1
|
Brambillasca S, Cera MR, Andronache A, Dey SK, Fagá G, Fancelli D, Frittoli E, Pasi M, Robusto M, Varasi M, Scita G, Mercurio C. Novel selective inhibitors of macropinocytosis-dependent growth in pancreatic ductal carcinoma. Biomed Pharmacother 2024; 177:116991. [PMID: 38906021 PMCID: PMC11287759 DOI: 10.1016/j.biopha.2024.116991] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Macropinocytosis is a cellular process that enables cells to engulf extracellular material, such as nutrients, growth factors, and even whole cells. It is involved in several physiological functions as well as pathological conditions. In cancer cells, macropinocytosis plays a crucial role in promoting tumor growth and survival under nutrient-limited conditions. In particular KRAS mutations have been identified as main drivers of macropinocytosis in pancreatic, breast, and non-small cell lung cancers. We performed a high-content screening to identify inhibitors of macropinocytosis in pancreatic ductal adenocarcinoma (PDAC)-derived cells, aiming to prevent nutrient scavenging of PDAC tumors. The screening campaign was conducted in a well-known pancreatic KRAS-mutated cell line (MIAPaCa-2) cultured under nutrient deprivation and using FITC-dextran to precisely quantify macropinocytosis. We assembled a collection of 3584 small molecules, including drugs approved by the Food and Drug Administration (FDA), drug-like molecules against molecular targets, kinase-targeted compounds, and molecules designed to hamper protein-protein interactions. We identified 28 molecules that inhibited macropinocytosis, with potency ranging from 0.4 to 29.9 μM (EC50). A few of them interfered with other endocytic pathways, while 11 compounds did not and were therefore considered specific "bona fide" macropinocytosis inhibitors and further characterized. Four compounds (Ivermectin, Tyrphostin A9, LY2090314, and Pyrvinium Pamoate) selectively hampered nutrient scavenging in KRAS-mutated cancer cells. Their ability to impair albumin-dependent proliferation was replicated both in different 2D cell culture systems and 3D organotypic models. These findings provide a new set of compounds specifically targeting macropinocytosis, which could have therapeutic applications in cancer and infectious diseases.
Collapse
Affiliation(s)
- Silvia Brambillasca
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Maria Rosaria Cera
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Adrian Andronache
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Sumit Kumar Dey
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giovanni Fagá
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Daniele Fancelli
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Maurizio Pasi
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Michela Robusto
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mario Varasi
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giorgio Scita
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy.
| | - Ciro Mercurio
- Experimental Therapeutics Program, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
2
|
Kumari S, Raj S, Babu MA, Bhatti GK, Bhatti JS. Antibody-drug conjugates in cancer therapy: innovations, challenges, and future directions. Arch Pharm Res 2024; 47:40-65. [PMID: 38153656 DOI: 10.1007/s12272-023-01479-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
The emergence of antibody-drug conjugates (ADCs) as a potential therapeutic avenue in cancer treatment has garnered significant attention. By combining the selective specificity of monoclonal antibodies with the cytotoxicity of drug molecules, ADCs aim to increase the therapeutic index, selectively targeting cancer cells while minimizing systemic toxicity. Various ADCs have been licensed for clinical usage, with ongoing research paving the way for additional options. However, the manufacture of ADCs faces several challenges. These include identifying suitable target antigens, enhancing antibodies, linkers, and payloads, and managing resistance mechanisms and side effects. This review focuses on the strategies to overcome these hurdles, such as site-specific conjugation techniques, novel antibody formats, and combination therapy. Our focus lies on current advancements in antibody engineering, linker technology, and cytotoxic payloads while addressing the challenges associated with ADC development. Furthermore, we explore the future potential of personalized medicine, leveraging individual patients' molecular profiles, to propel ADC treatments forward. As our understanding of the molecular mechanisms driving cancer progression continues to expand, we anticipate the development of new ADCs that offer more effective and personalized therapeutic options for cancer patients.
Collapse
Affiliation(s)
- Shivangi Kumari
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Sonam Raj
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
3
|
Elechalawar CK, Rao G, Gulla SK, Patel MM, Frickenstein A, Means N, Roy RV, Tsiokas L, Asfa S, Panja P, Rao C, Wilhelm S, Bhattacharya R, Mukherjee P. Gold Nanoparticles Inhibit Macropinocytosis by Decreasing KRAS Activation. ACS NANO 2023; 17:9326-9337. [PMID: 37129853 PMCID: PMC10718652 DOI: 10.1021/acsnano.3c00920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The RAS-transformed cells utilize macropinocytosis to acquire amino acids to support their uncontrolled growth. However, targeting RAS to inhibit macropinocytosis remains a challenge. Here, we report that gold nanoparticles (GNP) inhibit macropinocytosis by decreasing KRAS activation. Using surface-modified and unmodified GNP, we showed that unmodified GNP specifically sequestered both wild-type and mutant KRAS and inhibited its activation, irrespective of growth factor stimulation, while surface-passivated GNP had no effect. Alteration of KRAS activation is reflected on downstream signaling cascades, macropinocytosis and tumor cell growth in vitro, and two independent preclinical human xenograft models of pancreatic cancer in vivo. The current study demonstrates NP-mediated inhibition of macropinocytosis and KRAS activation and provides translational opportunities to inhibit tumor growth in a number of cancers where activation of KRAS plays a major role.
Collapse
Affiliation(s)
- Chandra Kumar Elechalawar
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Geeta Rao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Suresh Kumar Gulla
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Maulin Mukeshchandra Patel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Alex Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Nicolas Means
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Ram Vinod Roy
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Leonidas Tsiokas
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Sima Asfa
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Prasanta Panja
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Chinthalapally Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
4
|
Griffiths G, Gruenberg J, Marsh M, Wohlmann J, Jones AT, Parton RG. Nanoparticle entry into cells; the cell biology weak link. Adv Drug Deliv Rev 2022; 188:114403. [PMID: 35777667 DOI: 10.1016/j.addr.2022.114403] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
Nanoparticles (NP) are attractive options for the therapeutic delivery of active pharmaceutical drugs, proteins and nucleic acids into cells, tissues and organs. Research into the development and application of NP most often starts with a diverse group of scientists, including chemists, bioengineers and material and pharmaceutical scientists, who design, fabricate and characterize NP in vitro (Stage 1). The next step (Stage 2) generally investigates cell toxicity as well as the processes by which NP bind, are internalized and deliver their cargo to appropriate model tissue culture cells. Subsequently, in Stage 3, selected NP are tested in animal systems, mostly mouse. Whereas the chemistry-based development and analysis in Stage 1 is increasingly sophisticated, the investigations in Stage 2 are not what could be regarded as 'state-of-the-art' for the cell biology field and the quality of research into NP interactions with cells is often sub-standard. In this review we describe our current understanding of the mechanisms by which particles gain entry into mammalian cells via endocytosis. We summarize the most important areas for concern, highlight some of the most common mis-conceptions, and identify areas where NP scientists could engage with trained cell biologists. Our survey of the different mechanisms of uptake into cells makes us suspect that claims for roles for caveolae, as well as macropinocytosis, in NP uptake into cells have been exaggerated, whereas phagocytosis has been under-appreciated.
Collapse
Affiliation(s)
- Gareth Griffiths
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway.
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211-Geneva-4, Switzerland
| | - Mark Marsh
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jens Wohlmann
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, Wales CF103NB, UK
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Qld 4072, Australia
| |
Collapse
|
5
|
Abstract
Macropinocytosis is an evolutionarily conserved endocytic pathway that mediates the nonselective acquisition of extracellular material via large endocytic vesicles known as macropinosomes. In addition to other functions, this uptake pathway supports cancer cell metabolism through the uptake of nutrients. Cells harboring oncogene or tumor suppressor mutations are known to display heightened macropinocytosis, which confers to the cancer cells the ability to survive and proliferate despite the nutrient-scarce conditions of the tumor microenvironment. Thus, macropinocytosis is associated with cancer malignancy. Macropinocytic uptake can be induced in cancer cells by different stress stimuli, acting as an adaptive mechanism for the cells to resist stresses in the tumor milieu. Here, we review the cellular stresses that are known to promote macropinocytosis, as well as the underlying molecular mechanisms that drive this process.
Collapse
Affiliation(s)
- Guillem Lambies
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Cosimo Commisso
- Cell and Molecular Biology of Cancer Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
6
|
Abstract
Macropinocytosis is an evolutionarily conserved endocytic pathway that mediates non-selective uptake of extracellular fluid in bulk. Macropinocytosis is initiated by localized polymerization of the actin cytoskeleton, which generates plasma membrane protrusions that enclose part of the environment into large endocytic vesicles. From amoebae to mammalian cells, the actin dynamics that drive macropinosome formation are regulated by a conserved set of intracellular signaling proteins including Ras superfamily GTPases and PI3-kinases. In mammalian cells, multiple upstream signaling pathways control activity of these core regulators in response to cell-extrinsic and cell-intrinsic stimuli. Growth factor signaling pathways play a central role in macropinocytosis induction. In addition, an increasing number of functionally diverse processes has been identified as macropinocytosis regulators, including several nutrient-sensing and developmental signaling pathways. Many of these signaling pathways have proto-oncogenic properties, and their dysregulation drives the high macropinocytic activity that is commonly observed in cancer cells. These regulatory principles illustrate how macropinocytosis is controlled by complex upstream inputs to exert diverse cellular functions in physiological and pathological contexts.
Collapse
|
7
|
Hanada K, Kawada K, Nishikawa G, Toda K, Maekawa H, Nishikawa Y, Masui H, Hirata W, Okamoto M, Kiyasu Y, Honma S, Ogawa R, Mizuno R, Itatani Y, Miyoshi H, Sasazuki T, Shirasawa S, Taketo MM, Obama K, Sakai Y. Dual blockade of macropinocytosis and asparagine bioavailability shows synergistic anti-tumor effects on KRAS-mutant colorectal cancer. Cancer Lett 2021; 522:129-141. [PMID: 34543685 DOI: 10.1016/j.canlet.2021.09.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023]
Abstract
Mutations of KRAS gene are found in various types of cancer, including colorectal cancer (CRC). Despite intense efforts, no pharmacological approaches are expected to be effective against KRAS-mutant cancers. Macropinocytosis is an evolutionarily conserved actin-dependent endocytic process that internalizes extracellular fluids into large vesicles called macropinosomes. Recent studies have revealed macropinocytosis's important role in metabolic adaptation to nutrient stress in cancer cells harboring KRAS mutations. Here we showed that KRAS-mutant CRC cells enhanced macropinocytosis for tumor growth under nutrient-depleted conditions. We also demonstrated that activation of Rac1 and phosphoinositide 3-kinase were involved in macropinocytosis of KRAS-mutant CRC cells. Furthermore, we found that macropinocytosis was closely correlated with asparagine metabolism. In KRAS-mutant CRC cells engineered with knockdown of asparagine synthetase, macropinocytosis was accelerated under glutamine-depleted condition, and albumin addition could restore the glutamine depletion-induced growth suppression by recovering the intracellular asparagine level. Finally, we discovered that the combination of macropinocytosis inhibition and asparagine depletion dramatically suppressed the tumor growth of KRAS-mutant CRC cells in vivo. These results indicate that dual blockade of macropinocytosis and asparagine bioavailability could be a novel therapeutic strategy for KRAS-mutant cancers.
Collapse
Affiliation(s)
- Keita Hanada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Gen Nishikawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kosuke Toda
- Department of Surgery, Otsu City Hospital, Otsu, Japan
| | - Hisatsugu Maekawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuyo Nishikawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideyuki Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Hirata
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Michio Okamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiyuki Kiyasu
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shusaku Honma
- Department of Surgery, Kobe City Medical Center West Hospital, Kobe, Japan
| | - Ryotaro Ogawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rei Mizuno
- Department of Surgery, Uji Tokushukai Medical Center, Kyoto, Japan
| | - Yoshiro Itatani
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Miyoshi
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | | | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - M Mark Taketo
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Osaka Red Cross Hospital, Osaka, Japan
| |
Collapse
|
8
|
Zhao Y, Li X, Zhang W, Yu L, Wang Y, Deng Z, Liu M, Mo S, Wang R, Zhao J, Liu S, Hao Y, Wang X, Ji T, Zhang L, Wang C. Trends in the biological functions and medical applications of extracellular vesicles and analogues. Acta Pharm Sin B 2021; 11:2114-2135. [PMID: 34522580 PMCID: PMC8424226 DOI: 10.1016/j.apsb.2021.03.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/19/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Natural extracellular vesicles (EVs) play important roles in many life processes such as in the intermolecular transfer of substances and genetic information exchanges. Investigating the origins and working mechanisms of natural EVs may provide an understanding of life activities, especially regarding the occurrence and development of diseases. Additionally, due to their vesicular structure, EVs (in small molecules, nucleic acids, proteins, etc.) could act as efficient drug-delivery carriers. Herein, we describe the sources and biological functions of various EVs, summarize the roles of EVs in disease diagnosis and treatment, and review the application of EVs as drug-delivery carriers. We also assess the challenges and perspectives of EVs in biomedical applications.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiaolu Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Wenbo Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Lanlan Yu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yang Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Zhun Deng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Mingwei Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Shanshan Mo
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ruonan Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jinming Zhao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Shuli Liu
- Department of Clinical Laboratory, Peking University Civil Aviation School of Clinical Medicine, Beijing 100123, China
| | - Yun Hao
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Xiangdong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Tianjiao Ji
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Chenxuan Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
9
|
Hammood M, Craig AW, Leyton JV. Impact of Endocytosis Mechanisms for the Receptors Targeted by the Currently Approved Antibody-Drug Conjugates (ADCs)-A Necessity for Future ADC Research and Development. Pharmaceuticals (Basel) 2021; 14:ph14070674. [PMID: 34358100 PMCID: PMC8308841 DOI: 10.3390/ph14070674] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Biologically-based therapies increasingly rely on the endocytic cycle of internalization and exocytosis of target receptors for cancer therapies. However, receptor trafficking pathways (endosomal sorting (recycling, lysosome localization) and lateral membrane movement) are often dysfunctional in cancer. Antibody-drug conjugates (ADCs) have revitalized the concept of targeted chemotherapy by coupling inhibitory antibodies to cytotoxic payloads. Significant advances in ADC technology and format, and target biology have hastened the FDA approval of nine ADCs (four since 2019). Although the links between aberrant endocytic machinery and cancer are emerging, the impact of dysregulated internalization processes of ADC targets and response rates or resistance have not been well studied. This is despite the reliance on ADC uptake and trafficking to lysosomes for linker cleavage and payload release. In this review, we describe what is known about all the target antigens for the currently approved ADCs. Specifically, internalization efficiency and relevant intracellular sorting activities are described for each receptor under normal processes, and when complexed to an ADC. In addition, we discuss aberrant endocytic processes that have been directly linked to preclinical ADC resistance mechanisms. The implications of endocytosis in regard to therapeutic effectiveness in the clinic are also described. Unexpectedly, information on endocytosis is scarce (absent for two receptors). Moreover, much of what is known about endocytosis is not in the context of receptor-ADC/antibody complexes. This review provides a deeper understanding of the pertinent principles of receptor endocytosis for the currently approved ADCs.
Collapse
Affiliation(s)
- Manar Hammood
- Departément de Medécine Nucléaire et Radiobiologie, Faculté de Medécine et des Sciences de la Santé, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Andrew W. Craig
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Jeffrey V. Leyton
- Departément de Medécine Nucléaire et Radiobiologie, Faculté de Medécine et des Sciences de la Santé, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Centre d’Imagerie Moleculaire, Centre de Recherche, CHUS, Sherbrooke, QC J1H 5N4, Canada
- Correspondence: ; Tel.: +1-819-346-1110
| |
Collapse
|
10
|
Chang YY, Enninga J, Stévenin V. New methods to decrypt emerging macropinosome functions during the host-pathogen crosstalk. Cell Microbiol 2021; 23:e13342. [PMID: 33848057 PMCID: PMC8365644 DOI: 10.1111/cmi.13342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Large volumes of liquid and other materials from the extracellular environment are internalised by eukaryotic cells via an endocytic process called macropinocytosis. It is now recognised that this fundamental and evolutionarily conserved pathway is hijacked by numerous intracellular pathogens as an entry portal to the host cell interior. Yet, an increasing number of additional cellular functions of macropinosomes in pathologic processes have been reported beyond this role for fluid internalisation. It emerges that the identity of macropinosomes can vary hugely and change rapidly during their lifetime. A deeper understanding of this important multi-faceted compartment is based on novel methods for their investigation. These methods are either imaging-based for the tracking of macropinosome dynamics, or they provide the means to extract macropinosomes at high purity for comprehensive proteomic analyses. Here, we portray these new approaches for the investigation of macropinosomes. We document how these method developments have provided insights for a new understanding of the intracellular lifestyle of the bacterial pathogens Shigella and Salmonella. We suggest that a systematic complete characterisation of macropinosome subversion with these approaches during other infection processes and pathologies will be highly beneficial for our understanding of the underlying cellular and molecular processes.
Collapse
Affiliation(s)
- Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.,Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France
| | - Virginie Stévenin
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Université Paris Diderot, Ecole doctorale BioSPC, Paris, France
| |
Collapse
|
11
|
Doodnauth SA, Grinstein S, Maxson ME. Constitutive and stimulated macropinocytosis in macrophages: roles in immunity and in the pathogenesis of atherosclerosis. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180147. [PMID: 30967001 DOI: 10.1098/rstb.2018.0147] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Macrophages respond to several stimuli by forming florid membrane ruffles that lead to fluid uptake by macropinocytosis. This type of induced macropinocytosis, executed by a variety of non-malignant and malignant cells, is initiated by transmembrane receptors and is involved in nutrient acquisition and mTOR signalling. However, macrophages also perform a unique type of constitutive ruffling and macropinocytosis that is dependent on the presence of extracellular calcium. Calcium-sensing receptors are responsible for this activity. This distinct form of macropinocytosis enables macrophages to continuously sample their microenvironment for antigenic molecules and for pathogen- and danger-associated molecular patterns, as part of their immune surveillance functions. Interestingly, even within the monocyte lineage, there are differences in macropinocytic ability that reflect the polarized functional roles of distinct macrophage subsets. This review discusses the shared and distinct features of both induced and constitutive macropinocytosis displayed by the macrophage lineage and their roles in physiology, immunity and pathophysiology. In particular, we analyse the role of macropinocytosis in the uptake of modified low-density lipoprotein (LDL) and its contribution to foam cell and atherosclerotic plaque formation. We propose a combined role of scavenger receptors and constitutive macropinocytosis in oxidized LDL uptake, a process we have termed 'receptor-assisted macropinocytosis'. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Sasha A Doodnauth
- 1 Princess Margaret Cancer Center, University Health Network , Toronto, ON , Canada M5G 1L7.,2 Department of Medical Biophysics, University of Toronto , Toronto, ON , Canada M5G 1L7
| | - Sergio Grinstein
- 3 Program in Cell Biology, Hospital for Sick Children , 686 Bay Street, Toronto, ON , Canada M5G 0A4.,4 Department of Biochemistry, University of Toronto , 1 King's Circle, Toronto, ON , Canada M5S 1A8.,5 Keenan Research Centre of the Li Ka Shing Knowledge Institute , St. Michael's Hospital, 290 Victoria Street, Toronto, ON , Canada M5C 1N8
| | - Michelle E Maxson
- 3 Program in Cell Biology, Hospital for Sick Children , 686 Bay Street, Toronto, ON , Canada M5G 0A4
| |
Collapse
|
12
|
Commisso C. The pervasiveness of macropinocytosis in oncological malignancies. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180153. [PMID: 30967003 DOI: 10.1098/rstb.2018.0153] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In tumour cells, macropinocytosis functions as an amino acid supply route and supports cancer cell survival and proliferation. Initially demonstrated in oncogenic KRAS-driven models of pancreatic cancer, macropinocytosis triggers the internalization of extracellular proteins via discrete endocytic vesicles called macropinosomes. The incoming protein cargo is targeted for lysosome-dependent degradation, causing the intracellular release of amino acids. These protein-derived amino acids support metabolic fitness by contributing to the intracellular amino acid pools, as well as to the biosynthesis of central carbon metabolites. In this way, macropinocytosis represents a novel amino acid supply route that tumour cells use to survive the nutrient-poor conditions of the tumour microenvironment. Macropinocytosis has also emerged as an entry mechanism for a variety of nanomedicines, suggesting that macropinocytosis regulation in the tumour setting can be harnessed for the delivery of anti-cancer therapeutics. A slew of recent studies point to the possibility that macropinocytosis is a pervasive feature of many different tumour types. In this review, we focus on the role of this important uptake mechanism in a variety of cancers and highlight the main molecular drivers of macropinocytosis in these malignancies. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute , La Jolla, CA 92037 , USA
| |
Collapse
|
13
|
Disanza A, Bisi S, Frittoli E, Malinverno C, Marchesi S, Palamidessi A, Rizvi A, Scita G. Is cell migration a selectable trait in the natural evolution of cancer development? Philos Trans R Soc Lond B Biol Sci 2019; 374:20180224. [PMID: 31431177 DOI: 10.1098/rstb.2018.0224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Selective evolutionary pressure shapes the processes and genes that enable cancer survival and expansion in a tumour-suppressive environment. A distinguishing lethal feature of malignant cancer is its dissemination and seeding of metastatic foci. A key requirement for this process is the acquisition of a migratory/invasive ability. However, how the migratory phenotype is selected for during the natural evolution of cancer and what advantage, if any, it might provide to the growing malignant cells remain open issues. In this opinion piece, we discuss three possible answers to these issues. We will examine lines of evidence from mathematical modelling of cancer evolution that indicate that migration is an intrinsic selectable property of malignant cells that directly impacts on growth dynamics and cancer geometry. Second, we will argue that migratory phenotypes can emerge as an adaptive response to unfavourable growth conditions and endow cells not only with the ability to move/invade, but also with specific metastatic traits, including drug resistance, self-renewal and survival. Finally, we will discuss the possibility that migratory phenotypes are coincidental events that emerge by happenstance in the natural evolution of cancer. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Andrea Disanza
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Sara Bisi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Emanuela Frittoli
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Chiara Malinverno
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.,Department of Oncology and Haemato-Oncology-DIPO, School of Medicine, University of Milan, Milan, Italy
| | - Stefano Marchesi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Andrea Palamidessi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Abrar Rizvi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.,Department of Oncology and Haemato-Oncology-DIPO, School of Medicine, University of Milan, Milan, Italy
| | - Giorgio Scita
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.,Department of Oncology and Haemato-Oncology-DIPO, School of Medicine, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Zhang Y, Commisso C. Macropinocytosis in Cancer: A Complex Signaling Network. Trends Cancer 2019; 5:332-334. [PMID: 31208695 DOI: 10.1016/j.trecan.2019.04.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 10/26/2022]
Abstract
Macropinocytosis is an important nutrient-scavenging pathway in numerous cancer types, including pancreatic, lung, prostate, and bladder. This Forum highlights recent work identifying the key regulators of macropinocytosis that support tumor cell fitness in different contexts, providing a unique framework for strategies to target macropinocytosis in the treatment of cancer.
Collapse
Affiliation(s)
- Yijuan Zhang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
15
|
Abstract
Macropinocytosis is an actin-driven form of clathrin-independent endocytosis that generates an enlarged structure, the macropinosome. Although many studies focus on signaling molecules and phosphoinositides involved in initiating macropinocytosis, the commitment to forming a macropinosome and the handling of that membrane have not been studied in detail. Here we show in HT1080 cells, a human fibrosarcoma cell line, a requirement for microtubules, dynein, the JIP3 microtubule motor scaffold protein, and Arf6, a JIP3 interacting protein, for the formation and inward movement of the macropinosome. While actin and myosin II also play critical roles in the formation of ruffling membrane, microtubules provide an important tract for initiation, sealing, and transport of the macropinosome through the actin- and myosin-rich lamellar region.
Collapse
Affiliation(s)
- Chad D Williamson
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, and.,Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Julie G Donaldson
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, and
| |
Collapse
|
16
|
Colin M, Delporte C, Janky R, Lechon AS, Renard G, Van Antwerpen P, Maltese WA, Mathieu V. Dysregulation of Macropinocytosis Processes in Glioblastomas May Be Exploited to Increase Intracellular Anti-Cancer Drug Levels: The Example of Temozolomide. Cancers (Basel) 2019; 11:cancers11030411. [PMID: 30909495 PMCID: PMC6468498 DOI: 10.3390/cancers11030411] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 12/16/2022] Open
Abstract
Macropinocytosis is a clathrin-independent endocytosis of extracellular fluid that may contribute to cancer aggressiveness through nutrient supply, recycling of plasma membrane and receptors, and exosome internalization. Macropinocytosis may be notably triggered by epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR), two well-known markers for glioblastoma aggressiveness. Therefore, we studied whether the expression of key actors of macropinocytosis is modified in human glioma datasets. Strong deregulation has been evidenced at the mRNA level according to the grade of the tumor, and 38 macropinocytosis-related gene signatures allowed discrimination of the glioblastoma (GBM) samples. Honokiol-induced vacuolization was then compared to vacquinol-1 and MOMIPP, two known macropinocytosis inducers. Despite high phase-contrast morphological similarities, honokiol-induced vacuoles appeared to originate from both endocytosis and ER. Also, acridine orange staining suggested differences in the macropinosomes’ fate: their fusion with lysosomes appeared very limited in 3-(5-methoxy -2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (MOMIPP)-treated cells. Nevertheless, each of the compounds markedly increased temozolomide uptake by glioma cells, as evidenced by LC-MS. In conclusion, the observed deregulation of macropinocytosis in GBM makes them prone to respond to various compounds affecting their formation and/or intracellular fate. Considering that sustained macropinocytosis may also trigger cell death of both sensitive and resistant GBM cells, we propose to envisage macropinocytosis inducers in combination approaches to obtain dual benefits: increased drug uptake and additive/synergistic effects.
Collapse
Affiliation(s)
- Margaux Colin
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - Cédric Delporte
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | | | - Anne-Sophie Lechon
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - Gwendoline Renard
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - Pierre Van Antwerpen
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Université libre de Bruxelles (ULB), 1050 Brussels, Belgium.
| | - William A Maltese
- Department of Cancer Biology, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium.
- ULB Cancer Research Center, Université libre de Bruxelles (ULB), 1050 Bruxelles, Belgium.
| |
Collapse
|
17
|
Dendo K, Yugawa T, Nakahara T, Ohno SI, Goshima N, Arakawa H, Kiyono T. Induction of non-apoptotic programmed cell death by oncogenic RAS in human epithelial cells and its suppression by MYC overexpression. Carcinogenesis 2018; 39:202-213. [PMID: 29106503 PMCID: PMC5862353 DOI: 10.1093/carcin/bgx124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 10/27/2017] [Indexed: 12/17/2022] Open
Abstract
Oncogenic mutations of RAS genes, found in about 30% of human cancers, are considered to play important roles in cancer development. However, oncogenic RAS can also induce senescence in mouse and human normal fibroblasts. In some cell lines, oncogenic RAS has been reported to induce non-apoptotic programed cell death (PCD). Here, we investigated effects of oncogenic RAS expression in several types of normal human epithelial cells. Oncogenic RAS but not wild-type RAS stimulated macropinocytosis with accumulation of large-phase lucent vacuoles in the cytoplasm, subsequently leading to cell death which was indistinguishable from a recently proposed new type of PCD, methuosis. A RAC1 inhibitor suppressed accumulation of macropinosomes and overexpression of MYC attenuated oncogenic RAS-induced such accumulation, cell cycle arrest and cell death. MYC suppression or rapamycin treatment in some cancer cell lines harbouring oncogenic mutations in RAS genes induced cell death with accumulation of macropinosomes. These results suggest that this type of non-apoptotic PCD is a tumour-suppressing mechanism acting against oncogenic RAS mutations in normal human epithelial cells, which can be overcome by MYC overexpression, raising the possibility that its induction might be a novel approach to treatment of RAS-mutated human cancers.
Collapse
Affiliation(s)
- Kasumi Dendo
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan.,Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima, Bunkyou-ku, Tokyo, Japan
| | - Takashi Yugawa
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Tomomi Nakahara
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Shin-Ichi Ohno
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Naoki Goshima
- Molecular Profiling Research Center for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Aomi, Koto-ku, Tokyo, Japan
| | - Hirofumi Arakawa
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima, Bunkyou-ku, Tokyo, Japan.,Division of Cancer Biology, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Tohru Kiyono
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan
| |
Collapse
|
18
|
GRP75 modulates oncogenic Dbl-driven endocytosis derailed via the CHIP-mediated ubiquitin degradation pathway. Cell Death Dis 2018; 9:971. [PMID: 30250167 PMCID: PMC6155137 DOI: 10.1038/s41419-018-1039-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/13/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023]
Abstract
Chaperone-assisted proteasome degradation of oncogenic protein acts as an upstream signal controlling tumorigenesis and progression. The understanding of the co-regulation of chaperone and oncoprotein of endocytosis pathways is extremely limited. In this study, we showed for the first time that proto-Dbl (dbl proto-oncogene product) is co-enriched with mitochondrial chaperone GRP75 in endocytosis vesicles from ovarian cancer cells. onco-Dbl, produced by oncogenic mutation/degradation of proto-Dbl, markedly enhanced cellular macropinocytosis but suppressed clathrin-mediated endocytosis and clathrin-independent endocytosis pathways, presenting a derailed endocytosis phenotype. GRP75 was associated with proto-Dbl inside cells and modulated Dbl-driven endocytosis derailed by a co-regulatory mode. In spite of not being a component of the Hsc70/Hsp90/proto-Dbl complex, the degradation of proto-Dbl was promoted by GRP75 through the CHIP-mediated ubiquitin–proteasome pathway, of which GRP75 acts as a cooperator with CHIP but also acts as a competitor to Hsc70 and Hsp90 in the multiple chaperones-assisted pro-folding/pro-degradation machinery. Knockdown or inhibition of GRP75 attenuated proto-Dbl degradation and reduced the onco-Dbl level, which differentially impaired Rho GTPases activation and therefore shifted the endocytosis-derailed phenotype. Our data uncovered a novel GRP75-Dbl endocytosis regulatory axis and provided an alternative using chaperone inhibitor to shut down the oncoprotein-driven endocytosis derailment mechanism.
Collapse
|
19
|
Segerer G, Engelmann D, Kaestner A, Trötzmüller M, Köfeler H, Stigloher C, Thiele C, Jeanclos E, Gohla A. A phosphoglycolate phosphatase/AUM-dependent link between triacylglycerol turnover and epidermal growth factor signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2018. [DOI: 10.1016/j.bbalip.2018.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Yoshida S, Pacitto R, Inoki K, Swanson J. Macropinocytosis, mTORC1 and cellular growth control. Cell Mol Life Sci 2018; 75:1227-1239. [PMID: 29119228 PMCID: PMC5843684 DOI: 10.1007/s00018-017-2710-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/03/2017] [Indexed: 12/15/2022]
Abstract
The growth and proliferation of metazoan cells are driven by cellular nutrient status and by extracellular growth factors. Growth factor receptors on cell surfaces initiate biochemical signals that increase anabolic metabolism and macropinocytosis, an actin-dependent endocytic process in which relatively large volumes of extracellular solutes and nutrients are internalized and delivered efficiently into lysosomes. Macropinocytosis is prominent in many kinds of cancer cells, and supports the growth of cells transformed by oncogenic K-Ras. Growth factor receptor signaling and the overall metabolic status of the cell are coordinated in the cytoplasm by the mechanistic target-of-rapamycin complex-1 (mTORC1), which positively regulates protein synthesis and negatively regulates molecular salvage pathways such as autophagy. mTORC1 is activated by two distinct Ras-related small GTPases, Rag and Rheb, which associate with lysosomal membranes inside the cell. Rag recruits mTORC1 to the lysosomal surface where Rheb directly binds to and activates mTORC1. Rag is activated by both lysosomal luminal and cytosolic amino acids; Rheb activation requires phosphoinositide 3-kinase, Akt, and the tuberous sclerosis complex-1/2. Signals for activation of Rag and Rheb converge at the lysosomal membrane, and several lines of evidence support the idea that growth factor-dependent endocytosis facilitates amino acid transfer into the lysosome leading to the activation of Rag. This review summarizes evidence that growth factor-stimulated macropinocytosis is essential for amino acid-dependent activation of mTORC1, and that increased solute accumulation by macropinocytosis in transformed cells supports unchecked cell growth.
Collapse
Affiliation(s)
- Sei Yoshida
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA
| | - Regina Pacitto
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA
| | - Ken Inoki
- Department of Integrative and Molecular Physiology and Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joel Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA.
| |
Collapse
|
21
|
Sigismund S, Scita G. The 'endocytic matrix reloaded' and its impact on the plasticity of migratory strategies. Curr Opin Cell Biol 2018; 54:9-17. [PMID: 29544103 DOI: 10.1016/j.ceb.2018.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 12/14/2022]
Abstract
An explosive growth in knowledge, in the last two decades, has conferred a new dimension to the process of endocytosis. Endocytic circuitries have come into focus as a pervasive system that controls virtual all aspects of cell biology. A few years ago, we proposed the term 'endocytic matrix' to define a cellular network of signalling wiring that is at the core of the cellular blueprint. A primary role of the endocytic matrix is the delivery of space-resolved and time-resolved signals to the cell in an interpretable format, and, as such, it has profound consequences on polarized cellular and supra-cellular functions, first and foremost, cell motility. Here, we describe a set of recent results that expand this notion and illuminate how endocytic matrix dynamically controls the plasticity of migratory strategies. We further highlight the impact of inter-organelle contact sites on motility and the role of organelle positioning in this process. Finally, we illustrate how global perturbation of the endocytic circuitry influences cellular and supra-cellular mechanics, ultimately controlling a solid-to-liquid-like transition in the mode of motility with potential consequences on cancer dissemination.
Collapse
Affiliation(s)
- Sara Sigismund
- IFOM, The FIRC Institute of Molecular Oncology, via Adamello 16, 20139 Milan, Italy; Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, 20139 Milan, Italy
| | - Giorgio Scita
- IFOM, The FIRC Institute of Molecular Oncology, via Adamello 16, 20139 Milan, Italy; University of Milan, School of Medicine, Department of Oncology and Hemato-Oncology-DIPO, Milan, Italy.
| |
Collapse
|
22
|
Kühn S, Lopez-Montero N, Chang YY, Sartori-Rupp A, Enninga J. Imaging macropinosomes during Shigella infections. Methods 2017; 127:12-22. [PMID: 28522322 DOI: 10.1016/j.ymeth.2017.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/11/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022] Open
Abstract
Macropinocytosis is the uptake of extracellular fluid within vesicles of varying size that takes place during numerous cellular processes in a large variety of cells. A growing number of pathogens, including viruses, parasites, and bacteria are known to induce macropinocytosis during their entry into targeted host cells. We have recently discovered that the human enteroinvasive, bacterial pathogen Shigella causes in situ macropinosome formation during its entry into epithelial cells. These infection-associated macropinosomes are not generated to ingest the bacteria, but are instead involved in Shigella's intracellular niche formation. They make contacts with the phagocytosed shigellae to promote vacuolar membrane rupture and their cytosolic release. Here, we provide an overview of the different imaging approaches that are currently used to analyze macropinocytosis during infectious processes with a focus on Shigella entry. We detail the advantages and disadvantages of genetically encoded reporters as well as chemical probes to trace fluid phase uptake. In addition, we report how such reporters can be combined with ultrastructural approaches for correlative light electron microscopy either in thin sections or within large volumes. The combined imaging techniques introduced here provide a detailed characterization of macropinosomes during bacterial entry, which, apart from Shigella, are relevant for numerous other ones, including Salmonella, Brucella or Mycobacteria.
Collapse
Affiliation(s)
- Sonja Kühn
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | | | - Yuen-Yan Chang
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Anna Sartori-Rupp
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Jost Enninga
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France.
| |
Collapse
|
23
|
SNAT7 is the primary lysosomal glutamine exporter required for extracellular protein-dependent growth of cancer cells. Proc Natl Acad Sci U S A 2017; 114:E3602-E3611. [PMID: 28416685 DOI: 10.1073/pnas.1617066114] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lysosomes degrade cellular components sequestered by autophagy or extracellular material internalized by endocytosis and phagocytosis. The macromolecule building blocks released by lysosomal hydrolysis are then exported to the cytosol by lysosomal transporters, which remain undercharacterized. In this study, we designed an in situ assay of lysosomal amino acid export based on the transcription factor EB (TFEB), a master regulator of lysosomal biogenesis that detects lysosomal storage. This assay was used to screen candidate lysosomal transporters, leading to the identification of sodium-coupled neutral amino acid transporter 7 (SNAT7), encoded by the SLC38A7 gene, as a lysosomal transporter highly selective for glutamine and asparagine. Cell fractionation confirmed the lysosomal localization of SNAT7, and flux measurements confirmed its substrate selectivity and showed a strong activation by the lysosomal pH gradient. Interestingly, gene silencing or editing experiments revealed that SNAT7 is the primary permeation pathway for glutamine across the lysosomal membrane and it is required for growth of cancer cells in a low free-glutamine environment, when macropinocytosis and lysosomal degradation of extracellular proteins are used as an alternative source of amino acids. SNAT7 may, thus, represent a novel target for glutamine-related anticancer therapies.
Collapse
|
24
|
Recouvreux MV, Commisso C. Macropinocytosis: A Metabolic Adaptation to Nutrient Stress in Cancer. Front Endocrinol (Lausanne) 2017; 8:261. [PMID: 29085336 PMCID: PMC5649207 DOI: 10.3389/fendo.2017.00261] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/20/2017] [Indexed: 12/28/2022] Open
Abstract
Oncogenic mutations, such as Ras mutations, drive not only enhanced proliferation but also the metabolic adaptations that confer to cancer cells the ability to sustain cell growth in a harsh tumor microenvironment. These adaptations might represent metabolic vulnerabilities that can be exploited to develop novel and more efficient cancer therapies. Macropinocytosis is an evolutionarily conserved endocytic pathway that permits the internalization of extracellular fluid via large endocytic vesicles known as macropinosomes. Recently, macropinocytosis has been determined to function as a nutrient-scavenging pathway in Ras-driven cancer cells. Macropinocytic uptake of extracellular proteins, and their further degradation within endolysosomes, provides the much-needed amino acids that fuel cancer cell metabolism and tumor growth. Here, we review the molecular mechanisms that govern the process of macropinocytosis, as well as discuss recent work that provides evidence of the important role of macropinocytosis as a nutrient supply pathway in cancer cells.
Collapse
Affiliation(s)
- Maria Victoria Recouvreux
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
- *Correspondence: Cosimo Commisso,
| |
Collapse
|
25
|
Park JK, Peng H, Katsnelson J, Yang W, Kaplan N, Dong Y, Rappoport JZ, He C, Lavker RM. MicroRNAs-103/107 coordinately regulate macropinocytosis and autophagy. J Cell Biol 2016; 215:667-685. [PMID: 27872138 PMCID: PMC5146999 DOI: 10.1083/jcb.201604032] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/29/2016] [Accepted: 11/01/2016] [Indexed: 11/28/2022] Open
Abstract
The miR-103/107 family is preferentially expressed in the stem cell–enriched limbal epithelium and regulates multiple characteristics associated with stem cells. Park et al. show that miR-103/107 also contribute to limbal epithelial homeostasis by suppressing macropinocytosis and preserving end-stage autophagy. Macropinocytosis, by which cells ingest large amounts of fluid, and autophagy, the lysosome-based catabolic process, involve vesicular biogenesis (early stage) and turnover (end stage). Much is known about early-stage events; however, our understanding of how the end stages of these processes are governed is incomplete. Here we demonstrate that the microRNA-103/107(miR-103/107) family, which is preferentially expressed in the stem cell–enriched limbal epithelium, coordinately regulates aspects of both these activities. Loss of miR-103/107 causes dysregulation of macropinocytosis with the formation of large vacuoles, primarily through up-regulation of Src, Ras, and Ankfy1. Vacuole accumulation is not a malfunction of early-stage autophagy; rather, miR-103/107 ensure proper end-stage autophagy by regulating diacylglycerol/protein kinase C and cyclin-dependent kinase 5 signaling, which enables dynamin to function in vacuole clearance. Our findings unveil a key biological function for miR-103/107 in coordinately suppressing macropinocytosis and preserving end-stage autophagy, thereby contributing to maintenance of a stem cell–enriched epithelium.
Collapse
Affiliation(s)
- Jong Kook Park
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Han Peng
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | | | - Wending Yang
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Nihal Kaplan
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| | - Ying Dong
- Department of Dermatology, Northwestern University, Chicago, IL 60611.,Department of Ophthalmology, The First Affiliated Hospital, Chinese PLA General Hospital, Beijing 100048, China
| | - Joshua Z Rappoport
- Center for Advanced Microscopy and Nikon Imaging Center, Northwestern University, Chicago, IL 60611
| | - CongCong He
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Robert M Lavker
- Department of Dermatology, Northwestern University, Chicago, IL 60611
| |
Collapse
|
26
|
Bretou M, Kumari A, Malbec O, Moreau HD, Obino D, Pierobon P, Randrian V, Sáez PJ, Lennon-Duménil AM. Dynamics of the membrane-cytoskeleton interface in MHC class II-restricted antigen presentation. Immunol Rev 2016; 272:39-51. [DOI: 10.1111/imr.12429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Marine Bretou
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Anita Kumari
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Odile Malbec
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Hélène D. Moreau
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Dorian Obino
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Paolo Pierobon
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Violaine Randrian
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Pablo J. Sáez
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | | |
Collapse
|
27
|
Yoshida S, Pacitto R, Yao Y, Inoki K, Swanson JA. Growth factor signaling to mTORC1 by amino acid-laden macropinosomes. J Biophys Biochem Cytol 2015; 211:159-72. [PMID: 26438830 PMCID: PMC4602043 DOI: 10.1083/jcb.201504097] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022] Open
Abstract
The rapid activation of the mechanistic target of rapamycin complex-1 (mTORC1) by growth factors is increased by extracellular amino acids through yet-undefined mechanisms of amino acid transfer into endolysosomes. Because the endocytic process of macropinocytosis concentrates extracellular solutes into endolysosomes and is increased in cells stimulated by growth factors or tumor-promoting phorbol esters, we analyzed its role in amino acid-dependent activation of mTORC1. Here, we show that growth factor-dependent activation of mTORC1 by amino acids, but not glucose, requires macropinocytosis. In murine bone marrow-derived macrophages and murine embryonic fibroblasts stimulated with their cognate growth factors or with phorbol myristate acetate, activation of mTORC1 required an Akt-independent vesicular pathway of amino acid delivery into endolysosomes, mediated by the actin cytoskeleton. Macropinocytosis delivered small, fluorescent fluid-phase solutes into endolysosomes sufficiently fast to explain growth factor-mediated signaling by amino acids. Therefore, the amino acid-laden macropinosome is an essential and discrete unit of growth factor receptor signaling to mTORC1.
Collapse
Affiliation(s)
- Sei Yoshida
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Regina Pacitto
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yao Yao
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Ken Inoki
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Joel A. Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
28
|
Bloomfield G, Traynor D, Sander SP, Veltman DM, Pachebat JA, Kay RR. Neurofibromin controls macropinocytosis and phagocytosis in Dictyostelium. eLife 2015; 4. [PMID: 25815683 PMCID: PMC4374526 DOI: 10.7554/elife.04940] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
Cells use phagocytosis and macropinocytosis to internalise bulk material, which in phagotrophic organisms supplies the nutrients necessary for growth. Wildtype Dictyostelium amoebae feed on bacteria, but for decades laboratory work has relied on axenic mutants that can also grow on liquid media. We used forward genetics to identify the causative gene underlying this phenotype. This gene encodes the RasGAP Neurofibromin (NF1). Loss of NF1 enables axenic growth by increasing fluid uptake. Mutants form outsized macropinosomes which are promoted by greater Ras and PI3K activity at sites of endocytosis. Relatedly, NF1 mutants can ingest larger-than-normal particles using phagocytosis. An NF1 reporter is recruited to nascent macropinosomes, suggesting that NF1 limits their size by locally inhibiting Ras signalling. Our results link NF1 with macropinocytosis and phagocytosis for the first time, and we propose that NF1 evolved in early phagotrophs to spatially modulate Ras activity, thereby constraining and shaping their feeding structures. DOI:http://dx.doi.org/10.7554/eLife.04940.001 Dictyostelium amoebae are microbes that feed on bacteria living in the soil. They are unusual in that the amoebae can survive and grow in a single-celled form, but when food is scarce, many individual cells can gather together to form a simple multicellular organism. To feed on bacteria, the amoebae use a process called phagocytosis, which starts with the membrane that surrounds the cell growing outwards to completely surround the bacteria. This leads to the bacteria entering the amoeba within a membrane compartment called a vesicle, where they are broken down into small molecules by enzymes. The cells can also take up fluids and dissolved molecules using a similar process called macropinocytosis. With its short and relatively simple lifestyle, Dictyostelium is often used in research to study phagocytosis, cell movement and other processes that are also found in larger organisms. For example, some immune cells in animals use phagocytosis to capture and destroy invading microbes. Most studies using Dictyostelium as a model have used amoebae with genetic mutations that allow them to be grown in liquid cultures in the laboratory without needing to feed on bacteria. The mutations allow the ‘mutant’ amoebae to take up more liquid and dissolved nutrients by macropinocytosis, but it is not known where in the genome these mutations are. Here, Bloomfield et al. used genome sequencing to reveal that these mutations alter a gene that encodes a protein called Neurofibromin. The experiments show that the loss of Neurofibromin increases the amount of fluid taken up by the amoebae through macropinocytosis, and also enables the amoebae to take up larger-than-normal particles during phagocytosis. The experiments suggest that Neurofibromin controls both phagocytosis and macropinocytosis by inhibiting the activity of another protein called Ras. Neurofibromin is found in animals and many other organisms so Bloomfield et al. propose that it is an ancient protein that evolved in early single-celled organisms to control the size and shape of their feeding structures. In humans, mutations in the gene that encodes the Neurofibromin protein can lead to the development of a severe disorder—called Neurofibromatosis type 1—in which tumours form in the nervous system. Given that tumour cells can use phagocytosis and macropinocytosis to gain nutrients as they grow, understanding how this protein works in the Dictyostelium amoebae may help to inform future efforts to develop treatments for this human disease. DOI:http://dx.doi.org/10.7554/eLife.04940.002
Collapse
Affiliation(s)
| | - David Traynor
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Sophia P Sander
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Douwe M Veltman
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Justin A Pachebat
- Department of Plant Sciences, University of Cambridge, Cambridge, United Kingdom
| | - Robert R Kay
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
29
|
Ha KD, Bidlingmaier SM, Zhang Y, Su Y, Liu B. High-content analysis of antibody phage-display library selection outputs identifies tumor selective macropinocytosis-dependent rapidly internalizing antibodies. Mol Cell Proteomics 2014; 13:3320-31. [PMID: 25149096 PMCID: PMC4256486 DOI: 10.1074/mcp.m114.039768] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Many forms of antibody-based targeted therapeutics, including antibody drug conjugates, utilize the internalizing function of the targeting antibody to gain intracellular entry into tumor cells. Ideal antibodies for developing such therapeutics should be capable of both tumor-selective binding and efficient endocytosis. The macropinocytosis pathway is capable of both rapid and bulk endocytosis, and recent studies have demonstrated that it is selectively up-regulated by cancer cells. We hypothesize that receptor-dependent macropinocytosis can be achieved using tumor-targeting antibodies that internalize via the macropinocytosis pathway, improving potency and selectivity of the antibody-based targeted therapeutic. Although phage antibody display libraries have been utilized to find antibodies that bind and internalize to target cells, no methods have been described to screen for antibodies that internalize specifically via macropinocytosis. We hereby describe a novel screening strategy to identify phage antibodies that bind and rapidly enter tumor cells via macropinocytosis. We utilized an automated microscopic imaging-based, High Content Analysis platform to identify novel internalizing phage antibodies that colocalize with macropinocytic markers from antibody libraries that we have generated previously by laser capture microdissection-based selection, which are enriched for internalizing antibodies binding to tumor cells in situ residing in their tissue microenvironment (Ruan, W., Sassoon, A., An, F., Simko, J. P., and Liu, B. (2006) Identification of clinically significant tumor antigens by selecting phage antibody library on tumor cells in situ using laser capture microdissection. Mol. Cell. Proteomics. 5, 2364–2373). Full-length human IgG molecules derived from macropinocytosing phage antibodies retained the ability to internalize via macropinocytosis, validating our screening strategy. The target antigen for a cross-species binding antibody with a highly active macropinocytosis activity was identified as ephrin type-A receptor 2. Antibody-toxin conjugates created using this macropinocytosing IgG were capable of potent and receptor-dependent killing of a panel of EphA2-positive tumor cell lines in vitro. These studies identify novel methods to screen for and validate antibodies capable of receptor-dependent macropinocytosis, allowing further exploration of this highly efficient and tumor-selective internalization pathway for targeted therapy development.
Collapse
Affiliation(s)
- Kevin D Ha
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Scott M Bidlingmaier
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Yafeng Zhang
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Yang Su
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| | - Bin Liu
- From the ‡Department of Anesthesia, UCSF Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94110-1305
| |
Collapse
|
30
|
Wang JTH, Teasdale RD, Liebl D. Macropinosome quantitation assay. MethodsX 2014; 1:36-41. [PMID: 26150932 PMCID: PMC4472846 DOI: 10.1016/j.mex.2014.05.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 05/19/2014] [Indexed: 02/02/2023] Open
Abstract
In contrast to phagocytosis, macropinocytosis is not directly initiated by interactions between cell surface receptors and cargo ligands, but is a result of constitutive membrane ruffling driven by dynamic remodelling of cortical actin cytoskeleton in response to stimulation of growth factor receptors. Wang et al. (2010) [13] developed a reliable assay that allows quantitative assessment of the efficiency and kinetics of macropinosome biogenesis and/or maturation in cells where the function of a targeted protein has been perturbed by pharmacological inhibitors or by knock-down or knock-out approaches. In this manuscript we describe a modified quantitative protocol to measure the rate and volume of fluid phase uptake in adherent cells. This assay:uses fluorescent dextran, microscopy and semi-automated image analysis; allows quantitation of macropinosomes within large numbers of individual cells; can be applied also to non-homogenous cell populations including transiently transfected cell monolayers. We present the background necessary to consider when customising this protocol for application to new cell types or experimental variations.
Collapse
Affiliation(s)
- Jack T H Wang
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane 4072, Queensland, Australia ; School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Rohan D Teasdale
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane 4072, Queensland, Australia
| | - David Liebl
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane 4072, Queensland, Australia ; Dept. Bacterial Pathogenesis and Cellular Responses, Institute of Sciences Research and Technologies/CEA-Grenoble, Grenoble, France
| |
Collapse
|
31
|
Tisdale EJ, Shisheva A, Artalejo CR. Overexpression of atypical protein kinase C in HeLa cells facilitates macropinocytosis via Src activation. Cell Signal 2014; 26:1235-42. [PMID: 24582589 PMCID: PMC4149413 DOI: 10.1016/j.cellsig.2014.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/18/2014] [Indexed: 01/08/2023]
Abstract
Atypical protein kinase C (aPKC) is the first recognized kinase oncogene. However, the specific contribution of aPKC to cancer progression is unclear. The pseudosubstrate domain of aPKC is different from the other PKC family members, and therefore a synthetic peptide corresponding to the aPKC pseudosubstrate (aPKC-PS) sequence, which specifically blocks aPKC kinase activity, is a valuable tool to assess the role of aPKC in various cellular processes. Here, we learned that HeLa cells incubated with membrane permeable aPKC-PS peptide displayed dilated heterogeneous vesicles labeled with peptide that were subsequently identified as macropinosomes. A quantitative membrane binding assay revealed that aPKC-PS peptide stimulated aPKC recruitment to membranes and activated Src. Similarly, aPKC overexpression in transfected HeLa cells activated Src and induced macropinosome formation. Src-aPKC interaction was essential; substitution of the proline residues in aPKC that associate with the Src-SH3 binding domain rendered the mutant kinase unable to induce macropinocytosis in transfected cells. We propose that aPKC overexpression is a contributing factor to cell transformation by interacting with and consequently promoting Src activation and constitutive macropinocytosis, which increases uptake of extracellular factors, required for altered cell growth and accelerated cell migration.
Collapse
Affiliation(s)
- Ellen J Tisdale
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield Ave., 6374 Scott Hall, Detroit, MI 48201, USA.
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, 540 E. Canfield Ave., 5374 Scott Hall, Detroit, MI 48201, USA
| | - Cristina R Artalejo
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield Ave., 6374 Scott Hall, Detroit, MI 48201, USA
| |
Collapse
|
32
|
Commisso C, Flinn RJ, Bar-Sagi D. Determining the macropinocytic index of cells through a quantitative image-based assay. Nat Protoc 2014; 9:182-92. [PMID: 24385148 DOI: 10.1038/nprot.2014.004] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macropinocytosis serves as an internalization pathway for extracellular fluid and its contents. Macropinocytosis is upregulated in oncogene-expressing cells and, recently, we have revealed a functional role for macropinocytosis in fueling cancer cell growth through the internalization of extracellular albumin, which is degraded into a usable source of intracellular amino acids. Assessing macropinocytosis has been challenging in the past because of the lack of reliable assays capable of quantitatively measuring this uptake mechanism. Here we describe a protocol for visualizing and quantifying the extent of macropinocytosis in cells both in culture and growing in vivo as tumor xenografts. By using this approach, the 'macropinocytic index' of a particular cell line or subcutaneous tumor can be ascertained within 1-2 d. The protocol can be carried out with multiple samples in parallel and can be easily adapted for a variety of cell types and xenograft or allograft mouse models.
Collapse
Affiliation(s)
- Cosimo Commisso
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, USA
| | - Rory J Flinn
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, USA
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
33
|
Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 2013; 497:633-7. [PMID: 23665962 PMCID: PMC3810415 DOI: 10.1038/nature12138] [Citation(s) in RCA: 1271] [Impact Index Per Article: 105.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 04/02/2013] [Indexed: 12/22/2022]
Abstract
Macropinocytosis is a highly conserved endocytic process by which extracellular fluid and its contents are internalized into cells through large, heterogeneous vesicles known as macropinosomes. Oncogenic Ras proteins have been shown to stimulate macropinocytosis but the functional contribution of this uptake mechanism to the transformed phenotype remains unknown. Here we show that Ras-transformed cells use macropinocytosis to transport extracellular protein into the cell. The internalized protein undergoes proteolytic degradation, yielding amino acids including glutamine that can enter central carbon metabolism. Accordingly, the dependence of Ras-transformed cells on free extracellular glutamine for growth can be suppressed by the macropinocytic uptake of protein. Consistent with macropinocytosis representing an important route of nutrient uptake in tumours, its pharmacological inhibition compromises the growth of Ras-transformed pancreatic tumour xenografts. These results identify macropinocytosis as a mechanism by which cancer cells support their unique metabolic needs and point to the possible exploitation of this process in the design of anticancer therapies.
Collapse
|
34
|
Barthwal MK, Anzinger JJ, Xu Q, Bohnacker T, Wymann MP, Kruth HS. Fluid-phase pinocytosis of native low density lipoprotein promotes murine M-CSF differentiated macrophage foam cell formation. PLoS One 2013; 8:e58054. [PMID: 23536783 PMCID: PMC3594233 DOI: 10.1371/journal.pone.0058054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 01/31/2013] [Indexed: 12/21/2022] Open
Abstract
During atherosclerosis, low-density lipoprotein (LDL)-derived cholesterol accumulates in macrophages to form foam cells. Macrophage uptake of LDL promotes foam cell formation but the mechanism mediating this process is not clear. The present study investigates the mechanism of LDL uptake for macrophage colony-stimulating factor (M-CSF)-differentiated murine bone marrow-derived macrophages. LDL receptor-null (LDLR−/−) macrophages incubated with LDL showed non-saturable accumulation of cholesterol that did not down-regulate for the 24 h examined. Incubation of LDLR−/− macrophages with increasing concentrations of 125I-LDL showed non-saturable macrophage LDL uptake. A 20-fold excess of unlabeled LDL had no effect on 125I-LDL uptake by wild-type macrophages and genetic deletion of the macrophage scavenger receptors CD36 and SRA did not affect 125I-LDL uptake, showing that LDL uptake occurred by fluid-phase pinocytosis independently of receptors. Cholesterol accumulation was inhibited approximately 50% in wild-type and LDLR−/− mice treated with LY294002 or wortmannin, inhibitors of all classes of phosphoinositide 3-kinases (PI3K). Time-lapse, phase-contrast microscopy showed that macropinocytosis, an important fluid-phase uptake pathway in macrophages, was blocked almost completely by PI3K inhibition with wortmannin. Pharmacological inhibition of the class I PI3K isoforms alpha, beta, gamma or delta did not affect macrophage LDL-derived cholesterol accumulation or macropinocytosis. Furthermore, macrophages from mice expressing kinase-dead class I PI3K beta, gamma or delta isoforms showed no decrease in cholesterol accumulation or macropinocytosis when compared with wild-type macrophages. Thus, non-class I PI3K isoforms mediated macropinocytosis in these macrophages. Further characterization of the components necessary for LDL uptake, cholesterol accumulation, and macropinocytosis identified dynamin, microtubules, actin, and vacuolar type H(+)-ATPase as contributing to uptake. However, Pak1, Rac1, and Src-family kinases, which mediate fluid-phase pinocytosis in certain other cell types, were unnecessary. In conclusion, our findings provide evidence that targeting those components mediating macrophage macropinocytosis with inhibitors may be an effective strategy to limit macrophage accumulation of LDL-derived cholesterol in arteries.
Collapse
Affiliation(s)
- Manoj K. Barthwal
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joshua J. Anzinger
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Qing Xu
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas Bohnacker
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Howard S. Kruth
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
35
|
Poillot C, Bichraoui H, Tisseyre C, Bahemberae E, Andreotti N, Sabatier JM, Ronjat M, De Waard M. Small efficient cell-penetrating peptides derived from scorpion toxin maurocalcine. J Biol Chem 2012; 287:17331-17342. [PMID: 22433862 DOI: 10.1074/jbc.m112.360628] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Maurocalcine is the first demonstrated example of an animal toxin peptide with efficient cell penetration properties. Although it is a highly competitive cell-penetrating peptide (CPP), its relatively large size of 33 amino acids and the presence of three internal disulfide bridges may hamper its development for in vitro and in vivo applications. Here, we demonstrate that several efficient CPPs can be derived from maurocalcine by replacing Cys residues by isosteric 2-aminobutyric acid residues and sequence truncation down to peptides of up to 9 residues in length. A surprising finding is that all of the truncated maurocalcine analogues possessed cell penetration properties, indicating that the maurocalcine is a highly specialized CPP. Careful examination of the cell penetration properties of the truncated analogues indicates that several maurocalcine-derived peptides should be of great interest for cell delivery applications where peptide size matters.
Collapse
Affiliation(s)
- Cathy Poillot
- INSERM U836, Grenoble Neuroscience Institute, Site Santé La Tronche, Chemin Fortuné Ferrini, BP 170, 38042 Grenoble Cedex 9, France; Université Joseph Fourier, 38041 Grenoble, France
| | - Hicham Bichraoui
- INSERM U836, Grenoble Neuroscience Institute, Site Santé La Tronche, Chemin Fortuné Ferrini, BP 170, 38042 Grenoble Cedex 9, France; Université Joseph Fourier, 38041 Grenoble, France
| | - Céline Tisseyre
- INSERM U836, Grenoble Neuroscience Institute, Site Santé La Tronche, Chemin Fortuné Ferrini, BP 170, 38042 Grenoble Cedex 9, France; Université Joseph Fourier, 38041 Grenoble, France
| | - Eloi Bahemberae
- INSERM U836, Grenoble Neuroscience Institute, Site Santé La Tronche, Chemin Fortuné Ferrini, BP 170, 38042 Grenoble Cedex 9, France; Université Joseph Fourier, 38041 Grenoble, France
| | | | | | - Michel Ronjat
- INSERM U836, Grenoble Neuroscience Institute, Site Santé La Tronche, Chemin Fortuné Ferrini, BP 170, 38042 Grenoble Cedex 9, France; Université Joseph Fourier, 38041 Grenoble, France
| | - Michel De Waard
- INSERM U836, Grenoble Neuroscience Institute, Site Santé La Tronche, Chemin Fortuné Ferrini, BP 170, 38042 Grenoble Cedex 9, France; Université Joseph Fourier, 38041 Grenoble, France; Smartox Biotechnologies, Biopolis, 5 Avenue du Grand Sablon, 38700 La Tronche, France.
| |
Collapse
|
36
|
Grieve AG, Moss SE, Hayes MJ. Annexin A2 at the interface of actin and membrane dynamics: a focus on its roles in endocytosis and cell polarization. Int J Cell Biol 2012; 2012:852430. [PMID: 22505935 PMCID: PMC3296266 DOI: 10.1155/2012/852430] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 11/16/2011] [Accepted: 12/13/2011] [Indexed: 12/15/2022] Open
Abstract
Annexins are a family of calcium- and phospholipid-binding proteins found in nearly all eukaryotes. They are structurally highly conserved and have been implicated in a wide range of cellular activities. In this paper, we focus on Annexin A2 (AnxA2). Altered expression of this protein has been identified in a wide variety of cancers, has also been found on the HIV particle, and has been implicated in the maturation of the virus. Recently, it has also been shown to have an important role in the establishment of normal apical polarity in epithelial cells. We synthesize here the known biochemical properties of this protein and the extensive literature concerning its involvement in the endocytic pathway. We stress the importance of AnxA2 as a platform for actin remodeling in the vicinity of dynamic cellular membranes, in the hope that this may shed light on the normal functions of the protein and its contribution to disease.
Collapse
Affiliation(s)
- Adam G. Grieve
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - Stephen E. Moss
- Division of Cell Biology, UCL Institute of Ophthalmology, 11-43 Bath Street, EC1V 9EL London, UK
| | - Matthew J. Hayes
- Division of Cell Biology, UCL Institute of Ophthalmology, 11-43 Bath Street, EC1V 9EL London, UK
| |
Collapse
|
37
|
Raoof M, Mackeyev Y, Cheney MA, Wilson LJ, Curley SA. Internalization of C60 fullerenes into cancer cells with accumulation in the nucleus via the nuclear pore complex. Biomaterials 2012; 33:2952-60. [PMID: 22245558 DOI: 10.1016/j.biomaterials.2011.12.043] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 12/26/2011] [Indexed: 01/18/2023]
Abstract
A highly water-soluble, non-ionic, and non-cytotoxic fullerene malonodiserinolamide-derivatized fullerene C(60) (C(60)-ser) is under investigation as a potential nanovector to deliver biologic and cancer drugs across biological barriers. Using laser-scanning confocal microscopy and flow cytometry, we find that PF-633 fluorophore conjugated C(60)-ser nanoparticles (C(60)-serPF) are internalized within living cancer cells in association with serum proteins through multiple energy-dependent pathways, and escape endocytotic vesicles to eventually localize and accumulate in the nucleus of the cells through the nuclear pore complex. Furthermore, in a mouse model of liver cancer, the C(60)-serPF conjugate is detected in most tissues, permeating through the altered vasculature of the tumor and the tightly-regulated blood brain barrier while evading the reticulo-endothelial system.
Collapse
Affiliation(s)
- Mustafa Raoof
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230-1402, USA.
| | | | | | | | | |
Collapse
|
38
|
Anzinger JJ, Chang J, Xu Q, Barthwal MK, Bohnacker T, Wymann MP, Kruth HS. Murine bone marrow-derived macrophages differentiated with GM-CSF become foam cells by PI3Kγ-dependent fluid-phase pinocytosis of native LDL. J Lipid Res 2011; 53:34-42. [PMID: 22058424 DOI: 10.1194/jlr.m018887] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Accumulation of cholesterol by macrophage uptake of LDL is a key event in the formation of atherosclerotic plaques. Previous research has shown that granulocyte-macrophage colony-stimulating factor (GM-CSF) is present in atherosclerotic plaques and promotes aortic lipid accumulation. However, it has not been determined whether murine GM-CSF-differentiated macrophages take up LDL to become foam cells. GM-CSF-differentiated macrophages from LDL receptor-null mice were incubated with LDL, resulting in massive macrophage cholesterol accumulation. Incubation of LDL receptor-null or wild-type macrophages with increasing concentrations of ¹²⁵I-LDL showed nonsaturable macrophage LDL uptake that was linearly related to the amount of LDL added, indicating that LDL uptake was mediated by fluid-phase pinocytosis. Previous studies suggest that phosphoinositide 3-kinases (PI3K) mediate macrophage fluid-phase pinocytosis, although the isoform mediating this process has not been determined. Because PI3Kγ is known to promote aortic lipid accumulation, we investigated its role in mediating macrophage fluid-phase pinocytosis of LDL. Wild-type macrophages incubated with LDL and the PI3Kγ inhibitor AS605240 or PI3Kγ-null macrophages incubated with LDL showed an ∼50% reduction in LDL uptake and cholesterol accumulation compared with wild-type macrophages incubated with LDL only. These results show that GM-CSF-differentiated murine macrophages become foam cells by fluid-phase pinocytosis of LDL and identify PI3Kγ as contributing to this process.
Collapse
Affiliation(s)
- Joshua J Anzinger
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Macropinocytosis is a regulated form of endocytosis that mediates the non-selective uptake of solute molecules, nutrients and antigens. It is an actin-dependent process initiated from surface membrane ruffles that give rise to large endocytic vacuoles called macropinosomes. Macropinocytosis is important in a range of physiological processes; it is highly active in macrophages and dendritic cells where it is a major pathway for the capture of antigens, it is relevant to cell migration and tumour metastasis and it represents a portal of cell entry exploited by a range of pathogens. The molecular basis for the formation and maturation of macropinosomes has only recently begun to be defined. Here, we review the general characteristics of macropinocytosis, describe some of the regulators of this pathway, which have been identified to date and highlight strategies to explore the relevance of this endocytosis pathway in vivo.
Collapse
|
40
|
Acute ligand-independent Src activation mimics low EGF-induced EGFR surface signalling and redistribution into recycling endosomes. Exp Cell Res 2010; 316:3239-53. [PMID: 20832399 DOI: 10.1016/j.yexcr.2010.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 08/12/2010] [Accepted: 09/02/2010] [Indexed: 01/09/2023]
Abstract
Src, a non-receptor tyrosine kinase, is a key signal transduction partner of epidermal growth factor (EGF) receptor (EGFR). In human breast cancer, EGFR and Src are frequently over-expressed and/or over-activated. Although reciprocal activation is documented, mechanisms underlying Src:EGFR interactions are incompletely understood. We here exploited ts/v-Src thermo-activation in MDCK monolayers to test whether acute Src activation impacts on signalling and trafficking of non-liganded EGFR. We found that thermo-activation caused rapid Src recruitment to the plasma membrane, concomitant association with EGFR, and its phosphorylation at Y845 and Y1173 predominantly at the cell surface. Like low EGF concentrations, activated Src (i) decreased EGF surface binding without affecting the total EGFR pool; (ii) triggered EGFR endocytosis via clathrin-coated vesicles; (iii) and led to its sequestration in perinuclear/recycling endosomes with avoidance of multivesicular bodies and lysosomal degradation. Combined Src activation and EGF were synergistic for EGFR-Y845 and -Y1173 phosphorylation at some endosomes. We conclude that acute effects of Src in MDCK cells may mimic those of low EGF on EGFR activation and redistribution. Src:EGFR interactions may be sufficient to trigger EGFR activation and might contribute to its local signalling, without requiring either soluble extracellular signal or receptor over-expression.
Collapse
|
41
|
Koivusalo M, Welch C, Hayashi H, Scott CC, Kim M, Alexander T, Touret N, Hahn KM, Grinstein S. Amiloride inhibits macropinocytosis by lowering submembranous pH and preventing Rac1 and Cdc42 signaling. ACTA ACUST UNITED AC 2010; 188:547-63. [PMID: 20156964 PMCID: PMC2828922 DOI: 10.1083/jcb.200908086] [Citation(s) in RCA: 702] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Inhibitors of Na+/H+ exchange proteins block macropinocytosis by lowering the pH near the plasma membrane, which in turn inhibits actin remodeling by Rho family GTPases. Macropinocytosis is differentiated from other types of endocytosis by its unique susceptibility to inhibitors of Na+/H+ exchange. Yet, the functional relationship between Na+/H+ exchange and macropinosome formation remains obscure. In A431 cells, stimulation by EGF simultaneously activated macropinocytosis and Na+/H+ exchange, elevating cytosolic pH and stimulating Na+ influx. Remarkably, although inhibition of Na+/H+ exchange by amiloride or HOE-694 obliterated macropinocytosis, neither cytosolic alkalinization nor Na+ influx were required. Instead, using novel probes of submembranous pH, we detected the accumulation of metabolically generated acid at sites of macropinocytosis, an effect counteracted by Na+/H+ exchange and greatly magnified when amiloride or HOE-694 were present. The acidification observed in the presence of the inhibitors did not alter receptor engagement or phosphorylation, nor did it significantly depress phosphatidylinositol-3-kinase stimulation. However, activation of the GTPases that promote actin remodelling was found to be exquisitely sensitive to the submembranous pH. This sensitivity confers to macropinocytosis its unique susceptibility to inhibitors of Na+/H+ exchange.
Collapse
Affiliation(s)
- Mirkka Koivusalo
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Duterme C, Mertens-Strijthagen J, Tammi M, Flamion B. Two novel functions of hyaluronidase-2 (Hyal2) are formation of the glycocalyx and control of CD44-ERM interactions. J Biol Chem 2009; 284:33495-508. [PMID: 19783662 DOI: 10.1074/jbc.m109.044362] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It has long been predicted that the members of the hyaluronidase enzyme family have important non-enzymatic functions. However, their nature remains a mystery. The metabolism of hyaluronan (HA), their major enzymatic substrate, is also enigmatic. To examine the function of Hyal2, a glycosylphosphatidylinositol-anchored hyaluronidase with intrinsically weak enzymatic activity, we have compared stably transfected rat fibroblastic BB16 cell lines with various levels of expression of Hyal2. These cell lines continue to express exclusively the standard form (CD44s) of the main HA receptor, CD44. Hyal2, CD44, and one of its main intracellular partners, ezrin-radixin-moesin (ERM), were found to co-immunoprecipitate. Functionally, Hyal2 overexpression was linked to loss of the glycocalyx, the HA-rich pericellular coat. This effect could be mimicked by exposure of BB16 cells either to Streptomyces hyaluronidase, to HA synthesis inhibitors, or to HA oligosaccharides. This led to shedding of CD44, separation of CD44 from ERM, reduction in baseline level of ERM activation, and markedly decreased cell motility (50% reduction in a wound healing assay). The effects of Hyal2 on the pericellular coat and on CD44-ERM interactions were inhibited by treatment with the Na(+)/H(+) exchanger-1 inhibitor ethyl-N-isopropylamiloride. We surmise that Hyal2, through direct interactions with CD44 and possibly some pericellular hyaluronidase activity requiring acidic foci, suppresses the formation or the stability of the glycocalyx, modulates ERM-related cytoskeletal interactions, and diminishes cell motility. These effects may be relevant to the purported in vivo tumor-suppressive activity of Hyal2.
Collapse
Affiliation(s)
- Cecile Duterme
- Molecular Physiology Research Unit, University of Namur, 5000 Namur, Belgium
| | | | | | | |
Collapse
|
43
|
Kaposi's sarcoma-associated herpesvirus utilizes an actin polymerization-dependent macropinocytic pathway to enter human dermal microvascular endothelial and human umbilical vein endothelial cells. J Virol 2009; 83:4895-911. [PMID: 19279100 DOI: 10.1128/jvi.02498-08] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) utilizes clathrin-mediated endocytosis for its infectious entry into human foreskin fibroblast (HFF) cells (S. M. Akula, P. P. Naranatt, N.-S. Walia, F.-Z. Wang, B. Fegley, and B. Chandran, J. Virol. 77:7978-7990, 2003). Here, we characterized KSHV entry into primary human microvascular dermal endothelial (HMVEC-d) and human umbilical vein endothelial (HUVEC) cells. Similar to the results for HMVEC-d cells, KSHV infection of HUVEC cells also resulted in an initial high level and subsequent decline in the expression of the lytic switch gene, ORF50, while latent gene expression persisted. Internalized virus particles enclosed in irregular vesicles were observed by electron microscopy of infected HMVEC-d cells. At an early time of infection, colocalization of KSHV capsid with envelope was observed by immunofluorescence analysis, thus demonstrating endocytosis of intact enveloped virus particles. Chlorpromazine, an inhibitor of clathrin-mediated endocytosis, and filipin (C(35)H(58)O(11)), a caveolar endocytosis inhibitor, did not have any effect on KSHV binding, entry (DNA internalization), or gene expression in HMVEC-d and HUVEC cells. In contrast to the results for HFF cells, virus entry and gene expression in both types of endothelial cells were significantly blocked by macropinocytosis inhibitors (EIPA [5-N-ethyl-N-isoproamiloride] and rottlerin [C(30)H(28)O(8)]) and by cytochalasin D, which affects actin polymerization. Inhibition of lipid raft blocked viral gene expression in HMVEC-d cells but not in HUVEC or HFF cells. In HMVEC-d and HUVEC cells, KSHV induced the actin polymerization and formation of lamellipodial extensions that are essential for macropinocytosis. Inhibition of macropinocytosis resulted in the distribution of viral capsids at the HMVEC-d cell periphery, and capsids did not associate with microtubules involved in the nuclear delivery of viral DNA. Internalized KSHV in HMVEC-d and HUVEC cells colocalized with the macropinocytosis marker dextran and not with the clathrin pathway marker transferrin or with caveolin. Dynasore, an inhibitor of dynamin, did not block viral entry into endothelial cells but did inhibit entry into HFF cells. KSHV was not associated with the early endosome marker EEA-1 in HMVEC-d cells, but rather with the late endosome marker LAMP1, as well as with Rab34 GTPase that is known to regulate macropinocytosis. Silencing Rab34 with small interfering RNA dramatically inhibited KSHV gene expression. Bafilomycin-mediated disruption of endosomal acidification inhibited viral gene expression. Taken together, these findings suggest that KSHV utilizes the actin polymerization-dependent, dynamin-independent macropinocytic pathway that involves a Rab34 GTPase-dependent late endosome and low-pH environment for its infectious entry into HMVEC-d and HUVEC cells. These studies also demonstrate that KSHV utilizes different modes of endocytic entry in fibroblast and endothelial cells.
Collapse
|
44
|
Abstract
Endocytic mechanisms control the lipid and protein composition of the plasma membrane, thereby regulating how cells interact with their environments. Here, we review what is known about mammalian endocytic mechanisms, with focus on the cellular proteins that control these events. We discuss the well-studied clathrin-mediated endocytic mechanisms and dissect endocytic pathways that proceed independently of clathrin. These clathrin-independent pathways include the CLIC/GEEC endocytic pathway, arf6-dependent endocytosis, flotillin-dependent endocytosis, macropinocytosis, circular doral ruffles, phagocytosis, and trans-endocytosis. We also critically review the role of caveolae and caveolin1 in endocytosis. We highlight the roles of lipids, membrane curvature-modulating proteins, small G proteins, actin, and dynamin in endocytic pathways. We discuss the functional relevance of distinct endocytic pathways and emphasize the importance of studying these pathways to understand human disease processes.
Collapse
Affiliation(s)
- Gary J Doherty
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.
| | | |
Collapse
|
45
|
Donepudi M, Resh MD. c-Src trafficking and co-localization with the EGF receptor promotes EGF ligand-independent EGF receptor activation and signaling. Cell Signal 2008; 20:1359-67. [PMID: 18448311 PMCID: PMC2459337 DOI: 10.1016/j.cellsig.2008.03.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 03/10/2008] [Accepted: 03/10/2008] [Indexed: 01/01/2023]
Abstract
c-Src is a non-receptor tyrosine kinase that associates with both the plasma membrane and endosomal compartments. In many human cancers, especially breast cancer, c-Src and the EGF receptor (EGFR) are overexpressed. Dual overexpression of c-Src and EGFR correlates with a Src-dependent increase in activation of EGFR, and synergism between these two tyrosine kinases increases the mitogenic activity of EGFR. Despite extensive studies of the functional interaction between c-Src and EGFR, little is known about the interactions in the trafficking pathways for the two proteins and how that influences signaling. Given the synergism between c-Src and EGFR, and the finding that EGFR is internalized and can signal from endosomes, we hypothesized that c-Src and EGFR traffic together through the endocytic pathway. Here we use a regulatable c-SrcGFP fusion protein that is a bona fide marker for c-Src to show that c-Src undergoes constitutive macropinocytosis from the plasma membrane into endocytic compartments. The movement of c-Src was dependent on its tyrosine kinase activity. Stimulation of cells with EGF revealed that c-Src traffics into the cell with activated EGFR and that c-Src expression and kinase activity prolongs EGFR activation. Surprisingly, even in the absence of EGF addition, c-Src expression induced activation of EGFR and of EGFR-mediated downstream signaling targets ERK and Shc. These data suggest that the synergy between c-Src and EGFR also occurs as these two kinases traffic together, and that their co-localization promotes EGFR-mediated signaling.
Collapse
Affiliation(s)
| | - Marilyn D. Resh
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
46
|
Porat-Shliom N, Kloog Y, Donaldson JG. A unique platform for H-Ras signaling involving clathrin-independent endocytosis. Mol Biol Cell 2008; 19:765-75. [PMID: 18094044 PMCID: PMC2262976 DOI: 10.1091/mbc.e07-08-0841] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 11/05/2007] [Accepted: 12/07/2007] [Indexed: 11/11/2022] Open
Abstract
Trafficking of H-Ras was examined to determine whether it can enter cells through clathrin-independent endocytosis (CIE). H-Ras colocalized with the CIE cargo protein, class I major histocompatibility complex, and it was sequestered in vacuoles that formed upon expression of an active mutant of Arf6, Q67L. Activation of Ras, either through epidermal growth factor stimulation or the expression of an active mutant of Ras, G12V, induced plasma membrane ruffling and macropinocytosis, a stimulated form of CIE. Live imaging of cells expressing H-RasG12V and fluorescent protein chimeras with pleckstrin homology domains that recognize specific phosphoinositides showed that incoming macropinosomes contained phosphatidylinositol 4,5-bisphosphate (PIP(2)) and phosphatiylinositol 3,4,5-trisphosphate (PIP(3)). PIP(2) loss from the macropinosome was followed by the recruitment of Rab5, a downstream target of Ras, and then PIP(3) loss. Our studies support a model whereby Ras can signal on macropinosomes that pass through three distinct stages: PIP(2)/PIP(3), PIP(3)/Rab5, and Rab5. Vacuoles that form in cells expressing Arf6Q67L trap Ras signaling in the first stage, recruiting the active form of the Ras effectors extracellular signal-regulated kinase and protein kinase B (Akt) but not Rab5. Arf6 stimulation of macropinocytosis also involves passage through the distinct lipid phases, but recruitment of Akt is not observed.
Collapse
Affiliation(s)
- Natalie Porat-Shliom
- *Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
- Department of Neurobiochemistry, Tel Aviv University, Tel Aviv, Israel
| | - Yoel Kloog
- Department of Neurobiochemistry, Tel Aviv University, Tel Aviv, Israel
| | - Julie G. Donaldson
- *Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
47
|
Ivanov AI. Pharmacological inhibition of endocytic pathways: is it specific enough to be useful? Methods Mol Biol 2008; 440:15-33. [PMID: 18369934 DOI: 10.1007/978-1-59745-178-9_2] [Citation(s) in RCA: 474] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Eukaryotic cells constantly form and internalize plasma membrane vesicles in a process known as endocytosis. Endocytosis serves a variety of housekeeping and specialized cellular functions, and it can be mediated by distinct molecular pathways. Among them, internalization via clathrin-coated pits, lipid raft/caveolae-mediated endocytosis and macropinocytosis/phagocytosis are the most extensively characterized. The major endocytic pathways are usually distinguished on the basis of their differential sensitivity to pharmacological/chemical inhibitors, although the possibility of nonspecific effects of such inhibitors is frequently overlooked. This review provides a critical evaluation of the selectivity of the most widely used pharmacological inhibitors of clathrin-mediated, lipid raft/caveolae-mediated endocytosis and macropinocytosis/phagocytosis. The mechanisms of actions of these agents are described with special emphasis on their reported side effects on the alternative internalization modes and the actin cytoskeleton. The most and the least-selective inhibitors of each major endocytic pathway are highlighted.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Medicine, Gastroenterology and Hepatology Division, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
48
|
Gao YS, Hubbert CC, Lu J, Lee YS, Lee JY, Yao TP. Histone deacetylase 6 regulates growth factor-induced actin remodeling and endocytosis. Mol Cell Biol 2007; 27:8637-47. [PMID: 17938201 PMCID: PMC2169396 DOI: 10.1128/mcb.00393-07] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 04/15/2007] [Accepted: 09/28/2007] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a cytoplasmic deacetylase that uniquely catalyzes alpha-tubulin deacetylation and promotes cell motility. However, the mechanism underlying HDAC6-dependent cell migration and the role for microtubule acetylation in motility are not known. Here we show that HDAC6-induced global microtubule deacetylation was not sufficient to stimulate cell migration. Unexpectedly, in response to growth factor stimulation, HDAC6 underwent rapid translocation to actin-enriched membrane ruffles and subsequently became associated with macropinosomes, the vesicles for fluid-phase endocytosis. Supporting the importance of these associations, membrane ruffle formation, macropinocytosis, and cell migration were all impaired in HDAC6-deficient cells. Conversely, elevated HDAC6 levels promoted membrane ruffle formation with a concomitant increase in macropinocytosis and motility. In search for an HDAC6 target, we found that heat shock protein 90 (Hsp90), another prominent substrate of HDAC6, was also recruited to membrane ruffles and macropinosomes. Significantly, inhibition of Hsp90 activity suppressed membrane ruffling and cell migration, while expression of an acetylation-resistant Hsp90 mutant promoted ruffle formation. Our results uncover a surprising role for HDAC6 in actin remodeling-dependent processes and identify the actin cytoskeleton as an important target of HDAC6-regulated protein deacetylation.
Collapse
Affiliation(s)
- Ya-sheng Gao
- Duke University, Department of Pharmacology and Cancer Biology, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
49
|
Jones AT. Macropinocytosis: searching for an endocytic identity and role in the uptake of cell penetrating peptides. J Cell Mol Med 2007; 11:670-84. [PMID: 17760832 PMCID: PMC3823249 DOI: 10.1111/j.1582-4934.2007.00062.x] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Macropinocytosis defines a series of events initiated by extensive plasma membrane reorganization or ruffling to form an external macropinocytic structure that is then enclosed and internalized. The process is constitutive in some organisms and cell types but in others it is only pronounced after growth factor stimulation. Internalized macropinosomes share many features with phagosomes and both are distinguished from other forms of pinocytic vesicles by their large size, morphological heterogeneity and lack of coat structures. A paucity of information is available on other distinguishing features for macropinocytosis such as specific marker proteins and drugs that interfere with its mechanism over other endocytic processes. This has hampered efforts to characterize the dynamics of this pathway and to identify regulatory proteins that are expressed in order to allow it to proceed. Upon internalization, macropinosomes acquire regulatory proteins common to other endocytic pathways, suggesting that their identities as unique structures are short-lived. There is however less consensus regarding the overall fate of the macropinosome cargo or its limiting membrane and processes such as fusion, tubulation, recycling and regulated exocytosis have all been implicated in shaping the macropinosome and directing cargo traffic. Macropinocytosis has also been implicated in the internalization of cell penetrating peptides that are of significant interest to researchers aiming to utilize their translocation abilities to deliver therapeutic entities such as genes and proteins into cells. This review focuses on recent findings on the regulation of macropinocytosis, the intracellular fate of the macropinosome and discusses evidence for the role of this pathway as a mechanism of entry for cell penetrating peptides.
Collapse
Affiliation(s)
- Arwyn Tomos Jones
- Welsh School of Pharmacy, Redwood Building, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
50
|
Mettlen M, Platek A, Van Der Smissen P, Carpentier S, Amyere M, Lanzetti L, de Diesbach P, Tyteca D, Courtoy PJ. Src triggers circular ruffling and macropinocytosis at the apical surface of polarized MDCK cells. Traffic 2007; 7:589-603. [PMID: 16643281 DOI: 10.1111/j.1600-0854.2006.00412.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We addressed the role of Src on cortical actin dynamics and polarized endocytosis in MDCK cells harboring a thermosensitive v-src mutant. Shifting monolayers established at 40 degrees C (non-permissive temperature) to 34 degrees C (permissive temperature) rapidly reactivated v-Src kinase, but tight junctions and cell polarity resisted for >6 h. At this interval, activated v-src was recruited on apical vesicles, induced cortactin-associated apical circular ruffles productive of macropinosomes, thereby accelerating apical pinocytosis by approximately fivefold. Ruffling and macropinosome formation were selectively abrogated by inhibitors of actin polymerization, phosphoinositide 3-kinase, phospholipase C, and phospholipase D, which all returned apical pinocytosis to the level observed at 40 degrees C, underscoring the distinct control of apical micropinocytosis and macropinocytosis. Src promoted microtubule-dependent fusion of macropinosomes to the apical recycling endosome (ARE), causing its strong vacuolation. However, preservation of tubulation and apical polarity indicated that its function was not affected. The ARE was labeled for v-src, Rab11, and rabankyrin-5 but not early endosome antigen 1, thus distinguishing two separate Rab5-dependent apical pathways. The mechanisms of Src-induced apical ruffling and macropinocytosis could shed light on the triggered apical enteroinvasive pathogens entry and on the apical differentiation of osteoclasts.
Collapse
Affiliation(s)
- Marcel Mettlen
- CELL Unit, Université catholique de Louvain and Christian de Duve Institute of Cellular Pathology, 1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|