1
|
Cho Y, Hwang JW, Bedford MT, Song DG, Kim SN, Kim YK. CARM1 regulates tubulin autoregulation through PI3KC2α R175 methylation. Cell Commun Signal 2025; 23:120. [PMID: 40045375 PMCID: PMC11884010 DOI: 10.1186/s12964-025-02124-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/23/2025] [Indexed: 03/09/2025] Open
Abstract
Tubulin is crucial in several cellular processes, including intracellular organization, organelle transport, motility, and chromosome segregation. Intracellular tubulin concentration is tightly regulated by an autoregulation mechanism, in which excess free tubulin promotes tubulin mRNA degradation. However, the details of how changes in free tubulin levels initiate this autoregulation remain unclear. In this study, we identified coactivator-associated arginine methyltransferase 1 (CARM1)-phosphatidylinositol 3-kinase class 2α (PI3KC2α) axis as a novel regulator of tubulin autoregulation. CARM1 stabilizes PI3KC2α by methylating its R175 residue. Once PI3KC2α is not methylated, it becomes unstable, leading to decreased cellular levels. Loss of PI3KC2α results in the release of tetratricopeptide repeat domain 5 (TTC5), which initiates tubulin autoregulation. Thus, PI3KC2α, along with its CARM1-mediated arginine methylation, regulates the initiation of tubulin autoregulation. Additionally, disruption of the CARM1-PI3KC2α axis decreases intracellular tubulin levels, leading to a synergistic increase in the cytotoxicity of microtubule-targeting agents (MTAs). Taken together, our study demonstrates that the CARM1-PI3KC2α axis is a key regulator of TTC5-mediated tubulin autoregulation and that disrupting this axis enhances the anti-cancer activity of MTAs.
Collapse
Affiliation(s)
- Yena Cho
- Muscle Physiome Research Center and Research Institute of Pharmaceutical Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jee Won Hwang
- College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dae-Geun Song
- Natural Products Research Institute, KIST Gangneung, Gangneung, 25451, Republic of Korea
- Division of Natural Product Applied Science, University of Science and Technology KIST School, Seoul, 02792, Republic of Korea
| | - Su-Nam Kim
- Natural Products Research Institute, KIST Gangneung, Gangneung, 25451, Republic of Korea
- Division of Natural Product Applied Science, University of Science and Technology KIST School, Seoul, 02792, Republic of Korea
| | - Yong Kee Kim
- Muscle Physiome Research Center and Research Institute of Pharmaceutical Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
- College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
2
|
Fan L, Xiao H, Ren J, Hou Y, Cai J, Wu W, Xiang B, Lin Q, Liao M, Ren T, Chen L. Newcastle disease virus induces clathrin-mediated endocytosis to establish infection through the activation of PI3K/AKT signaling pathway by VEGFR2. J Virol 2024; 98:e0132224. [PMID: 39254313 PMCID: PMC11494881 DOI: 10.1128/jvi.01322-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/24/2024] [Indexed: 09/11/2024] Open
Abstract
The phosphatidyl-inositol 3-kinase/serine-threonine kinase (PI3K/ AKT) signaling pathway constitutes a classical phosphorylation cascade that integrates tyrosine, lipid, and serine acid-threonine phosphorylation, affecting cell function. The pathway is vulnerable to viral infection. Newcastle disease virus (NDV) poses a significant threat to the global poultry industry; however, its mechanism of early viral cell invasion and pathogenesis remain unclear. Previous in vivo and in vitro studies have shown that NDV infection activates PI3K/AKT signaling; however, it remains unclear whether NDV establishes infection through endocytosis regulated by this pathway. This study aimed to examine whether different genotypes of NDV strains could activate the PI3K/AKT signaling pathway within 2 h of in vitro infection. This activation, which relies on PI3K phosphorylation, remains unaffected by the phosphorylation-phosphatase and tensin homolog/phosphatase and tensin homolog (p-PTEN/PTEN) signaling pathway. Moreover, inhibition of PI3K activity impedes NDV replication. Additionally, interfering with the PI3K regulatory subunit p85 has no significant effect on NDV replication. Conversely, the tyrosine kinase activity upstream of PI3K can influence AKT activation and viral replication, particularly through vascular endothelial growth factor receptor 2 (VEGFR2). Additionally, NDV F protein primarily mediates PI3K and AKT phosphorylation to activate the PI3K/AKT signaling pathway. NDV F and VEGFR2 proteins, along with the PI3K p85α subunit, interact and co-localize at the cell membrane. NDV-induced PI3K/AKT signaling pathway activation impacts clathrin-mediated endocytosis, with VEGFR2 playing a pivotal role. In conclusion, this study shows that NDV infection is established early through F protein binding to VEGFR2, activating the PI3K/AKT signaling pathway and inducing clathrin-mediated endocytosis, supporting infection prevention and control measures. IMPORTANCE Newcastle disease virus (NDV) is a threat to the global poultry industry; however, the mechanisms of NDV infection remain unclear. NDV affects the phosphatidyl-inositol 3-kinase/serine-threonine kinase (PI3K/ AKT) signaling pathway, requiring endocytosis for successful infection. Based on previous studies, we identified a close correlation between NDV infection and replication and the PI3K/AKT signaling pathway activity. This study examined the molecular mechanisms through which NDV activates the PI3K/AKT signaling pathway to regulate endocytosis and facilitate infection. This study showed that early-stage in vitro NDV infection activated the PI3K/AKT signaling pathway, enhancing clathrin-mediated endocytosis, crucial for infection onset. Notably, this process involves the interaction between NDV F protein and the vascular endothelial growth factor receptor 2 tyrosine kinase, leading to the subsequent binding and phosphorylation of the PI3K p85α regulatory subunit. This activation primes PI3K, initiating a cascade that promotes clathrin-mediated endocytosis. Our findings elucidate how NDV capitalizes on the PI3K/AKT signaling pathway to establish infection through endocytosis.
Collapse
Affiliation(s)
- Lei Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Hongtao Xiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Jinlian Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Yuechi Hou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Juncheng Cai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Wanyan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Bin Xiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Qiuyan Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Libin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| |
Collapse
|
3
|
Ling S, Kwak D, Takuwa Y, Ge C, Franceschi R, Kim KK. Discoidin domain receptor 2 signaling through PIK3C2α in fibroblasts promotes lung fibrosis. J Pathol 2024; 262:505-516. [PMID: 38332727 PMCID: PMC10940211 DOI: 10.1002/path.6253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/30/2023] [Accepted: 12/12/2023] [Indexed: 02/10/2024]
Abstract
Pulmonary fibrosis, especially idiopathic pulmonary fibrosis (IPF), portends significant morbidity and mortality, and current therapeutic options are suboptimal. We have previously shown that type I collagen signaling through discoidin domain receptor 2 (DDR2), a receptor tyrosine kinase expressed by fibroblasts, is critical for the regulation of fibroblast apoptosis and progressive fibrosis. However, the downstream signaling pathways for DDR2 remain poorly defined and could also be attractive potential targets for therapy. A recent phosphoproteomic approach indicated that PIK3C2α, a poorly studied member of the PI3 kinase family, could be a downstream mediator of DDR2 signaling. We hypothesized that collagen I/DDR2 signaling through PIK3C2α regulates fibroblast activity during progressive fibrosis. To test this hypothesis, we found that primary murine fibroblasts and IPF-derived fibroblasts stimulated with endogenous or exogenous type I collagen led to the formation of a DDR2/PIK3C2α complex, resulting in phosphorylation of PIK3C2α. Fibroblasts treated with an inhibitor of PIK3C2α or with deletion of PIK3C2α had fewer markers of activation after stimulation with TGFβ and more apoptosis after stimulation with a Fas-activating antibody. Finally, mice with fibroblast-specific deletion of PIK3C2α had less fibrosis after bleomycin treatment than did littermate control mice with intact expression of PIK3Cα. Collectively, these data support the notion that collagen/DDR2/PIK3C2α signaling is critical for fibroblast function during progressive fibrosis, making this pathway a potential target for antifibrotic therapy. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Song Ling
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Doyun Kwak
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa Ishikawa, Japan
| | - Chunxi Ge
- Departments of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Renny Franceschi
- Departments of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Kevin K. Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Chen ZY, Mortha A. INPP4B ensures that ILC1s and NK cells set up a productive home office. J Exp Med 2024; 221:e20232375. [PMID: 38329467 PMCID: PMC10866016 DOI: 10.1084/jem.20232375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
In this issue of JEM, Peng et al. (https://doi.org/10.1084/jem.20230124) identify inositol polyphosphate 4-phosphatase type II (encoded by Inpp4b) as an important enzyme for tissue-resident ILC1 and NK cell survival, signal transduction, and anti-tumor immunity.
Collapse
Affiliation(s)
- Zi Yan Chen
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, Canada
| |
Collapse
|
5
|
Peng V, Trsan T, Sudan R, Bhattarai B, Cortez VS, Molgora M, Vacher J, Colonna M. Inositol phosphatase INPP4B sustains ILC1s and intratumoral NK cells through an AKT-driven pathway. J Exp Med 2024; 221:e20230124. [PMID: 38197946 PMCID: PMC10783437 DOI: 10.1084/jem.20230124] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 11/17/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous population of lymphocytes that coordinate early immune responses and maintain tissue homeostasis. Type 1 innate immune responses are mediated by natural killer (NK) cells and group 1 ILCs (ILC1s). Despite their shared features, NK cells and ILC1s display profound differences among various tissue microenvironments. Here, we identify the inositol polyphosphatase INPP4B as a hallmark feature of tissue-resident ILC1s and intratumoral NK cells using an scRNA-seq atlas of tissue-associated and circulating NK/ILC1s. Conditional deletion of Inpp4b in ILC1s and NK cells reveals that it is necessary for the homeostasis of tissue-resident ILC1s but not circulating NK cells at steady-state. Inpp4b-deficient cells display increased rates of apoptosis and reduced activation of the prosurvival molecule AKT. Furthermore, expression of Inpp4b by NK/ILC1s is necessary for their presence in the intratumoral environment, and lack of Inpp4b impairs antitumor immunity. These findings highlight INPP4B as a novel regulator of tissue residency and antitumor function in ILC1s and NK cells.
Collapse
Affiliation(s)
- Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Raki Sudan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bishan Bhattarai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Victor S. Cortez
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jean Vacher
- Institut de Recherches Cliniques de Montréal, Montréal, Canada
- Département de Médecine, Université de Montréal, Montréal, Canada
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Sakata J, Tatsumi T, Sugiyama A, Shimizu A, Inagaki Y, Katoh H, Yamashita T, Takahashi K, Aki S, Kaneko Y, Kawamura T, Miura M, Ishii M, Osawa T, Tanaka T, Ishikawa S, Tsukagoshi M, Chansler M, Kodama T, Kanai M, Tokuyama H, Yamatsugu K. Antibody-mimetic drug conjugate with efficient internalization activity using anti-HER2 VHH and duocarmycin. Protein Expr Purif 2024; 214:106375. [PMID: 37797818 DOI: 10.1016/j.pep.2023.106375] [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: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Antibody-mimetic drug conjugate (AMDC) is a cancer cell-targeted drug delivery system based on the non-covalent binding of mutated streptavidin and modified biotin, namely Cupid and Psyche. However, the development of AMDCs is hampered by difficulties in post-translational modification or poor internalization activity. Here, we report an expression, refolding, and purification method for AMDC using a variable heavy chain of heavy chain-only antibodies (VHHs). Monomeric anti-HER2 VHH fused to Cupid was expressed in Escherichia coli inclusion bodies. Solubilization and refolding at optimized reducing conditions and pH levels were selected to form a functional, tetrameric protein (anti-HER2 VHH-Cupid) that can be easily purified based on molecular weight. Anti-HER2 VHH-Cupid non-covalently creates a tight complex with Psyche linked to a potent DNA-alkylating agent, duocarmycin. This complex can be absorbed by the HER2-expressing human breast cancer cell line, KPL-4, and kills KPL-4 cells in vitro and in vivo. The production of a targeting protein with internalizing activity, combined with the non-covalent conjugation of a highly potent payload, renders AMDC a promising platform for developing cancer-targeted therapy.
Collapse
Affiliation(s)
- Juri Sakata
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Toshifumi Tatsumi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Akira Sugiyama
- Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunyo-ku, Tokyo, 113-0032, Japan; Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan.
| | - Akihiro Shimizu
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Yuya Inagaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takefumi Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan; Department of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Sho Aki
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Yudai Kaneko
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan; Medical & Biological Laboratories Co., Ltd, 2-11-8 Shibadaimon, Minato-ku, Tokyo, 105-0012, Japan
| | - Takeshi Kawamura
- Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunyo-ku, Tokyo, 113-0032, Japan; Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Mai Miura
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Masazumi Ishii
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Tsuyoshi Osawa
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Toshiya Tanaka
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | | | - Michael Chansler
- Savid Therapeutics Inc., Eifuku 3-9-10, Suginami-ku, Tokyo, 168-0064, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hidetoshi Tokuyama
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8675, Japan.
| |
Collapse
|
7
|
Morgan RC, Frank C, Greger M, Attaway M, Sigvardsson M, Bartom ET, Kee BL. TGF-β Promotes the Postselection Thymic Development and Peripheral Function of IFN-γ-Producing Invariant NKT cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1376-1384. [PMID: 37702745 PMCID: PMC10592054 DOI: 10.4049/jimmunol.2200809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 08/29/2023] [Indexed: 09/14/2023]
Abstract
IFN-γ-producing invariant NKT (iNKT)1 cells are lipid-reactive innate-like lymphocytes that are resident in the thymus and peripheral tissues where they protect against pathogenic infection. The thymic functions of iNKT1 cells are not fully elucidated, but subsets of thymic iNKT cells modulate CD8 T cell, dendritic cell, B cell, and thymic epithelial cell numbers or function. In this study, we show that a subset of murine thymic iNKT1 cells required TGF-β-induced signals for their postselection development, to maintain hallmark TGF-β-induced genes, and for expression of the adhesion receptors CD49a and CD103. However, the residency-associated receptor CD69 was not TGF-β signaling-dependent. Recently described CD244+ c2 thymic iNKT1 cells, which produce IFN-γ without exogenous stimulation and have NK-like characteristics, reside in this TGF-β-responsive population. Liver and spleen iNKT1 cells do not share this TGF-β gene signature, but nonetheless TGF-β impacts liver iNKT1 cell phenotype and function. Our findings provide insight into the heterogeneity of mechanisms guiding iNKT1 cell development in different tissues and suggest a close association between a subset of iNKT1 cells and TGF-β-producing cells in the thymus that support their development.
Collapse
Affiliation(s)
- Roxroy C. Morgan
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637
| | - Cameron Frank
- Dept. of Pathology, The University of Chicago, Chicago, IL 60637
| | - Munmun Greger
- Dept. of Pathology, The University of Chicago, Chicago, IL 60637
- Committees on Cancer Biology and Immunology, The University of Chicago, Chicago, IL 60637
| | - Mary Attaway
- Committees on Cancer Biology and Immunology, The University of Chicago, Chicago, IL 60637
| | | | - Elizabeth T. Bartom
- Dept. of Biochemistry and Molecular Genetics, Northwestern Feinberg School of Medicine, Chicago IL
| | - Barbara L. Kee
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637
- Dept. of Pathology, The University of Chicago, Chicago, IL 60637
- Committees on Cancer Biology and Immunology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
8
|
Xu X, Zhou R, Duan Q, Miao Y, Zhang T, Wang M, Jones OD, Xu M. Circulating macrophages as the mechanistic link between mosaic loss of Y-chromosome and cardiac disease. Cell Biosci 2023; 13:135. [PMID: 37488646 PMCID: PMC10364353 DOI: 10.1186/s13578-023-01075-7] [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: 11/23/2022] [Accepted: 06/19/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Genetics evidences have long linked mosaic loss of Y-chromosome (mLOY) in peripheral leukocytes with a wide range of male age-associated diseases. However, a lack of cellular and molecular mechanistic explanations for this link has limited further investigation into the relationship between mLOY and male age-related disease. Excitingly, Sano et al. have provided the first piece of evidence directly linking mLOY to cardiac fibrosis through mLOY enriched profibrotic transforming growth factor β1 (TGF-β1) regulons in hematopoietic macrophages along with suppressed interleukin-1β (IL-1β) proinflammatory regulons. The results of this novel finding can be extrapolated to other disease related to mLOY, such as cancer, cardiac disease, and age-related macular degeneration. RESULTS Sano et al. used a CRISPR-Cas9 gRNAs gene editing induced Y-chromosome ablation mouse model to assess results of a UK biobank prospective analysis implicating the Y-chromosome in male age-related disease. Using this in vivo model, Sano et al. showed that hematopoietic mLOY accelerated cardiac fibrosis and heart failure in male mice through profibrotic pathways. This process was linked to monocyte-macrophage differentiation during hematopoietic development. Mice confirmed to have mLOY in leukocytes, by loss of Y-chromosome genes Kdm5d, Uty, Eif2s3y, and Ddx3y, at similar percentages to the human population were shown to have accelerated rates of interstitial and perivascular fibrosis and abnormal echocardiograms. These mice also recovered poorly from the transverse aortic constriction (TAC) model of heart failure and developed left ventricular dysfunction at higher rates. This was attributed to aberrant proliferation of cardiac MEF-SK4 + fibroblasts promoted by mLOY macrophages enriched in profibrotic regulons and lacking in proinflammatory regulons. These pro-fibrotic macrophages localized to heart and eventually resulted in cardiac fibrosis via enhanced TGF-β1 and suppressed IL-1β signaling. Furthermore, treatment of mLOY mice with TGFβ1 neutralizing antibody was able to improve their cardiac function. This study by Sano et al. was able to provide a causative link between the known association between mLOY and male cardiac disease morbidity and mortality for the first time, and thereby provide a new target for improving human health. CONCLUSIONS Using a CRISPR-Cas9 induced Y-chromosome ablation mouse model, Sano et al. has proven mosaic loss of Y-chromosome in peripheral myeloid cells to have a causative effect on male mobility and mortality due to male age-related cardiac disease. They traced the mechanism of this effect to hyper-expression of the profibrotic TGF-β1 and reduced pro-inflammatory IL-1β signaling, attenuation of which could provide another potential strategy in improving outcomes against age-related diseases in men.
Collapse
Affiliation(s)
- Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences and University Hospital Medical Center, 620 West Chang'an, Chang'an District, Xi'an, 710119, China.
| | - Rong Zhou
- Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences and University Hospital Medical Center, 620 West Chang'an, Chang'an District, Xi'an, 710119, China
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences and University Hospital Medical Center, 620 West Chang'an, Chang'an District, Xi'an, 710119, China
| | - Yuanlin Miao
- Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences and University Hospital Medical Center, 620 West Chang'an, Chang'an District, Xi'an, 710119, China
| | - Tingting Zhang
- Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences and University Hospital Medical Center, 620 West Chang'an, Chang'an District, Xi'an, 710119, China
| | - Mofei Wang
- Laboratory of Cell Biology, Genetics and Developmental Biology, Shaanxi Normal University College of Life Sciences and University Hospital Medical Center, 620 West Chang'an, Chang'an District, Xi'an, 710119, China
| | - Odell D Jones
- University of Pennsylvania School of Medicine ULAR, Philadelphia, PA, 19144, USA
| | - MengMeng Xu
- Department of Pediatrics, Morgan Stanley Children's Hospital, Columbia University, 3959 Broadway, New York, NY, 10032, USA.
| |
Collapse
|
9
|
Aki S, Nakahara R, Maeda K, Osawa T. Cancer metabolism within tumor microenvironments. Biochim Biophys Acta Gen Subj 2023; 1867:130330. [PMID: 36804842 DOI: 10.1016/j.bbagen.2023.130330] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/06/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Tumor microenvironments could determine cancer heterogeneity and malignancy. Hypoxia, nutrition starvation, and acidic pH could contribute to cancer malignancy associated with genetic, epigenetic, and metabolic alterations, promoting invasion and metastasis. Cancer cells adapting to extreme tumor microenvironments could enable evasion of cell death and immune responses. It could stimulate drug resistance and recurrence, resulting in poor patient prognosis. Therefore, investigating druggable targets of the malignant cancer cells within tumor microenvironments is necessary, but such treatments are limited. Cell-cell metabolic interaction may also contribute to cancer malignancy within the tumor microenvironments. Organelle-organelle interactions have recently gained attention as new cancer therapy targets as they play essential roles in the metabolic adaptation to the tumor microenvironment. In this review, we overview (1) metabolic alterations within tumor microenvironments, (2) cell-to-cell, and (3) organelle-to-organelle metabolic interactions, and we add novel insights into cancer therapy.
Collapse
Affiliation(s)
- Sho Aki
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan; Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Ryuichi Nakahara
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan; Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Keisuke Maeda
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Tsuyoshi Osawa
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan; Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan.
| |
Collapse
|
10
|
Nakahara R, Maeda K, Aki S, Osawa T. Metabolic adaptations of cancer in extreme tumor microenvironments. Cancer Sci 2023; 114:1200-1207. [PMID: 36630222 PMCID: PMC10067430 DOI: 10.1111/cas.15722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/26/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
Cancer cells are highly heterogeneous to adapt to extreme tumor microenvironments (TMEs). TMEs challenge cancer cells via hypoxia, nutrition starvation, and acidic pH, promoting invasion and metastasis concomitant with genetic, epigenetic, and metabolic alterations. Metabolic adaptation to an extreme TME could allow cancer cells to evade cell death and immune responses, as well as resulting in drug resistance, recurrence, and poor patient prognosis. Therefore, elucidation of the metabolic adaptation of malignant cancer cells within TMEs is necessary, however, most are still elusive. Recently, adaptation of cancer cells within the TME can be analyzed via cell-cell interactions at the single-cell level. In addition, information into organelle-organelle interactions has recently been obtained. These cell-cell, and organelle-organelle interactions demonstrate the potential as new cancer therapy targets, as they play essential roles in the metabolic adaptation of cancer cells to the TME. In this manuscript, we review (1) metabolic adaptations within tumor microenvironments through (2) cell-to-cell, and (3) organelle-organelle metabolic interactions.
Collapse
Affiliation(s)
- Ryuichi Nakahara
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, Tokyo, Japan.,Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Keisuke Maeda
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, Tokyo, Japan
| | - Sho Aki
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, Tokyo, Japan.,Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tsuyoshi Osawa
- Division of Nutriomics and Oncology, RCAST, The University of Tokyo, Tokyo, Japan.,Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Lo WT, Belabed H, Kücükdisli M, Metag J, Roske Y, Prokofeva P, Ohashi Y, Horatscheck A, Cirillo D, Krauss M, Schmied C, Neuenschwander M, von Kries JP, Médard G, Kuster B, Perisic O, Williams RL, Daumke O, Payrastre B, Severin S, Nazaré M, Haucke V. Development of selective inhibitors of phosphatidylinositol 3-kinase C2α. Nat Chem Biol 2023; 19:18-27. [PMID: 36109648 PMCID: PMC7613998 DOI: 10.1038/s41589-022-01118-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/20/2022] [Indexed: 01/01/2023]
Abstract
Phosphatidylinositol 3-kinase type 2α (PI3KC2α) and related class II PI3K isoforms are of increasing biomedical interest because of their crucial roles in endocytic membrane dynamics, cell division and signaling, angiogenesis, and platelet morphology and function. Herein we report the development and characterization of PhosphatidylInositol Three-kinase Class twO INhibitors (PITCOINs), potent and highly selective small-molecule inhibitors of PI3KC2α catalytic activity. PITCOIN compounds exhibit strong selectivity toward PI3KC2α due to their unique mode of interaction with the ATP-binding site of the enzyme. We demonstrate that acute inhibition of PI3KC2α-mediated synthesis of phosphatidylinositol 3-phosphates by PITCOINs impairs endocytic membrane dynamics and membrane remodeling during platelet-dependent thrombus formation. PITCOINs are potent and selective cell-permeable inhibitors of PI3KC2α function with potential biomedical applications ranging from thrombosis to diabetes and cancer.
Collapse
Affiliation(s)
- Wen-Ting Lo
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Hassane Belabed
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Murat Kücükdisli
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Juliane Metag
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Yvette Roske
- grid.419491.00000 0001 1014 0849Max-Delbrück-Centrum für Molekulare Medizin, Kristallographie, Berlin, Germany
| | - Polina Prokofeva
- grid.6936.a0000000123222966Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Yohei Ohashi
- grid.42475.300000 0004 0605 769XMRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - André Horatscheck
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Davide Cirillo
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Michael Krauss
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Christopher Schmied
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Martin Neuenschwander
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Jens Peter von Kries
- grid.418832.40000 0001 0610 524XLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Guillaume Médard
- grid.6936.a0000000123222966Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Bernhard Kuster
- grid.6936.a0000000123222966Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Olga Perisic
- grid.42475.300000 0004 0605 769XMRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Roger L. Williams
- grid.42475.300000 0004 0605 769XMRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Oliver Daumke
- grid.419491.00000 0001 1014 0849Max-Delbrück-Centrum für Molekulare Medizin, Kristallographie, Berlin, Germany
| | - Bernard Payrastre
- Inserm, U1297-Université, Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France ,grid.411175.70000 0001 1457 2980Centre Hospitalier Universitaire de Toulouse, Laboratoire d’Hématologie, Toulouse, France
| | - Sonia Severin
- Inserm, U1297-Université, Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Marc Nazaré
- Departments of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany.
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany. .,Departments of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Dutta P, Ray K. Ciliary membrane, localised lipid modification and cilia function. J Cell Physiol 2022; 237:2613-2631. [PMID: 35661356 DOI: 10.1002/jcp.30787] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/08/2022]
Abstract
Cilium, a tiny microtubule-based cellular appendage critical for cell signalling and physiology, displays a large variety of receptors. The composition and turnover of ciliary lipids and receptors determine cell behaviour. Due to the exclusion of ribosomal machinery and limited membrane area, a cilium needs adaptive logistics to actively reconstitute the lipid and receptor compositions during development and differentiation. How is this dynamicity generated? Here, we examine whether, along with the Intraflagellar-Transport, targeted changes in sector-wise lipid composition could control the receptor localisation and functions in the cilia. We discuss how an interplay between ciliary lipid composition, localised lipid modification, and receptor function could contribute to cilia growth and signalling. We argue that lipid modification at the cell-cilium interface could generate an added thrust for a selective exchange of membrane lipids and the transmembrane and membrane-associated proteins.
Collapse
Affiliation(s)
- Priya Dutta
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
13
|
Lo WT, Zhang Y, Vadas O, Roske Y, Gulluni F, De Santis MC, Zagar AV, Stephanowitz H, Hirsch E, Liu F, Daumke O, Kudryashev M, Haucke V. Structural basis of phosphatidylinositol 3-kinase C2α function. Nat Struct Mol Biol 2022; 29:218-228. [PMID: 35256802 PMCID: PMC8930771 DOI: 10.1038/s41594-022-00730-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/21/2022] [Indexed: 12/16/2022]
Abstract
Phosphatidylinositol 3-kinase type 2α (PI3KC2α) is an essential member of the structurally unresolved class II PI3K family with crucial functions in lipid signaling, endocytosis, angiogenesis, viral replication, platelet formation and a role in mitosis. The molecular basis of these activities of PI3KC2α is poorly understood. Here, we report high-resolution crystal structures as well as a 4.4-Å cryogenic-electron microscopic (cryo-EM) structure of PI3KC2α in active and inactive conformations. We unravel a coincident mechanism of lipid-induced activation of PI3KC2α at membranes that involves large-scale repositioning of its Ras-binding and lipid-binding distal Phox-homology and C-C2 domains, and can serve as a model for the entire class II PI3K family. Moreover, we describe a PI3KC2α-specific helical bundle domain that underlies its scaffolding function at the mitotic spindle. Our results advance our understanding of PI3K biology and pave the way for the development of specific inhibitors of class II PI3K function with wide applications in biomedicine.
Collapse
Affiliation(s)
- Wen-Ting Lo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.
| | - Yingyi Zhang
- Max Planck Institute for Biophysics, Frankfurt am Main, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt am Main, Germany.,Biological Cryo-EM Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Oscar Vadas
- University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Yvette Roske
- Max Delbrück Centre for Molecular Medicine (MDC), Crystallography, Berlin, Germany
| | - Federico Gulluni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Maria Chiara De Santis
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | | | - Heike Stephanowitz
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Fan Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Oliver Daumke
- Max Delbrück Centre for Molecular Medicine (MDC), Crystallography, Berlin, Germany
| | - Misha Kudryashev
- Max Planck Institute for Biophysics, Frankfurt am Main, Germany.,Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt am Main, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany. .,Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
14
|
Yamatsugu K, Katoh H, Yamashita T, Takahashi K, Aki S, Tatsumi T, Kaneko Y, Kawamura T, Miura M, Ishii M, Ohkubo K, Osawa T, Kodama T, Ishikawa S, Kanai M, Sugiyama A. Antibody mimetic drug conjugate manufactured by high-yield Escherichia coli expression and non-covalent binding system. Protein Expr Purif 2022; 192:106043. [PMID: 34973460 DOI: 10.1016/j.pep.2021.106043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
Antibody-drug conjugates (ADCs) are a major therapeutic tool for the treatment of advanced cancer. Malignant cells in advanced cancer often display multiple genetic mutations and become resistant to monotherapy. Therefore, a therapeutic regimen that simultaneously targets multiple molecules with multiple payloads is desirable. However, the development of ADCs is hampered by issues in biopharmaceutical manufacturing and the complexity of the conjugation process of low-molecular-weight payloads to biologicals. Here, we report antibody mimetic-drug conjugates (AMDCs) developed by exploiting the non-covalent binding property of payloads based on high-affinity binding of mutated streptavidin and modified iminobiotin. Miniprotein antibodies were fused to a low immunogenic streptavidin variant, which was then expressed in Escherichia coli inclusion bodies, solubilized, and refolded into functional tetramers. The AMDC developed against human epidermal growth factor receptor 2 (HER2) effectively killed cultured cancer cells using bis-iminobiotin conjugated to photo-activating silicon phthalocyanine. The HER2-targeting AMDC was also effective in vivo against a mouse KPL-4 xenograft model. This AMDC platform provides rapid, stable, and high-yield therapeutics against multiple targets.
Collapse
Affiliation(s)
- Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takefumi Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Sho Aki
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Toshifumi Tatsumi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yudai Kaneko
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Takeshi Kawamura
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan; Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunyo-ku, Tokyo, 113-0032, Japan
| | - Mai Miura
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Masazumi Ishii
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Kei Ohkubo
- Institute for Advanced Co-Creation Studies, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-1 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tsuyoshi Osawa
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Akira Sugiyama
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan; Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunyo-ku, Tokyo, 113-0032, Japan.
| |
Collapse
|
15
|
Abstract
Phosphoinositides are signalling lipids derived from phosphatidylinositol, a ubiquitous phospholipid in the cytoplasmic leaflet of eukaryotic membranes. Initially discovered for their roles in cell signalling, phosphoinositides are now widely recognized as key integrators of membrane dynamics that broadly impact on all aspects of cell physiology and on disease. The past decade has witnessed a vast expansion of our knowledge of phosphoinositide biology. On the endocytic and exocytic routes, phosphoinositides direct the inward and outward flow of membrane as vesicular traffic is coupled to the conversion of phosphoinositides. Moreover, recent findings on the roles of phosphoinositides in autophagy and the endolysosomal system challenge our view of lysosome biology. The non-vesicular exchange of lipids, ions and metabolites at membrane contact sites in between organelles has also been found to depend on phosphoinositides. Here we review our current understanding of how phosphoinositides shape and direct membrane dynamics to impact on cell physiology, and provide an overview of emerging concepts in phosphoinositide regulation.
Collapse
|
16
|
Hamila SA, Ooms LM, Rodgers SJ, Mitchell CA. The INPP4B paradox: Like PTEN, but different. Adv Biol Regul 2021; 82:100817. [PMID: 34216856 DOI: 10.1016/j.jbior.2021.100817] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/28/2021] [Accepted: 06/10/2021] [Indexed: 06/13/2023]
Abstract
Cancer is a complex and heterogeneous disease marked by the dysregulation of cancer driver genes historically classified as oncogenes or tumour suppressors according to their ability to promote or inhibit tumour development and growth, respectively. Certain genes display both oncogenic and tumour suppressor functions depending on the biological context, and as such have been termed dual-role cancer driver genes. However, because of their context-dependent behaviour, the tumourigenic mechanism of many dual-role genes is elusive and remains a significant knowledge gap in our effort to understand and treat cancer. Inositol polyphosphate 4-phosphatase type II (INPP4B) is an emerging dual-role cancer driver gene, primarily known for its role as a negative regulator of the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway. In response to growth factor stimulation, class I PI3K generates PtdIns(3,4,5)P3 at the plasma membrane. PtdIns(3,4,5)P3 can be hydrolysed by inositol polyphosphate 5-phosphatases to generate PtdIns(3,4)P2, which, together with PtdIns(3,4,5)P3, facilitates the activation of AKT to promote cell proliferation, survival, migration, and metabolism. Phosphatase and tensin homology on chromosome 10 (PTEN) and INPP4B are dual-specificity phosphatases that hydrolyse PtdIns(3,4,5)P3 and PtdIns(3,4)P2, respectively, and thus negatively regulate PI3K/AKT signalling. PTEN is a bona fide tumour suppressor that is frequently lost in human tumours. INPP4B was initially characterised as a tumour suppressor akin to PTEN, and has been implicated as such in a number of cancers, including prostate, thyroid, and basal-like breast cancers. However, evidence has since emerged revealing INPP4B as a paradoxical oncogene in several malignancies, with increased INPP4B expression reported in AML, melanoma and colon cancers among others. Although the tumour suppressive function of INPP4B has been mostly ascribed to its ability to negatively regulate PI3K/AKT signalling, its oncogenic function remains less clear, with proposed mechanisms including promotion of PtdIns(3)P-dependent SGK3 signalling, inhibition of PTEN-dependent AKT activation, and enhancing DNA repair mechanisms to confer chemoresistance. Nevertheless, research is ongoing to identify the factors that dictate the tumourigenic output of INPP4B in different human cancers. In this review we discuss the dualistic role that INPP4B plays in the context of cancer development, progression and treatment, drawing comparisons to PTEN to explore how their similarities and, importantly, their differences may account for their diverging roles in tumourigenesis.
Collapse
Affiliation(s)
- Sabryn A Hamila
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Lisa M Ooms
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Samuel J Rodgers
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
17
|
Kostopoulou N, Bellou S, Bagli E, Markou M, Kostaras E, Hyvönen M, Kalaidzidis Y, Papadopoulos A, Chalmantzi V, Kyrkou A, Panopoulou E, Fotsis T, Murphy C. Embryonic stem cells are devoid of macropinocytosis, a trafficking pathway for activin A in differentiated cells. J Cell Sci 2021; 134:jcs246892. [PMID: 34313314 DOI: 10.1242/jcs.246892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Ligand-receptor complexes formed at the plasma membrane are internalised via various endocytic pathways that influence the ultimate signalling output by regulating the selection of interaction partners by the complex along the trafficking route. We report that, in differentiated cells, activin A-receptor complexes are internalised via clathrin-mediated endocytosis (CME) and macropinocytosis (MP), whereas in human embryonic stem cells (hESCs) internalisation occurs via CME. We further show that hESCs are devoid of MP, which becomes functional upon differentiation towards endothelial cells through mesoderm mediators. Our results reveal, for the first time, that MP is an internalisation route for activin A in differentiated cells, and that MP is not active in hESCs and is induced as cells differentiate.
Collapse
Affiliation(s)
- Nikoleta Kostopoulou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Sofia Bellou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Confocal Laser Scanning Microscopy Unit, Network of Research Supporting Laboratories, University of Ioannina, Ioannina, 45110, Greece
| | - Eleni Bagli
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Maria Markou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Eleftherios Kostaras
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Yiannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Angelos Papadopoulos
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Varvara Chalmantzi
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Athena Kyrkou
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
| | - Ekaterini Panopoulou
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Theodore Fotsis
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- Laboratory of Biological Chemistry, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Carol Murphy
- Foundation for Research & Technology-Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), Department of Biomedical Research, Ioannina, 45110, Greece
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham, A118 Aston Webb, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
18
|
Class II phosphatidylinositol 3-kinase isoforms in vesicular trafficking. Biochem Soc Trans 2021; 49:893-901. [PMID: 33666217 PMCID: PMC8106491 DOI: 10.1042/bst20200835] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 01/14/2023]
Abstract
Phosphatidylinositol 3-kinases (PI3Ks) are critical regulators of many cellular processes including cell survival, proliferation, migration, cytoskeletal reorganization, and intracellular vesicular trafficking. They are a family of lipid kinases that phosphorylate membrane phosphoinositide lipids at the 3′ position of their inositol rings, and in mammals they are divided into three classes. The role of the class III PI3K Vps34 is well-established, but recent evidence suggests the physiological significance of class II PI3K isoforms in vesicular trafficking. This review focuses on the recently discovered functions of the distinct PI3K-C2α and PI3K-C2β class II PI3K isoforms in clathrin-mediated endocytosis and consequent endosomal signaling, and discusses recently reported data on class II PI3K isoforms in different physiological contexts in comparison with class I and III isoforms.
Collapse
|
19
|
Class II phosphatidylinositol 3-kinase-C2α is essential for Notch signaling by regulating the endocytosis of γ-secretase in endothelial cells. Sci Rep 2021; 11:5199. [PMID: 33664344 PMCID: PMC7933152 DOI: 10.1038/s41598-021-84548-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/16/2021] [Indexed: 01/09/2023] Open
Abstract
The class II α-isoform of phosphatidylinositol 3-kinase (PI3K-C2α) plays a crucial role in angiogenesis at least in part through participating in endocytosis and, thereby, endosomal signaling of several cell surface receptors including VEGF receptor-2 and TGFβ receptor in vascular endothelial cells (ECs). The Notch signaling cascade regulates many cellular processes including cell proliferation, cell fate specification and differentiation. In the present study, we explored a role of PI3K-C2α in Delta-like 4 (Dll4)-induced Notch signaling in ECs. We found that knockdown of PI3K-C2α inhibited Dll4-induced generation of the signaling molecule Notch intracellular domain 1 (NICD1) and the expression of Notch1 target genes including HEY1, HEY2 and NOTCH3 in ECs but not in vascular smooth muscle cells. PI3K-C2α knockdown did not inhibit Dll4-induced endocytosis of cell surface Notch1. In contrast, PI3K-C2α knockdown as well as clathrin heavy chain knockdown impaired endocytosis of Notch1-cleaving protease, γ-secretase complex, with the accumulation of Notch1 at the perinuclear endolysosomes. Pharmacological blockage of γ-secretase also induced the intracellular accumulation of Notch1. Taken together, we conclude that PI3K-C2α is required for the clathrin-mediated endocytosis of γ-secretase complex, which allows for the cleavage of endocytosed Notch1 by γ-secretase complex at the endolysosomes to generate NICD1 in ECs.
Collapse
|
20
|
Koch PA, Dornan GL, Hessenberger M, Haucke V. The molecular mechanisms mediating class II PI 3-kinase function in cell physiology. FEBS J 2021; 288:7025-7042. [PMID: 33387369 DOI: 10.1111/febs.15692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) family of lipid-modifying enzymes plays vital roles in cell signaling and membrane trafficking through the production of 3-phosphorylated phosphoinositides. Numerous studies have analyzed the structure and function of class I and class III PI3Ks. In contrast, we know comparably little about the structure and physiological functions of the class II enzymes. Only recent studies have begun to unravel their roles in development, endocytic and endolysosomal membrane dynamics, signal transduction, and cell migration, while the mechanisms that control their localization and enzymatic activity remain largely unknown. Here, we summarize our current knowledge of the class II PI3Ks and outline open questions related to their structure, enzymatic activity, and their physiological and pathophysiological functions.
Collapse
Affiliation(s)
- Philipp Alexander Koch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Faculty of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Germany
| | | | - Manuel Hessenberger
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Faculty of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Germany
| |
Collapse
|
21
|
Islam S, Yoshioka K, Aki S, Ishimaru K, Yamada H, Takuwa N, Takuwa Y. Class II phosphatidylinositol 3-kinase α and β isoforms are required for vascular smooth muscle Rho activation, contraction and blood pressure regulation in mice. J Physiol Sci 2020; 70:18. [PMID: 32192434 PMCID: PMC7082390 DOI: 10.1186/s12576-020-00745-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Class II phosphatidylinositol 3-kinases (PI3K), PI3K-C2α and PI3K-C2β, are involved in cellular processes including endocytosis, cilia formation and autophagy. However, the role of PI3K-C2α and PI3K-C2β at the organismal level is not well understood. We found that double knockout (KO) mice with both smooth muscle-specific KO of PI3K-C2α and global PI3K-C2β KO, but not single KO mice of either PI3K-C2α or PI3K-C2β, exhibited reductions in arterial blood pressure and substantial attenuation of contractile responses of isolated aortic rings. In wild-type vascular smooth muscle cells, double knockdown of PI3K-C2α and PI3K-C2β but not single knockdown of either PI3K markedly inhibited contraction with reduced phosphorylation of 20-kDa myosin light chain and MYPT1 and Rho activation, but without inhibition of the intracellular Ca2+ mobilization. These data indicate that PI3K-C2α and PI3K-C2β play the redundant but essential role for vascular smooth muscle contraction and blood pressure regulation mainly through their involvement in Rho activation.
Collapse
MESH Headings
- Animals
- Blood Pressure/physiology
- Calcium/metabolism
- Cells, Cultured
- Class II Phosphatidylinositol 3-Kinases/genetics
- Class II Phosphatidylinositol 3-Kinases/metabolism
- Disease Models, Animal
- Isoenzymes
- Mice
- Mice, Knockout
- Muscle Contraction/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Shahidul Islam
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Sho Aki
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kazuhiro Ishimaru
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroki Yamada
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Noriko Takuwa
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
- Department of Health Science, Ishikawa Prefectural University, Kahoku, Ishikawa, 929-1210, Japan
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|