, Paris, France
| | | | - Christophe Lamaze
- Membrane Mechanics and Dynamics of Intracellular Signalling Laboratory, Institut Curie-Research Center, CNRS UMR3666, INSERM U1143, PSL Research University, Paris, France.
| | - Philippe Chavrier
- Actin and Membrane Dynamics Laboratory, Institut Curie-Research Center, CNRS UMR144, PSL Research University, Paris, France.
| |
Collapse
|
20
|
Saito K, Ozawa S, Chiba Y, Takahashi R, Ogomori R, Mukai K, Taguchi T, Hatakeyama H, Ohta Y. FilGAP, a GAP for Rac1, down-regulates invadopodia formation in breast cancer cells. Cell Struct Funct 2023; 48:161-174. [PMID: 37482421 PMCID: PMC11496788 DOI: 10.1247/csf.23032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023] Open
Abstract
Invadopodia are protrusive structures that mediate the extracellular matrix (ECM) degradation required for tumor invasion and metastasis. Rho small GTPases regulate invadopodia formation, but the molecular mechanisms of how Rho small GTPase activities are regulated at the invadopodia remain unclear. Here we have identified FilGAP, a GTPase-activating protein (GAP) for Rac1, as a negative regulator of invadopodia formation in tumor cells. Depletion of FilGAP in breast cancer cells increased ECM degradation and conversely, overexpression of FilGAP decreased it. FilGAP depletion promoted the formation of invadopodia with ECM degradation. In addition, FilGAP depletion and Rac1 overexpression increased the emergence of invadopodia induced by epidermal growth factor, whereas FilGAP overexpression suppressed it. Overexpression of GAP-deficient FilGAP mutant enhanced invadopodia emergence as well as FilGAP depletion. The pleckstrin-homology (PH) domain of FilGAP binds phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2], which is distributed on membranes of the invadopodia. FilGAP localized to invadopodia in breast cancer cells on the ECM, but FilGAP mutant lacking PI(3,4)P2-binding showed low localization. Similarly, the decrease of PI(3,4)P2 production reduced the FilGAP localization. Our results suggest that FilGAP localizes to invadopodia through its PH domain binding to PI(3,4)P2 and down-regulates invadopodia formation by inactivating Rac1, inhibiting ECM degradation in invasive tumor cells.Key words: invadopodia, breast carcinoma, Rac1, FilGAP, PI(3,4)P2.
Collapse
Affiliation(s)
- Koji Saito
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | - Sakino Ozawa
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | - Yosuke Chiba
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | - Ruri Takahashi
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | - Ryoya Ogomori
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | - Kojiro Mukai
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Tomohiko Taguchi
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Hiroyasu Hatakeyama
- Department of Physiology, School of Medicine, Kitasato University, Kanagawa, Japan
| | - Yasutaka Ohta
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| |
Collapse
|
21
|
Li D, Wang X, Miao H, Liu H, Pang M, Guo H, Ge M, Glass SE, Emmrich S, Ji S, Zhou Y, Ye X, Mao H, Wang J, Liu Q, Kim T, Klusmann JH, Li C, Liu Z, Jin H, Nie Y, Wu K, Fan D, Song X, Wang X, Li L, Lu Y, Zhao X. The lncRNA MIR99AHG directs alternative splicing of SMARCA1 by PTBP1 to enable invadopodia formation in colorectal cancer cells. Sci Signal 2023; 16:eadh4210. [PMID: 37725664 DOI: 10.1126/scisignal.adh4210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
Alternative splicing regulates gene expression and functional diversity and is often dysregulated in human cancers. Here, we discovered that the long noncoding RNA (lncRNA) MIR99AHG regulated alternative splicing to alter the activity of a chromatin remodeler and promote metastatic behaviors in colorectal cancer (CRC). MIR99AHG was abundant in invasive CRC cells and metastatic tumors from patients and promoted motility and invasion in cultured CRC cells. MIR99AHG bound to and stabilized the RNA splicing factor PTBP1, and this complex increased cassette exon inclusion in the mRNA encoding the chromatin remodeling gene SMARCA1. Specifically, MIR99AHG altered the nature of PTBP1 binding to the splice sites on intron 12 of SMARCA1 pre-mRNA, thereby triggering a splicing switch from skipping to including exon 13 to produce the long isoform, SMARCA1-L. SMARCA1, but not SMARCA1-L, suppressed invadopodia formation, cell migration, and invasion. Analysis of CRC samples revealed that the abundance of MIR99AHG transcript positively correlated with that of SMARCA1-L mRNA and PTBP1 protein and with poor prognosis in patients with CRC. Furthermore, TGF-β1 secretion from cancer-associated fibroblasts increased MIR99AHG expression in CRC cells. Our findings identify an lncRNA that is induced by cues from the tumor microenvironment and that interacts with PTBP1 to regulate alternative splicing, potentially providing a therapeutic target and predictive biomarker for metastatic CRC.
Collapse
Affiliation(s)
- Danxiu Li
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hui Miao
- Center for Functional Genomics and Bioinformatics, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hao Liu
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Maogui Pang
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hao Guo
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd., Nanjing, Jiangsu 210042, China
| | - Minghui Ge
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co. Ltd., Nanjing, Jiangsu 210042, China
| | - Sarah E Glass
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Stephan Emmrich
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Songtao Ji
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yun Zhou
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiaoni Ye
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Huajie Mao
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing Wang
- Department of Biomedical Informatics and Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Qi Liu
- Department of Biomedical Informatics and Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Taewan Kim
- Department of Anatomy, Histology & Developmental Biology, Base for International Science and Technology Cooperation, Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Jan-Henning Klusmann
- Pediatric Hematology and Oncology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt (Main) 60590, Germany
| | - Cunxi Li
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing 100191, China
| | - Zhenxiong Liu
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Haifeng Jin
- Center for Functional Genomics and Bioinformatics, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yongzhan Nie
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Kaichun Wu
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Daiming Fan
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xu Song
- Center for Functional Genomics and Bioinformatics, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ling Li
- Center for Functional Genomics and Bioinformatics, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yuanyuan Lu
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiaodi Zhao
- Department of Gastroenterology, Tangdu Hospital, Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
22
|
Neumann AJ, Prekeris R. A Rab-bit hole: Rab40 GTPases as new regulators of the actin cytoskeleton and cell migration. Front Cell Dev Biol 2023; 11:1268922. [PMID: 37736498 PMCID: PMC10509765 DOI: 10.3389/fcell.2023.1268922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/23/2023] Open
Abstract
The regulation of machinery involved in cell migration is vital to the maintenance of proper organism function. When migration is dysregulated, a variety of phenotypes ranging from developmental disorders to cancer metastasis can occur. One of the primary structures involved in cell migration is the actin cytoskeleton. Actin assembly and disassembly form a variety of dynamic structures which provide the pushing and contractile forces necessary for cells to properly migrate. As such, actin dynamics are tightly regulated. Classically, the Rho family of GTPases are considered the major regulators of the actin cytoskeleton during cell migration. Together, this family establishes polarity in the migrating cell by stimulating the formation of various actin structures in specific cellular locations. However, while the Rho GTPases are acknowledged as the core machinery regulating actin dynamics and cell migration, a variety of other proteins have become established as modulators of actin structures and cell migration. One such group of proteins is the Rab40 family of GTPases, an evolutionarily and functionally unique family of Rabs. Rab40 originated as a single protein in the bilaterians and, through multiple duplication events, expanded to a four-protein family in higher primates. Furthermore, unlike other members of the Rab family, Rab40 proteins contain a C-terminally located suppressor of cytokine signaling (SOCS) box domain. Through the SOCS box, Rab40 proteins interact with Cullin5 to form an E3 ubiquitin ligase complex. As a member of this complex, Rab40 ubiquitinates its effectors, controlling their degradation, localization, and activation. Because substrates of the Rab40/Cullin5 complex can play a role in regulating actin structures and cell migration, the Rab40 family of proteins has recently emerged as unique modulators of cell migration machinery.
Collapse
Affiliation(s)
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
23
|
Okletey J, Angelis D, Jones TM, Montagna C, Spiliotis ET. An oncogenic isoform of septin 9 promotes the formation of juxtanuclear invadopodia by reducing nuclear deformability. Cell Rep 2023; 42:112893. [PMID: 37516960 PMCID: PMC10530659 DOI: 10.1016/j.celrep.2023.112893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/17/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Invadopodia are extracellular matrix (ECM) degrading structures, which promote cancer cell invasion. The nucleus is increasingly viewed as a mechanosensory organelle that determines migratory strategies. However, how the nucleus crosstalks with invadopodia is little known. Here, we report that the oncogenic septin 9 isoform 1 (SEPT9_i1) is a component of breast cancer invadopodia. SEPT9_i1 depletion diminishes invadopodium formation and the clustering of the invadopodium precursor components TKS5 and cortactin. This phenotype is characterized by deformed nuclei and nuclear envelopes with folds and grooves. We show that SEPT9_i1 localizes to the nuclear envelope and juxtanuclear invadopodia. Moreover, exogenous lamin A rescues nuclear morphology and juxtanuclear TKS5 clusters. Importantly, SEPT9_i1 is required for the amplification of juxtanuclear invadopodia, which is induced by the epidermal growth factor. We posit that nuclei of low deformability favor the formation of juxtanuclear invadopodia in a SEPT9_i1-dependent manner, which functions as a tunable mechanism for overcoming ECM impenetrability.
Collapse
Affiliation(s)
- Joshua Okletey
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Dimitrios Angelis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Tia M Jones
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Cristina Montagna
- Department of Radiology and Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Whitehead CA, Fang H, Su H, Morokoff AP, Kaye AH, Hanssen E, Nowell CJ, Drummond KJ, Greening DW, Vella LJ, Mantamadiotis T, Stylli SS. Small extracellular vesicles promote invadopodia activity in glioblastoma cells in a therapy-dependent manner. Cell Oncol (Dordr) 2023; 46:909-931. [PMID: 37014551 PMCID: PMC10356899 DOI: 10.1007/s13402-023-00786-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
PURPOSE The therapeutic efficacy of radiotherapy/temozolomide treatment for glioblastoma (GBM) is limited by the augmented invasiveness mediated by invadopodia activity of surviving GBM cells. As yet, however the underlying mechanisms remain poorly understood. Due to their ability to transport oncogenic material between cells, small extracellular vesicles (sEVs) have emerged as key mediators of tumour progression. We hypothesize that the sustained growth and invasion of cancer cells depends on bidirectional sEV-mediated cell-cell communication. METHODS Invadopodia assays and zymography gels were used to examine the invadopodia activity capacity of GBM cells. Differential ultracentrifugation was utilized to isolate sEVs from conditioned medium and proteomic analyses were conducted on both GBM cell lines and their sEVs to determine the cargo present within the sEVs. In addition, the impact of radiotherapy and temozolomide treatment of GBM cells was studied. RESULTS We found that GBM cells form active invadopodia and secrete sEVs containing the matrix metalloproteinase MMP-2. Subsequent proteomic studies revealed the presence of an invadopodia-related protein sEV cargo and that sEVs from highly invadopodia active GBM cells (LN229) increase invadopodia activity in sEV recipient GBM cells. We also found that GBM cells displayed increases in invadopodia activity and sEV secretion post radiation/temozolomide treatment. Together, these data reveal a relationship between invadopodia and sEV composition/secretion/uptake in promoting the invasiveness of GBM cells. CONCLUSIONS Our data indicate that sEVs secreted by GBM cells can facilitate tumour invasion by promoting invadopodia activity in recipient cells, which may be enhanced by treatment with radio-chemotherapy. The transfer of pro-invasive cargos may yield important insights into the functional capacity of sEVs in invadopodia.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Huaqi Su
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew P Morokoff
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - Andrew H Kaye
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Eric Hanssen
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Advanced Microscopy Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, 3052, Australia
| | - Katharine J Drummond
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Laura J Vella
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia.
| |
Collapse
|
25
|
Okletey J, Angelis D, Jones TM, Montagna C, Spiliotis ET. An oncogenic isoform of septin 9 promotes the formation of juxtanuclear invadopodia by reducing nuclear deformability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.18.545473. [PMID: 37398172 PMCID: PMC10312791 DOI: 10.1101/2023.06.18.545473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Invadopodia are extracellular matrix (ECM) degrading structures, which promote cancer cell invasion. The nucleus is increasingly viewed as a mechanosensory organelle that determines migratory strategies. However, how the nucleus crosstalks with invadopodia is little known. Here, we report that the oncogenic septin 9 isoform 1 (SEPT9_i1) is a component of breast cancer invadopodia. SEPT9_i1 depletion diminishes invadopodia formation and the clustering of invadopodia precursor components TKS5 and cortactin. This phenotype is characterized by deformed nuclei, and nuclear envelopes with folds and grooves. We show that SEPT9_i1 localizes to the nuclear envelope and juxtanuclear invadopodia. Moreover, exogenous lamin A rescues nuclear morphology and juxtanuclear TKS5 clusters. Importantly, SEPT9_i1 is required for the amplification of juxtanuclear invadopodia, which is induced by the epidermal growth factor. We posit that nuclei of low deformability favor the formation of juxtanuclear invadopodia in a SEPT9_i1-dependent manner, which functions as a tunable mechanism for overcoming ECM impenetrability. Highlights The oncogenic SEPT9_i1 is enriched in breast cancer invadopodia in 2D and 3D ECMSEPT9_i1 promotes invadopodia precursor clustering and invadopodia elongationSEPT9_i1 localizes to the nuclear envelope and reduces nuclear deformabilitySEPT9_i1 is required for EGF-induced amplification of juxtanuclear invadopodia. eTOC Blurb Invadopodia promote the invasion of metastatic cancers. The nucleus is a mechanosensory organelle that determines migratory strategies, but how it crosstalks with invadopodia is unknown. Okletey et al show that the oncogenic isoform SEPT9_i1 promotes nuclear envelope stability and the formation of invadopodia at juxtanuclear areas of the plasma membrane.
Collapse
|
26
|
Chinnathambi S, Das R. Microglia degrade Tau oligomers deposit via purinergic P2Y12-associated podosome and filopodia formation and induce chemotaxis. Cell Biosci 2023; 13:95. [PMID: 37221563 DOI: 10.1186/s13578-023-01028-0] [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: 10/20/2022] [Accepted: 04/02/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Tau protein forms neurofibrillary tangles and becomes deposited in the brain during Alzheimer's disease (AD). Tau oligomers are the most reactive species, mediating neurotoxic and inflammatory activity. Microglia are the immune cells in the central nervous system, sense the extracellular Tau via various cell surface receptors. Purinergic P2Y12 receptor can directly interact with Tau oligomers and mediates microglial chemotaxis via actin remodeling. The disease-associated microglia are associated with impaired migration and express a reduced level of P2Y12, but elevate the level of reactive oxygen species and pro-inflammatory cytokines. RESULTS Here, we studied the formation and organization of various actin microstructures such as-podosome, filopodia and uropod in colocalization with actin nucleator protein Arp2 and scaffold protein TKS5 in Tau-induced microglia by fluorescence microscopy. Further, the relevance of P2Y12 signaling either by activation or blockage was studied in terms of actin structure formations and Tau deposits degradation by N9 microglia. Extracellular Tau oligomers facilitate the microglial migration via Arp2-associated podosome and filopodia formation through the involvement of P2Y12 signaling. Similarly, Tau oligomers induce the TKS5-associated podosome clustering in microglial lamella in a time-dependent manner. Moreover, the P2Y12 was evidenced to localize with F-actin-rich podosome and filopodia during Tau-deposit degradation. The blockage of P2Y12 signaling resulted in decreased microglial migration and Tau-deposit degradation. CONCLUSIONS The P2Y12 signaling mediate the formation of migratory actin structures like- podosome and filopodia to exhibit chemotaxis and degrade Tau deposit. These beneficial roles of P2Y12 in microglial chemotaxis, actin network remodeling and Tau clearance can be intervened as a therapeutic target in AD.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Hosur Road, Bangalore, 560029, Karnataka, India.
| | - Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
27
|
Joshi VB, Gutierrez Ruiz OL, Razidlo GL. The Cell Biology of Metastatic Invasion in Pancreatic Cancer: Updates and Mechanistic Insights. Cancers (Basel) 2023; 15:cancers15072169. [PMID: 37046830 PMCID: PMC10093482 DOI: 10.3390/cancers15072169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality worldwide. This is largely due to the lack of routine screening protocols, an absence of symptoms in early-stage disease leading to late detection, and a paucity of effective treatment options. Critically, the majority of patients either present with metastatic disease or rapidly develop metastatic disease. Thus, there is an urgent need to deepen our understanding of metastasis in PDAC. During metastasis, tumor cells escape from the primary tumor, enter the circulation, and travel to a distant site to form a secondary tumor. In order to accomplish this relatively rare event, tumor cells develop an enhanced ability to detach from the primary tumor, migrate into the surrounding matrix, and invade across the basement membrane. In addition, cancer cells interact with the various cell types and matrix proteins that comprise the tumor microenvironment, with some of these factors working to promote metastasis and others working to suppress it. In PDAC, many of these processes are not well understood. The purpose of this review is to highlight recent advances in the cell biology of the early steps of the metastatic cascade in pancreatic cancer. Specifically, we will examine the regulation of epithelial-to-mesenchymal transition (EMT) in PDAC and its requirement for metastasis, summarize our understanding of how PDAC cells invade and degrade the surrounding matrix, and discuss how migration and adhesion dynamics are regulated in PDAC to optimize cancer cell motility. In addition, the role of the tumor microenvironment in PDAC will also be discussed for each of these invasive processes.
Collapse
Affiliation(s)
- Vidhu B Joshi
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Omar L Gutierrez Ruiz
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Gina L Razidlo
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
28
|
Linder S, Cervero P, Eddy R, Condeelis J. Mechanisms and roles of podosomes and invadopodia. Nat Rev Mol Cell Biol 2023; 24:86-106. [PMID: 36104625 DOI: 10.1038/s41580-022-00530-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 01/28/2023]
Abstract
Cell invasion into the surrounding extracellular matrix or across tissue boundaries and endothelial barriers occurs in both physiological and pathological scenarios such as immune surveillance or cancer metastasis. Podosomes and invadopodia, collectively called 'invadosomes', are actin-based structures that drive the proteolytic invasion of cells, by forming highly regulated platforms for the localized release of lytic enzymes that degrade the matrix. Recent advances in high-resolution microscopy techniques, in vivo imaging and high-throughput analyses have led to considerable progress in understanding mechanisms of invadosomes, revealing the intricate inner architecture of these structures, as well as their growing repertoire of functions that extends well beyond matrix degradation. In this Review, we discuss the known functions, architecture and regulatory mechanisms of podosomes and invadopodia. In particular, we describe the molecular mechanisms of localized actin turnover and microtubule-based cargo delivery, with a special focus on matrix-lytic enzymes that enable proteolytic invasion. Finally, we point out topics that should become important in the invadosome field in the future.
Collapse
Affiliation(s)
- Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany.
| | - Pasquale Cervero
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Robert Eddy
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - John Condeelis
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Remy D, Macé AS, Chavrier P, Monteiro P. Invadopodia Methods: Detection of Invadopodia Formation and Activity in Cancer Cells Using Reconstituted 2D and 3D Collagen-Based Matrices. Methods Mol Biol 2023; 2608:225-246. [PMID: 36653711 DOI: 10.1007/978-1-0716-2887-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tumor dissemination involves cancer cell migration through the extracellular matrix (ECM). ECM is mainly composed of collagen fibers that oppose cell invasion. To overcome hindrance in the matrix, cancer cells deploy a protease-dependent program in order to remodel the matrix fibers. Matrix remodeling requires the formation of actin-based matrix/plasma membrane contact sites called invadopodia, responsible for collagen cleavage through the accumulation and activity of the transmembrane type-I matrix metalloproteinase (MT1-MMP). In this article, we describe experimental procedures designed to assay for invadopodia formation and for invadopodia activity using 2D and 3D models based on gelatin (denatured collagen) and fibrillar type-I collagen matrices.
Collapse
Affiliation(s)
- David Remy
- Institut Curie, CNRS UMR144, PSL Research University, Research Center, Actin and Membrane Dynamics Laboratory, Paris, France
| | - Anne-Sophie Macé
- Institut Curie, PSL Research University, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Philippe Chavrier
- Institut Curie, CNRS UMR144, PSL Research University, Research Center, Actin and Membrane Dynamics Laboratory, Paris, France
| | - Pedro Monteiro
- Institut Curie, CNRS UMR144, PSL Research University, Research Center, Actin and Membrane Dynamics Laboratory, Paris, France.
| |
Collapse
|
30
|
Pan MH, Xu R, Zhang Y, Yin L, Li R, Wen D, Lu S, Gao Y, Zhao X, Wei Q, Han B, Ma B. The Impact of Arp2/3 Complex Inhibition on Cytoskeleton Dynamics and Mitochondrial Function during Goat Oocyte Meiosis. Animals (Basel) 2023; 13:ani13020263. [PMID: 36670803 PMCID: PMC9854427 DOI: 10.3390/ani13020263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
F-actin is of critical importance in oocyte meiotic maturation. Actin assembly and its dynamics are mainly regulated by actin nucleation factors. The actin-related protein complex 2/3 (Arp2/3) is responsible for the organization of F-actin filaments. However, the role of Arp2/3 complex in goat oocytes has not been fully elucidated. Our findings demonstrate that Arp2/3 complex activity is necessary for the maturation of goat oocytes. The Arp2/3 complex-specific inhibitor CK666 impairs the maturation of goat oocytes and alters the genes associated with cumulus expansion, both of which suggest that normal meiosis is affected. Arp2, one of the subunits of the Arp2/3 complex, was found to be mainly accumulated at the oocyte cortex and to co-localize with F-actin during goat oocyte maturation in our results. Thus, we further investigated the cytoskeleton dynamics and found that Arp2/3 complex inhibition disrupts the F-actin assembly and spindle organization. Further analysis revealed that, in addition to direct effects on the cytoskeleton, Arp2/3 complex could also induce ROS accumulation and oxidative stress by disrupting mitochondrial distribution and function, ultimately increasing the rate of early apoptosis in goat oocytes. Our study provides evidence that the Arp2/3 complex is a key regulator of goat oocyte maturation through its regulation of the cytoskeleton dynamics and mitochondrial function.
Collapse
Affiliation(s)
- Meng-Hao Pan
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Rui Xu
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Yiqian Zhang
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Lu Yin
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Ruoyu Li
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Dongxu Wen
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Sihai Lu
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Yan Gao
- Yulin Animal Husbandry and Veterinary Service Center, Yulin 719000, China
| | - Xiaoe Zhao
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Qiang Wei
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
| | - Bin Han
- Yulin Animal Husbandry and Veterinary Service Center, Yulin 719000, China
- Correspondence: (B.H.); (B.M.)
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University/Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling 712100, China
- Correspondence: (B.H.); (B.M.)
| |
Collapse
|
31
|
Cao M, Peng B, Chen H, Yang M, Chen P, Ye L, Wang H, Ren L, Xie J, Zhu J, Xu X, Xu W, Geng L, Gong S. miR-34a induces neutrophil apoptosis by regulating Cdc42-WASP-Arp2/3 pathway-mediated F-actin remodeling and ROS production. Redox Rep 2022; 27:167-175. [PMID: 35938579 PMCID: PMC9364709 DOI: 10.1080/13510002.2022.2102843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background The number of neutrophils is significantly reduced in myelodysplastic syndrome (MDS), but the molecular basis remains unclear. We recently found that miR-34a was significantly increased in MDS neutrophils. Therefore, this study aims to clarify the effects of aberrant miR-34a expression on neutrophil counts. Methods miR-34a mimics/inhibitor transfection were performed in neutrophil-like differentiated HL60 (dHL60) cells, and a FACSCalibur flow cytometer was used to measure ROS production and apoptosis. In addition, the Cdc42-WASP-Arp2/3 pathway inhibitor (ML141) and activator (CN02) treated the dHL60 cells, and then ROS production, apoptosis and related proteins expression were detected. And, luciferase reporter assay to verify the relationship of miR-34a and the Cdc42-WASP-Arp2/3 pathway. Results overexpression of miR-34a could induce ROS production and apoptosis, decrease the expression levels of DOCK8, p-WASP, WASP, Arp2, Arp3, and increase F-actin’s expression. Meanwhile, knockdown of miR-34a could decrease ROS production and apoptosis, increase the expression of DOCK8, p-WASP, WASP, Arp2, Arp3, and decrease F-actin’s expression. Immunofluorescence staining showed aberrant miR-34a and Cdc42-WASP-Arp2/3 pathway could induce F-actin membrane transfer. Luciferase reporter assay indicated that DOCK8 was a direct target gene of miR-34a. Conclusion These data indicates miR-34a may induce neutrophil apoptosis by regulating Cdc42-WASP-Arp2/3 pathway-mediated F-actin remodeling and ROS production.
Collapse
Affiliation(s)
- Meiwan Cao
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Baoling Peng
- Center for child health and mental health, Shenzhen Childen’s Hospital, Shenzhen, People’s Republic of China
| | - Huan Chen
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Min Yang
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Peiyu Chen
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Liping Ye
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Hongli Wang
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Lu Ren
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jing Xie
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jingnan Zhu
- Department of Hematology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Xiangye Xu
- Department of Hematology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
32
|
Tahtamouni L, Alzghoul A, Alderfer S, Sun J, Ahram M, Prasad A, Bamburg J. The role of activated androgen receptor in cofilin phospho-regulation depends on the molecular subtype of TNBC cell line and actin assembly dynamics. PLoS One 2022; 17:e0279746. [PMID: 36584207 PMCID: PMC9803305 DOI: 10.1371/journal.pone.0279746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC) is highly metastatic and of poor prognosis. Metastasis involves coordinated actin filament dynamics mediated by cofilin and associated proteins. Activated androgen receptor (AR) is believed to contribute to TNBC tumorigenesis. Our current work studied roles of activated AR and cofilin phospho-regulation during migration of three AR+ TNBC cell lines to determine if altered cofilin regulation can explain their migratory differences. Untreated or AR agonist-treated BT549, MDA-MB-453, and SUM159PT cells were compared to cells silenced for cofilin (KD) or AR expression/function (bicalutamide). Cofilin-1 was found to be the only ADF/cofilin isoform expressed in each TNBC line. Despite a significant increase in cofilin kinase caused by androgens, the ratio of cofilin:p-cofilin (1:1) did not change in SUM159PT cells. BT549 and MDA-MB-453 cells contain high p-cofilin levels which underwent androgen-induced dephosphorylation through increased cofilin phosphatase expression, but surprisingly maintain a leading-edge with high p-cofilin/total cofilin not found in SUM159PT cells. Androgens enhanced cell polarization in all lines, stimulated wound healing and transwell migration rates and increased N/E-cadherin mRNA ratios while reducing cell adhesion in BT549 and MDA-MB-453 cells. Cofilin KD negated androgen effects in MDA-MB-453 except for cell adhesion, while in BT549 cells it abrogated androgen-reduced cell adhesion. In SUM159PT cells, cofilin KD with and without androgens had similar effects in almost all processes studied. AR dependency of the processes were confirmed. In conclusion, cofilin regulation downstream of active AR is dependent on which actin-mediated process is being examined in addition to being cell line-specific. Although MDA-MB-453 cells demonstrated some control of cofilin through an AR-dependent mechanism, other AR-dependent pathways need to be further studied. Non-cofilin-dependent mechanisms that modulate migration of SUM159PT cells need to be investigated. Categorizing TNBC behavior as AR responsive and/or cofilin dependent can inform on decisions for therapeutic treatment.
Collapse
Affiliation(s)
- Lubna Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States of America
- * E-mail: ,
| | - Ahmad Alzghoul
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Sydney Alderfer
- Department of Chemical and Biological Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States of America
| | - Jiangyu Sun
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States of America
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Ashok Prasad
- Department of Chemical and Biological Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States of America
| | - James Bamburg
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO, United States of America
| |
Collapse
|
33
|
Kreider-Letterman G, Castillo A, Mahlandt EK, Goedhart J, Rabino A, Goicoechea S, Garcia-Mata R. ARHGAP17 regulates the spatiotemporal activity of Cdc42 at invadopodia. J Cell Biol 2022; 222:213782. [PMID: 36571786 PMCID: PMC9794838 DOI: 10.1083/jcb.202207020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/18/2022] [Accepted: 11/28/2022] [Indexed: 12/27/2022] Open
Abstract
Invadopodia formation is regulated by Rho GTPases. However, the molecular mechanisms that control Rho GTPase signaling at invadopodia remain poorly understood. Here, we have identified ARHGAP17, a Cdc42-specific RhoGAP, as a key regulator of invadopodia in breast cancer cells and characterized a novel ARHGAP17-mediated signaling pathway that controls the spatiotemporal activity of Cdc42 during invadopodia turnover. Our results show that during invadopodia assembly, ARHGAP17 localizes to the invadopodia ring and restricts the activity of Cdc42 to the invadopodia core, where it promotes invadopodia growth. Invadopodia disassembly starts when ARHGAP17 translocates from the invadopodia ring to the core, in a process that is mediated by its interaction with the Cdc42 effector CIP4. Once at the core, ARHGAP17 inactivates Cdc42 to promote invadopodia disassembly. Our results in invadopodia provide new insights into the coordinated transition between the activation and inactivation of Rho GTPases.
Collapse
Affiliation(s)
| | - Abel Castillo
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Eike K. Mahlandt
- https://ror.org/04dkp9463Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Joachim Goedhart
- https://ror.org/04dkp9463Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Agustin Rabino
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Silvia Goicoechea
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Rafael Garcia-Mata
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA,Correspondence to Rafael Garcia-Mata:
| |
Collapse
|
34
|
Sun X, Zhou L, Wang X, Li Y, Liu X, Chen Y, Zhong Z, Chen J. FYCO1 regulates migration, invasion, and invadopodia formation in HeLa cells through CDC42/N-WASP/Arp2/3 signaling pathway. Biochem Cell Biol 2022; 100:458-472. [PMID: 36342046 DOI: 10.1139/bcb-2021-0575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
FYCO1, an autophagy adaptor, plays an essential role in the trafficking toward the plus-end of microtubules and the fusion of autophagosomes. Autophagic dysfunction is involved in numerous disease states, including cancers. Previous studies have implicated FYCO1 as one of the critical genes involved in the adenoma to carcinoma transition, but the biological function and mechanism of FYCO1 in carcinogenesis remain unclear. This study aims to elucidate the role and mechanism of up- and downregulation of FYCO1 in mediating tumor effects in HeLa cells. Functionally, FYCO1 promotes cellular migration, invasion, epithelial-mesenchymal transition, invadopodia formation, and matrix degradation, which are detected through wound healing, transwell, immunofluorescence, and Western blot approaches. Interestingly, the data show that although FYCO1 does not affect HeLa cell proliferation, cell cycle distribution, nor vessels' formation, FYCO1 can block the apoptotic function. FYCO1 inhibits cleavage of PARP, caspase3, and caspase9 and increases Bcl-2/Bax ratio. Then, we used CK666, an Arp2/3 specific inhibitor, to confirm that FYCO1 may promote the migration and invasion of HeLa cells through the CDC42/N-WASP/Arp2/3 signaling pathway. Taken together, these results provide a new insight that FYCO1, an autophagy adaptor, may also be a new regulator of tumor metastasis.
Collapse
Affiliation(s)
- Xuejiao Sun
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Linlin Zhou
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xinyao Wang
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yuying Li
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Xiangyuan Liu
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Yu Chen
- Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Zilin Zhong
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Jianjun Chen
- Translational Research Institute of Brain and Brain-like Intelligence, People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.,Department of Medical Genetics, School of Medicine, Tongji University, Shanghai 200092, China.,Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| |
Collapse
|
35
|
Lv S, Chen Z, Mi H, Yu X. Cofilin Acts as a Booster for Progression of Malignant Tumors Represented by Glioma. Cancer Manag Res 2022; 14:3245-3269. [PMID: 36452435 PMCID: PMC9703913 DOI: 10.2147/cmar.s389825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/10/2022] [Indexed: 07/20/2023] Open
Abstract
Cofilin, as a depolymerization factor of actin filaments, has been widely studied. Evidences show that cofilin has a role in actin structural reorganization and dynamic regulation. In recent years, several studies have demonstrated a regulatory role for cofilin in the migration and invasion mediated by cell dynamics and epithelial to mesenchymal transition (EMT)/EMT-like process, apoptosis, radiotherapy resistance, immune escape, and transcriptional dysregulation of malignant tumor cells, particularly glioma cells. On this basis, it is practical to evaluate cofilin as a biomarker for predicting tumor metastasis and prognosis. Targeting cofilin regulating kinases, Lin11, Isl-1 and Mec-3 kinases (LIM kinases/LIMKs) and their major upstream molecules inhibits tumor cell migration and invasion and targeting cofilin-mediated mitochondrial pathway induces apoptosis of tumor cells represent effective options for the development of novel anti-malignant tumor drug, especially anti-glioma drugs. This review explores the structure, general biological function, and regulation of cofilin, with an emphasis on the critical functions and prospects for clinical therapeutic applications of cofilin in malignant tumors represented by glioma.
Collapse
Affiliation(s)
- Shihong Lv
- Department of Gastroenterology, The Second Affiliated Hospital of Mudanjiang Medical College, Mudanjiang Medical College, Mudanjiang, 157011, People’s Republic of China
| | - Zhiye Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Hailong Mi
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xingjiang Yu
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
36
|
Feng D, Zhu W, Shi X, Wei W, Han P, Wei Q, Yang L. Leucine zipper protein 2 serves as a prognostic biomarker for prostate cancer correlating with immune infiltration and epigenetic regulation. Heliyon 2022; 8:e10750. [PMID: 36217461 PMCID: PMC9547219 DOI: 10.1016/j.heliyon.2022.e10750] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/19/2022] [Accepted: 09/20/2022] [Indexed: 11/04/2022] Open
Abstract
Background We sought to determine whether leucine zipper protein 2 (LUZP2) could benefit men with prostate cancer (PCa) undergoing radical radiotherapy (RT) or prostatectomy (RP). Methods Analysis was done on differentiating expression, clinical prognosis, co-expressed genes, immune infiltration, and epigenetic changes. All of our analyses were done using the R software (version 3.6.3) and the appropriate packages. Results In terms of PCa, tumor samples expressed LUZP2 more than normal samples did. In the TCGA database and GSE116918, we found that LUZP2 was the only independent risk factor for PCa. The shared enriched pathways for patients undergoing RP or RT were cell-cell adhesion, regulation of filopodium assembly, and extracellular matrix containing collagen. With the exception of TNFRSF14, we discovered that LUZP2 was negatively correlated with 21 immune checkpoints in PCa patients receiving RT. We found a significant inverse relationship between LUZP2 expression and the tumor immune environment, which included B cells, CD4+ T cells, neutrophils, macrophages, dendritic cells, stromal score, immune score, and estimate score, in patients receiving RP or RT. Additionally, tumor purity was positively correlated with LUZP2. We found that the drug bortezomib may be susceptible to the LUZP2. DNA methylation was significantly associated with the mRNA expression of LUZP2 in PCa patients from the TCGA database, and LUZP2 methylation was positively correlated with immune cells. The proliferative activity of various PCa cells, which correlated to different stages of this disease, was also found to be significantly reduced by LUZP2 reduction, according to the results of our experimental work. Conclusions We proposed a relatively comprehensive understanding of the roles of LUZP2 on PCa from the fresh perspective of senescence.
Collapse
|
37
|
Xue Y, Li M, Hu J, Song Y, Guo W, Miao C, Ge D, Hou Y, Wang X, Huang X, Liu T, Zhang X, Huang Q. Ca v2.2-NFAT2-USP43 axis promotes invadopodia formation and breast cancer metastasis through cortactin stabilization. Cell Death Dis 2022; 13:812. [PMID: 36137995 PMCID: PMC9500045 DOI: 10.1038/s41419-022-05174-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 01/23/2023]
Abstract
Distant metastasis is the main cause of mortality in breast cancer patients. Using the breast cancer genomic data from The Cancer Genome Atlas (TCGA), we identified brain specific Cav2.2 as a critical regulator of metastasis. Cav2.2 expression is significantly upregulated in breast cancer and its higher expression is inversely correlated with survival suggesting a previously unappreciated role of Cav2.2 in breast cancer. Cav2.2 is required for breast cancer migration, invasion, and metastasis. Interestingly, Cav2.2 promotes invadopodia formation and extracellular matrix (ECM) degradation through the stabilization of invadopodia component cortactin in a proteosome-dependent manner. Moreover, deubiquitinating enzyme USP43 mediated the functions of Cav2.2 in cortactin stabilization, invadopodia formation, ECM degradation, and metastasis. Interestingly, Cav2.2 upregulates USP43 expression through NFAT2 dephosphorylation and nuclear localization. Our study uncovered a novel pathway that regulates cortactin expression and invadopodia formation in breast cancer metastasis.
Collapse
Affiliation(s)
- Ying Xue
- grid.8547.e0000 0001 0125 2443Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, PR China ,grid.8547.e0000 0001 0125 2443Institute of Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Min Li
- grid.8547.e0000 0001 0125 2443Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, PR China ,grid.8547.e0000 0001 0125 2443Institute of Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Jie Hu
- grid.8547.e0000 0001 0125 2443Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yuanlin Song
- grid.8547.e0000 0001 0125 2443Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Wei Guo
- grid.8547.e0000 0001 0125 2443Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Changhong Miao
- grid.8547.e0000 0001 0125 2443Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, PR China ,grid.8547.e0000 0001 0125 2443Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Di Ge
- grid.8547.e0000 0001 0125 2443Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yingyong Hou
- grid.8547.e0000 0001 0125 2443Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Xuefei Wang
- grid.8547.e0000 0001 0125 2443Department of General Surgery/Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Xingxu Huang
- grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, PR China
| | - Tianshu Liu
- grid.8547.e0000 0001 0125 2443Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, PR China ,grid.8547.e0000 0001 0125 2443Department of Medicial Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Xiaoping Zhang
- grid.24516.340000000123704535The Institute of Intervention Vessel, Tongji University School of Medicine, Shanghai, PR China
| | - Qihong Huang
- grid.8547.e0000 0001 0125 2443Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, PR China ,grid.8547.e0000 0001 0125 2443Institute of Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, PR China ,grid.413087.90000 0004 1755 3939Shanghai Respiratory Research Institute, Shanghai, PR China
| |
Collapse
|
38
|
Li L, Wen Z, Kou N, Liu J, Jin D, Wang L, Wang F, Gao L. LIS1 interacts with CLIP170 to promote tumor growth and metastasis via the Cdc42 signaling pathway in salivary gland adenoid cystic carcinoma. Int J Oncol 2022; 61:129. [PMID: 36102310 PMCID: PMC9477107 DOI: 10.3892/ijo.2022.5419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 08/05/2022] [Indexed: 11/20/2022] Open
Abstract
Salivary gland adenoid cystic carcinoma (SACC) is one of the most common malignant tumors, with high aggressive potential in the oral and maxillofacial regions. Lissencephaly 1 (LIS1) is a microtubule-organizing center-associated protein that regulates the polymerization and stability of microtubules by mediating the motor function of dynein. Recent studies have suggested that LIS1 plays a potential role in the malignant development of tumors, such as in mitosis and migration. However, the role of LIS1 in SACC development and its related molecular mechanisms remain unclear. Thus, the effects of LIS1 on the proliferation, apoptosis, invasion and metastasis of SACC were studied, in vivo and in vitro. The results of immunohistochemical staining showed that LIS1 was highly expressed in SACC tissues, and its expression level was associated with malignant progression. In vitro, the results of CCK-8, TUNEL, wound healing and Transwell assays demonstrated that LIS1 promotes proliferation, inhibits apoptosis, and enhances the migration and invasion of SACC-LM cells. In vivo, knockdown of LIS1 effectively suppressed the growth of subcutaneous tumors in a mouse xenograft and distant metastasis of tumor cells in the metastasis model. The co-immunoprecipitation, immunofluorescence and western blot results also revealed that LIS1 binds to cytoplasmic linker protein 170 (CLIP170) to form a protein complex (LIS1/CLIP170), which activates the cell division control protein 42 homolog (Cdc42) signaling pathway to modulate the proliferation and anti-apoptosis of tumor cells, and enhanced invasion and metastasis by regulating the formation of invadopodia and the expression of MMPs in SACC-LM cells. Therefore, the present study demonstrated that LIS1 is a cancer promoter in SACC, and the molecular mechanism of the LIS1/CLIP170/Cdc42 signaling pathway is involved in the malignant progression, which offers a promising strategy for targeted therapy of SACC.
Collapse
Affiliation(s)
- Lijun Li
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Zhihao Wen
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ni Kou
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jing Liu
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Dong Jin
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lina Wang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Fu Wang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lu Gao
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
39
|
Allen JL, Hames RA, Mastroianni NM, Greenstein AE, Weed SA. Evaluation of the matrix metalloproteinase 9 (MMP9) inhibitor Andecaliximab as an Anti-invasive therapeutic in Head and neck squamous cell carcinoma. Oral Oncol 2022; 132:106008. [PMID: 35803110 DOI: 10.1016/j.oraloncology.2022.106008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/21/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Locoregional and lymphovascular involvement of invasive head and neck squamous cell carcinoma (HNSCC) complicates curative treatment. Matrix metalloproteinase (MMP) 9 is a negative prognostic marker in HNSCC and targets multiple extracellular matrix (ECM) substrates, where it contributes to breaching basement membrane and stromal barriers enabling invasive spread. Andecaliximab (ADX) is a second-generation MMP9 inhibitor well tolerated in clinical trials of gastric and pancreatic adenocarcinoma. The impact of selective MMP9 targeting by ADX in HNSCC has not been evaluated. MATERIALS AND METHODS Established and patient-derived xenograft (PDX) cell lines were utilized in HNSCC invasion assays to determine the inhibitory ability of MMP9-mediated invasion by ADX. MMP9 expression was confirmed using immunohistochemistry (IHC) and immunoblotting. ECM degradation was evaluated with confocal microscopy. Cell invasion from tumor spheroids was monitored by phase microscopy. Histological evaluation was used to determine ADX efficacy in three-dimensional organotypic cultures containing cancer associated fibroblasts (CAFs). RESULTS MMP9 was expressed in all established and PDX-derived cell lines. While the broad spectrum clinical MMP inhibitor marimastat (BB2516) blocked HNSCC invadopodia function and tumor spheroid invasion, ADX treatment failed to inhibit invadopodia-based matrix degradation, tumor cell or fibroblast-driven ECM invasion in collagen I-based matrices. CONCLUSION ADX monotherapy was ineffective at blocking initial MMP-dependent events of HNSCC invasion, likely due to redundant functions of additional non-targeted MMPs produced by tumor cells and microenvironment. Combination of ADX with existing and emerging therapies targeting additional MMP activation pathways may warrant future investigation.
Collapse
Affiliation(s)
- Jessica L Allen
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, United States
| | - River A Hames
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, United States
| | - Natalie M Mastroianni
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, United States
| | | | - Scott A Weed
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, United States.
| |
Collapse
|
40
|
The dual action of glioma-derived exosomes on neuronal activity: synchronization and disruption of synchrony. Cell Death Dis 2022; 13:705. [PMID: 35963860 PMCID: PMC9376103 DOI: 10.1038/s41419-022-05144-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/28/2022] [Accepted: 07/28/2022] [Indexed: 01/21/2023]
Abstract
Seizures represent a frequent symptom in gliomas and significantly impact patient morbidity and quality of life. Although the pathogenesis of tumor-related seizures is not fully understood, accumulating evidence indicates a key role of the peritumoral microenvironment. Brain cancer cells interact with neurons by forming synapses with them and by releasing exosomes, cytokines, and other small molecules. Strong interactions among neurons often lead to the synchronization of their activity. In this paper, we used an in vitro model to investigate the role of exosomes released by glioma cell lines and by patient-derived glioma stem cells (GSCs). The addition of exosomes released by U87 glioma cells to neuronal cultures at day in vitro (DIV) 4, when neurons are not yet synchronous, induces synchronization. At DIV 7-12 neurons become highly synchronous, and the addition of the same exosomes disrupts synchrony. By combining Ca2+ imaging, electrical recordings from single neurons with patch-clamp electrodes, substrate-integrated microelectrode arrays, and immunohistochemistry, we show that synchronization and de-synchronization are caused by the combined effect of (i) the formation of new neuronal branches, associated with a higher expression of Arp3, (ii) the modification of synaptic efficiency, and (iii) a direct action of exosomes on the electrical properties of neurons, more evident at DIV 7-12 when the threshold for spike initiation is significantly reduced. At DIV 7-12 exosomes also selectively boost glutamatergic signaling by increasing the number of excitatory synapses. Remarkably, de-synchronization was also observed with exosomes released by glioma-associated stem cells (GASCs) from patients with low-grade glioma but not from patients with high-grade glioma, where a more variable outcome was observed. These results show that exosomes released from glioma modify the electrical properties of neuronal networks and that de-synchronization caused by exosomes from low-grade glioma can contribute to the neurological pathologies of patients with brain cancers.
Collapse
|
41
|
Liu J, Zhu X, Gao L, Geng R, Tao X, Xu H, Chen Z. Expression and Prognostic Role of Glia Maturation Factor-γ in Gliomas. Front Mol Neurosci 2022; 15:906762. [PMID: 35845613 PMCID: PMC9277395 DOI: 10.3389/fnmol.2022.906762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background Glia maturation factor-γ (GMFG) regulates actin cytoskeletal organization and promotes the invasion of cancer cells. However, its expression pattern and molecular function in gliomas have not been clearly defined. Methods In this study, public datasets comprising 2,518 gliomas samples were used to explore GMFG expression and its correlation with malignancy in gliomas. Immunohistochemistry (IHC) staining was performed to determine the expression of GMFG in gliomas using an in-house cohort that contained 120 gliomas samples. Gene ontology enrichment analysis was conducted using the DAVID tool. The correlation between GMFG expression and immune cell infiltration was evaluated using TIMER, Tumor Immune Single-Cell Hub (TISCH) database, and IHC staining assays. The Kaplan-Meier analysis was performed to determine the prognostic role of GMFG and its association with temozolomide (TMZ) response in gliomas. Results The GMFG expression was higher in gliomas compared with non-tumor brain tissues both in public datasets and in-house cohort. High expression of GMFG was significantly associated with WHO grade IV, IDH 1/2 wild-type, and mesenchymal (ME) subtypes. Bioinformatic prediction and IHC analysis revealed that GMFG expression obviously correlated with the macrophage marker CD163 in gliomas. Moreover, both lower grade glioma (LGG) and glioblastoma multiforme (GBM) patients with high GMFG expression had shorter overall survival than those with low GMFG expression. These results indicate that GMFG may be a therapeutic target for the treatment of such patients. Patients with low GMFG expression who received chemotherapy had a longer survival time than those with high GMFG expression. For patients who received ion radiotherapy (IR) only, the GMFG expression level had no effect on the overall survival neither in CGGA and TCGA datasets. Conclusion The GMFG is a novel prognostic biomarker for patients with both LGG and GBM. Increased GMFG expression is associated with tumor-associated macrophages (TAMs) infiltration and with a bad response to TMZ treatment.
Collapse
Affiliation(s)
- Junhui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaonan Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rongxin Geng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiang Tao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haitao Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
42
|
Li TD, Bieling P, Weichsel J, Mullins RD, Fletcher DA. The molecular mechanism of load adaptation by branched actin networks. eLife 2022; 11:e73145. [PMID: 35748355 PMCID: PMC9328761 DOI: 10.7554/elife.73145] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Branched actin networks are self-assembling molecular motors that move biological membranes and drive many important cellular processes, including phagocytosis, endocytosis, and pseudopod protrusion. When confronted with opposing forces, the growth rate of these networks slows and their density increases, but the stoichiometry of key components does not change. The molecular mechanisms governing this force response are not well understood, so we used single-molecule imaging and AFM cantilever deflection to measure how applied forces affect each step in branched actin network assembly. Although load forces are observed to increase the density of growing filaments, we find that they actually decrease the rate of filament nucleation due to inhibitory interactions between actin filament ends and nucleation promoting factors. The force-induced increase in network density turns out to result from an exponential drop in the rate constant that governs filament capping. The force dependence of filament capping matches that of filament elongation and can be explained by expanding Brownian Ratchet theory to cover both processes. We tested a key prediction of this expanded theory by measuring the force-dependent activity of engineered capping protein variants and found that increasing the size of the capping protein increases its sensitivity to applied forces. In summary, we find that Brownian Ratchets underlie not only the ability of growing actin filaments to generate force but also the ability of branched actin networks to adapt their architecture to changing loads.
Collapse
Affiliation(s)
- Tai-De Li
- Department of Bioengineering & Biophysics Program, University of California, BerkeleyBerkeleyUnited States
- Division of Biological Systems & Engineering, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Advanced Science Research Center, City University of New YorkNew YorkUnited States
| | - Peter Bieling
- Division of Biological Systems & Engineering, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San FranciscoSan FranciscoUnited States
- Department of Systemic Cell Biology, Max Planck Institute of Molecular PhysiologyDortmundGermany
| | - Julian Weichsel
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San FranciscoSan FranciscoUnited States
| | - Daniel A Fletcher
- Department of Bioengineering & Biophysics Program, University of California, BerkeleyBerkeleyUnited States
- Division of Biological Systems & Engineering, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| |
Collapse
|
43
|
Limaye AJ, Whittaker MK, Bendzunas GN, Cowell JK, Kennedy EJ. Targeting the WASF3 complex to suppress metastasis. Pharmacol Res 2022; 182:106302. [PMID: 35691539 DOI: 10.1016/j.phrs.2022.106302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
Wiskott-Aldrich syndrome protein family members (WASF) regulate the dynamics of the actin cytoskeleton, which plays an instrumental role in cancer metastasis and invasion. WASF1/2/3 forms a hetero-pentameric complex with CYFIP1/2, NCKAP1/1 L, Abi1/2/3 and BRK1 called the WASF Regulatory Complex (WRC), which cooperatively regulates actin nucleation by WASF1/2/3. Activation of the WRC enables actin networking and provides the mechanical force required for the formation of lamellipodia and invadopodia. Although the WRC drives cell motility essential for several routine physiological functions, its aberrant deployment is observed in cancer metastasis and invasion. WASF3 expression is correlated with metastatic potential in several cancers and inversely correlates with overall progression-free survival. Therefore, disruption of the WRC may serve as a novel strategy for targeting metastasis. Given the complexity involved in the formation of the WRC which is largely comprised of large protein-protein interfaces, there are currently no inhibitors for WASF3. However, several constrained peptide mimics of the various protein-protein interaction interfaces within the WRC were found to successfully disrupt WASF3-mediated migration and invasion. This review explores the role of the WASF3 WRC in driving metastasis and how it may be selectively targeted for suppression of metastasis.
Collapse
Affiliation(s)
- Ameya J Limaye
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, 240W. Green St, Athens, GA 30602, United States
| | - Matthew K Whittaker
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, 240W. Green St, Athens, GA 30602, United States
| | - George N Bendzunas
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, 240W. Green St, Athens, GA 30602, United States
| | - John K Cowell
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA 30912, United States
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, 240W. Green St, Athens, GA 30602, United States.
| |
Collapse
|
44
|
Mazzolini J, Le Clerc S, Morisse G, Coulonges C, Zagury J, Sieger D. Wasl is crucial to maintain microglial core activities during glioblastoma initiation stages. Glia 2022; 70:1027-1051. [PMID: 35194846 PMCID: PMC9306864 DOI: 10.1002/glia.24154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
Abstract
Microglia actively promotes the growth of high-grade gliomas. Within the glioma microenvironment an amoeboid microglial morphology has been observed, however the underlying causes and the related impact on microglia functions and their tumor promoting activities is unclear. Using the advantages of the larval zebrafish model, we identified the underlying mechanism and show that microglial morphology and functions are already impaired during glioma initiation stages. The presence of pre-neoplastic HRasV12 expressing cells induces an amoeboid morphology of microglia, increases microglial numbers and decreases their motility and phagocytic activity. RNA sequencing analysis revealed lower expression levels of the actin nucleation promoting factor wasla in microglia. Importantly, a microglia specific rescue of wasla expression restores microglial morphology and functions. This results in increased phagocytosis of pre-neoplastic cells and slows down tumor progression. In conclusion, we identified a mechanism that de-activates core microglial functions within the emerging glioma microenvironment. Restoration of this mechanism might provide a way to impair glioma growth.
Collapse
Affiliation(s)
- Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Sigrid Le Clerc
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Gregoire Morisse
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Cédric Coulonges
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Jean‐François Zagury
- Laboratoire GBCM, EA7528, Conservatoire National des Arts et MétiersHESAM UniversitéParisFrance
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
45
|
Kramer DA, Piper HK, Chen B. WASP family proteins: Molecular mechanisms and implications in human disease. Eur J Cell Biol 2022; 101:151244. [PMID: 35667337 PMCID: PMC9357188 DOI: 10.1016/j.ejcb.2022.151244] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023] Open
Abstract
Proteins of the Wiskott-Aldrich syndrome protein (WASP) family play a central role in regulating actin cytoskeletal dynamics in a wide range of cellular processes. Genetic mutations or misregulation of these proteins are tightly associated with many diseases. The WASP-family proteins act by transmitting various upstream signals to their conserved WH2-Central-Acidic (WCA) peptide sequence at the C-terminus, which in turn binds to the Arp2/3 complex to stimulate the formation of branched actin networks at membranes. Despite this common feature, the regulatory mechanisms and cellular functions of distinct WASP-family proteins are very different. Here, we summarize and clarify our current understanding of WASP-family proteins and how disruption of their functions is related to human disease.
Collapse
Affiliation(s)
- Daniel A Kramer
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Hannah K Piper
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Baoyu Chen
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA.
| |
Collapse
|
46
|
Ostrowska-Podhorodecka Z, Ding I, Norouzi M, McCulloch CA. Impact of Vimentin on Regulation of Cell Signaling and Matrix Remodeling. Front Cell Dev Biol 2022; 10:869069. [PMID: 35359446 PMCID: PMC8961691 DOI: 10.3389/fcell.2022.869069] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Vimentin expression contributes to cellular mechanoprotection and is a widely recognized marker of fibroblasts and of epithelial-mesenchymal transition. But it is not understood how vimentin affects signaling that controls cell migration and extracellular matrix (ECM) remodeling. Recent data indicate that vimentin controls collagen deposition and ECM structure by regulating contractile force application to the ECM and through post-transcriptional regulation of ECM related genes. Binding of cells to the ECM promotes the association of vimentin with cytoplasmic domains of adhesion receptors such as integrins. After initial adhesion, cell-generated, myosin-dependent forces and signals that impact vimentin structure can affect cell migration. Post-translational modifications of vimentin determine its adaptor functions, including binding to cell adhesion proteins like paxillin and talin. Accordingly, vimentin regulates the growth, maturation and adhesive strength of integrin-dependent adhesions, which enables cells to tune their attachment to collagen, regulate the formation of cell extensions and control cell migration through connective tissues. Thus, vimentin tunes signaling cascades that regulate cell migration and ECM remodeling. Here we consider how specific properties of vimentin serve to control cell attachment to the underlying ECM and to regulate mesenchymal cell migration and remodeling of the ECM by resident fibroblasts.
Collapse
|
47
|
Kalli M, Li R, Mills GB, Stylianopoulos T, Zervantonakis IK. Mechanical Stress Signaling in Pancreatic Cancer Cells Triggers p38 MAPK- and JNK-Dependent Cytoskeleton Remodeling and Promotes Cell Migration via Rac1/cdc42/Myosin II. Mol Cancer Res 2022; 20:485-497. [PMID: 34782370 PMCID: PMC8898300 DOI: 10.1158/1541-7786.mcr-21-0266] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/24/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022]
Abstract
Advanced or metastatic pancreatic cancer is highly resistant to existing therapies, and new treatments are urgently needed to improve patient outcomes. Current studies focus on alternative treatment approaches that target the abnormal microenvironment of pancreatic tumors and the resulting elevated mechanical stress in the tumor interior. Nevertheless, the underlying mechanisms by which mechanical stress regulates pancreatic cancer metastatic potential remain elusive. Herein, we used a proteomic assay to profile mechanical stress-induced signaling cascades that drive the motility of pancreatic cancer cells. Proteomic analysis, together with selective protein inhibition and siRNA treatments, revealed that mechanical stress enhances cell migration through activation of the p38 MAPK/HSP27 and JNK/c-Jun signaling axes, and activation of the actin cytoskeleton remodelers: Rac1, cdc42, and myosin II. In addition, mechanical stress upregulated transcription factors associated with epithelial-to-mesenchymal transition and stimulated the formation of stress fibers and filopodia. p38 MAPK and JNK inhibition resulted in lower cell proliferation and more effectively blocked cell migration under mechanical stress compared with control conditions. The enhanced tumor cell motility under mechanical stress was potently reduced by cdc42 and Rac1 silencing with no effects on proliferation. Our results highlight the importance of targeting aberrant signaling in cancer cells that have adapted to mechanical stress in the tumor microenvironment, as a novel approach to effectively limit pancreatic cancer cell migration. IMPLICATIONS Our findings highlight that mechanical stress activated the p38 MAPK and JNK signaling axis and stimulated pancreatic cancer cell migration via upregulation of the actin cytoskeleton remodelers cdc42 and Rac1.
Collapse
Affiliation(s)
- Maria Kalli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ruxuan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health Sciences University, Oregon, Pennsylvania
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ioannis K. Zervantonakis
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Brunetti RM, Kockelkoren G, Raghavan P, Bell GR, Britain D, Puri N, Collins SR, Leonetti MD, Stamou D, Weiner OD. WASP integrates substrate topology and cell polarity to guide neutrophil migration. J Cell Biol 2022; 221:e202104046. [PMID: 34964841 PMCID: PMC8719638 DOI: 10.1083/jcb.202104046] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/19/2021] [Accepted: 12/10/2021] [Indexed: 12/02/2022] Open
Abstract
To control their movement, cells need to coordinate actin assembly with the geometric features of their substrate. Here, we uncover a role for the actin regulator WASP in the 3D migration of neutrophils. We show that WASP responds to substrate topology by enriching to sites of inward, substrate-induced membrane deformation. Superresolution imaging reveals that WASP preferentially enriches to the necks of these substrate-induced invaginations, a distribution that could support substrate pinching. WASP facilitates recruitment of the Arp2/3 complex to these sites, stimulating local actin assembly that couples substrate features with the cytoskeleton. Surprisingly, WASP only enriches to membrane deformations in the front half of the cell, within a permissive zone set by WASP's front-biased regulator Cdc42. While WASP KO cells exhibit relatively normal migration on flat substrates, they are defective at topology-directed migration. Our data suggest that WASP integrates substrate topology with cell polarity by selectively polymerizing actin around substrate-induced membrane deformations in the front half of the cell.
Collapse
Affiliation(s)
- Rachel M. Brunetti
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
- Center for Geometrically Engineered Cellular Membranes, University of California, San Francisco, San Francisco, CA
| | - Gabriele Kockelkoren
- Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
- Center for Geometrically Engineered Cellular Membranes, University of Copenhagen, Copenhagen, Denmark
| | - Preethi Raghavan
- University of California, Berkeley–University of California, San Francisco Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| | - George R.R. Bell
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA
| | - Derek Britain
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
- Center for Geometrically Engineered Cellular Membranes, University of California, San Francisco, San Francisco, CA
| | - Natasha Puri
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
| | - Sean R. Collins
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA
| | | | - Dimitrios Stamou
- Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
- Center for Geometrically Engineered Cellular Membranes, University of Copenhagen, Copenhagen, Denmark
| | - Orion D. Weiner
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA
- Center for Geometrically Engineered Cellular Membranes, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
49
|
Decoding Single Cell Morphology in Osteotropic Breast Cancer Cells for Dissecting Their Migratory, Molecular and Biophysical Heterogeneity. Cancers (Basel) 2022; 14:cancers14030603. [PMID: 35158871 PMCID: PMC8833404 DOI: 10.3390/cancers14030603] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is a heterogeneous disease and the mechanistic framework for differential osteotropism among intrinsic breast cancer subtypes is unknown. Hypothesizing that cell morphology could be an integrated readout for the functional state of a cancer cell, we established a catalogue of the migratory, molecular and biophysical traits of MDA-MB-231 breast cancer cells, compared it with two enhanced bone-seeking derivative cell lines and integrated these findings with single cell morphology profiles. Such knowledge could be essential for predicting metastatic capacities in breast cancer. High-resolution microscopy revealed a heterogeneous and specific spectrum of single cell morphologies in bone-seeking cells, which correlated with differential migration and stiffness. While parental MDA-MB-231 cells showed long and dynamic membrane protrusions and were enriched in motile cells with continuous and mesenchymal cell migration, bone-seeking cells appeared with discontinuous mesenchymal or amoeboid-like migration. Although non-responsive to CXCL12, bone-seeking cells responded to epidermal growth factor with a morphotype shift and differential expression of genes controlling cell shape and directional migration. Hence, single cell morphology encodes the molecular, migratory and biophysical architecture of breast cancer cells and is specifically altered among osteotropic phenotypes. Quantitative morpho-profiling could aid in dissecting breast cancer heterogeneity and in refining clinically relevant intrinsic breast cancer subtypes.
Collapse
|
50
|
Zhu Y, Ye L, Huang H, Xu X, Liu Y, Wang J, Jin Y. Case report: Primary immunodeficiency due to a novel mutation in CARMIL2 and its response to combined immunomodulatory therapy. Front Pediatr 2022; 10:1042302. [PMID: 36727012 PMCID: PMC9884805 DOI: 10.3389/fped.2022.1042302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/06/2022] [Indexed: 01/18/2023] Open
Abstract
Capping protein regulator and myosin 1 linker 2 (CARMIL2) is necessary for invadopodia formation, cell polarity, lamellipodial assembly, membrane ruffling, acropinocytosis, and collective cell migration. CARMIL2 deficiency is a rare autosomal recessive disease characterized by dysfunction in naïve T-cell activation, proliferation, differentiation, and effector function and insufficient responses in T-cell memory. In this paper, we report a 9-year-old female patient with a novel pathogenic variant in CARMIL2 (c.2063C > G:p.Thr688Arg) who presented with various symptoms of primary immunodeficiencies including recurrent upper and lower respiratory infections, perioral and perineum papules, reddish impetiginized atopic dermatitis, oral ulcer, painful urination and vaginitis, otitis media, and failure to thrive. A missense mutation leading to insufficient CARMIL2 protein expression, reduced absolute T-cell and natural killer cell (NK cell) counts, and marked skewing to the naïve T-cell form was identified and indicated defective maturation of T cells and B cells. Following 1 year of multitargeted treatment with corticosteroids, hydroxychloroquine, mycophenolate mofetil, and thymosin, the patient presented with significant regression in rashes. CD4+ T-cell, CD8+ T-cell, and NK cell counts were significantly improved.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lili Ye
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hua Huang
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xuemei Xu
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yu Liu
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yanliang Jin
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|