1
|
Mao R, Zhou F, Hong Y, Li Y, Zhu C, Jin L, Li S. CRH upregulates supervillin through ERK and AKT pathways to promote bladder cancer cell migration. Cell Biol Int 2024; 48:1743-1754. [PMID: 39090812 DOI: 10.1002/cbin.12227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Corticotropin-releasing hormone (CRH) has been well documented playing a role in the regulation of cellular processes, immune responses, and inflammatory processes that can influence the occurrence and development of tumors. Supervillin (SVIL) is a membrane-associated and actin-binding protein, which is actively involved in the proliferation, spread, and migration of cancer cells. This work investigated CRH's influence on bladder cancer cells' migration and relevant mechanisms. By using human bladder cancer cells T24 and RT4 in wound healing experiments and transwell assay, we found that the migration ability of the T24 cells was significantly increased after CRH treatment. In vivo experiments showed that CRH significantly promoted the metastases of T24 cells in cell line-derived xenograft (CDX) mouse model. Interestingly, downregulation of SVIL by SVIL-specifc small hairpin RNAs significantly reduced the promoting effect of CRH on bladder cancer cell migration. Furthermore, CRH significantly increased SVIL messenger RNA and protein expression in T24 cells, accompanied with AKT and ERK phosphorylation in T24 cells. Pretreatment with AKT inhibitor (MK2206) blocked the CRH-induced SVIL expression and ERK phosphorylation. Also, inhibition of ERK signaling pathway by U0126 significantly reduced the CRH-induced SVIL expression and AKT phosphorylation. It suggested that cross-talking between AKT and ERK pathways was involved in the effect of CRH on SVIL. Taken together, we demonstrated that CRH induced migration of bladder cancer cells, in which AKT and ERK pathways -SVIL played a key role.
Collapse
Affiliation(s)
- Rongchen Mao
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feier Zhou
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yali Hong
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yongqi Li
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Chao Zhu
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lai Jin
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Shengnan Li
- Department of Pharmacology, Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Shahriari S, Ghildyal R. The actin-binding protein palladin associates with the respiratory syncytial virus matrix protein. J Virol 2024; 98:e0143524. [PMID: 39360826 PMCID: PMC11494977 DOI: 10.1128/jvi.01435-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024] Open
Abstract
The respiratory syncytial virus (RSV) matrix (M) protein plays an important role in infection as it can interact with viral components as well as the host cell actin microfilaments. The M-actin interaction may play a role in facilitating the transportation of virion components to the apical surface, where RSV is released. We show that M protein's association with actin is facilitated by palladin, an actin-binding protein. Cells were infected with RSV or transfected to express full-length M as a green fluorescent protein (GFP)-tagged protein, followed by removal of nuclear and cytosolic proteins to enrich for cytoskeleton and its associated proteins. M protein was present in inclusion bodies tethered to microfilaments in infected cells. In transfected cells, GFP-M was presented close to microfilaments, without association, suggesting the possible involvement of an additional protein in this interaction. As palladin can bind to proteins that also bind actin, we investigated its interaction with M. Cells were co-transfected to express GFP-M and palladin as an mCherry fluorescent-tagged protein, followed by cytoskeleton enrichment. M and palladin were observed to colocalize towards microfilaments, suggesting that palladin is involved in the M-actin interaction. In co-immunoprecipitation studies, M was found to associate with two isoforms of palladin, of 140 and 37 kDa. Interestingly, siRNA downregulation of palladin resulted in reduced titer of released RSV, while cell associated RSV titer increased, suggesting a role for palladin in virus release. Together, our data show that the M-actin interaction mediated by palladin is important for RSV budding and release.IMPORTANCERespiratory syncytial virus is responsible for severe lower respiratory tract infections in young children under 5 years old, the elderly, and the immunosuppressed. The interaction of the respiratory syncytial virus matrix protein with the host actin cytoskeleton is important in infection but has not been investigated in depth. In this study, we show that the respiratory syncytial virus matrix protein associates with actin microfilaments and the actin-binding protein palladin, suggesting a role for palladin in respiratory syncytial virus release. This study provides new insight into the role of the actin cytoskeleton in respiratory syncytial virus infection, a key host-RSV interaction in assembly. Understanding the mechanism by which the RSV M protein and actin interact will ultimately provide a basis for the development of therapeutics targeted at RSV infections.
Collapse
Affiliation(s)
- Shadi Shahriari
- Biomedical Research Cluster, Faculty of Science and Technology, University of Canberra, Canberra, Australia
| | - Reena Ghildyal
- Biomedical Research Cluster, Faculty of Science and Technology, University of Canberra, Canberra, Australia
| |
Collapse
|
3
|
Hunek G, Zembala J, Januszewski J, Bełżek A, Syty K, Jabiry-Zieniewicz Z, Ludwin A, Flieger J, Baj J. Micro- and Macronutrients in Endometrial Cancer-From Metallomic Analysis to Improvements in Treatment Strategies. Int J Mol Sci 2024; 25:9918. [PMID: 39337406 PMCID: PMC11432114 DOI: 10.3390/ijms25189918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
Endometrial cancer is reported to be one of the most prevalent cancers of the female reproductive organs worldwide, with increasing incidence and mortality rates over the past decade. Early diagnosis is critical for effective treatment. Recently, there has been a growing focus on the role of nutrition and micronutrient and macronutrient status in patients with gynecologic cancers, including endometrial cancer. In the following paper, we have conducted an in-depth narrative literature review with the aim of evaluating the results of metallomic studies specifically concerning the micro- and macronutrient status of patients with endometrial cancer. The main objective of the paper was to analyze the results regarding the nutritional status of endometrial cancer patients and describe the role of chosen elements in the onset and progression of endometrial carcinogenesis. Further, we have focused on the evaluation of the usage of the described elements in the potential treatment of the abovementioned cancer, as well as the possible prevention of cancer considering proper supplementation of chosen elements in healthy individuals. Calcium supplementation has been proposed to reduce the risk of endometrial cancer, although some studies offer conflicting evidence. Deficiencies in phosphorus, selenium, and zinc have been inversely associated with endometrial cancer risk, suggesting they may play a protective role, whereas excessive levels of iron, copper, and cadmium have been positively correlated with increased risk. However, the molecular mechanisms by which these elements affect endometrial carcinogenesis are not fully understood, and current findings are often contradictory. Further research is needed to clarify these relationships and to evaluate the potential of nutritional interventions for the prevention and treatment of endometrial cancer.
Collapse
Affiliation(s)
- Gabriela Hunek
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Julita Zembala
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Jacek Januszewski
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Aleksandra Bełżek
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | - Kinga Syty
- Institute of Health Sciences, John Paul the II Catholic University of Lublin, Konstantynów 1G, 20-708 Lublin, Poland
| | - Zoulikha Jabiry-Zieniewicz
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Artur Ludwin
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| |
Collapse
|
4
|
Koh JYC, Chen L, Gong L, Tan SJ, Hou HW, Tay CY. Lost in Rotation: How TiO 2 and ZnO Nanoparticles Disrupt Coordinated Epithelial Cell Rotation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312007. [PMID: 38708799 DOI: 10.1002/smll.202312007] [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] [Received: 12/21/2023] [Revised: 04/10/2024] [Indexed: 05/07/2024]
Abstract
Coordinated cell movement is a cardinal feature in tissue organization that highlights the importance of cells working together as a collective unit. Disruptions to this synchronization can have far-reaching pathological consequences, ranging from developmental disorders to tissue repair impairment. Herein, it is shown that metal oxide nanoparticles (NPs), even at low and non-toxic doses (1 and 10 µg mL-1), can perturb the coordinated epithelial cell rotation (CECR) in micropatterned human epithelial cell clusters via distinct nanoparticle-specific mechanisms. Zinc oxide (ZnO) NPs are found to induce significant levels of intracellular reactive oxygen species (ROS) to promote mitogenic activity. Generation of a new localized force field through changes in the cytoskeleton organization and an increase in cell density leads to the arrest of CECR. Conversely, epithelial cell clusters exposed to titanium dioxide (TiO2) NPs maintain their CECR directionality but display suppressed rotational speed in an autophagy-dependent manner. Thus, these findings reveal that nanoparticles can actively hijack the nano-adaptive responses of epithelial cells to disrupt the fundamental mechanics of cooperation and communication in a collective setting.
Collapse
Affiliation(s)
- Jie Yan Cheryl Koh
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, 637141, Singapore
| | - Liuying Chen
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lingyan Gong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Shao Jie Tan
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Chor Yong Tay
- School of Material Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, Nanyang Technological University, Singapore, 637141, Singapore
| |
Collapse
|
5
|
Park SY, Park JH, Yang JW, Jung EJ, Ju YT, Jeong CY, Kim JY, Park T, Kim TH, Park M, Lee YJ, Jeong SH. SMARCD3 Overexpression Promotes Epithelial-Mesenchymal Transition in Gastric Cancer. Cancers (Basel) 2024; 16:2282. [PMID: 38927986 PMCID: PMC11201906 DOI: 10.3390/cancers16122282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
This study investigates the role of SMARCD3 in gastric cancer by comparing its expression in signet ring cell (SRC) and well-differentiated (WD) groups within gastric cancer cell lines and tissues. We observed elevated SMARCD3 levels in the SRC group compared to the WD group. Functional analysis was conducted through both SMARCD3 knock-in and knock-out methods. Kaplan-Meier survival analysis indicated that higher SMARCD3 expression correlates with poorer overall survival in gastric cancer patients (HR 2.16, p < 0.001). SMARCD3 knock-out cells showed decreased proliferation, migration, invasion, and expression of epithelial-mesenchymal transition (EMT) markers, contrasting with results from temporary and stable SMARCD3 overexpression experiments, which demonstrated increased cell area and irregularity (p < 0.001). Further analysis revealed that SMARCD3 overexpression in MKN-74 cells significantly enhanced p-AKT-S473 and p-ERK levels (p < 0.05), and in KATO III cells, it increased β-catenin and PI3Kp85 activities (p < 0.05). Conversely, these activities decreased in SNU 601 cells following SMARCD3 depletion. The study concludes that SMARCD3 overexpression may serve as a negative prognostic marker and a potential therapeutic target in gastric cancer treatment due to its role in promoting EMT.
Collapse
Affiliation(s)
- Sun Yi Park
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea (C.-Y.J.)
| | - Ji-Ho Park
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea (C.-Y.J.)
| | - Jung Wook Yang
- Department of Pathology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Eun-Jung Jung
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea; (E.-J.J.); (T.-H.K.)
| | - Young-Tae Ju
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea (C.-Y.J.)
| | - Chi-Young Jeong
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea (C.-Y.J.)
| | - Ju-Yeon Kim
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea (C.-Y.J.)
| | - Taejin Park
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea; (E.-J.J.); (T.-H.K.)
| | - Tae-Han Kim
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea; (E.-J.J.); (T.-H.K.)
| | - Miyeong Park
- Department of Anesthesiology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea
| | - Young-Joon Lee
- Department of Surgery, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea (C.-Y.J.)
| | - Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea; (E.-J.J.); (T.-H.K.)
| |
Collapse
|
6
|
Zhang W, Wang J, Shan C. The eEF1A protein in cancer: Clinical significance, oncogenic mechanisms, and targeted therapeutic strategies. Pharmacol Res 2024; 204:107195. [PMID: 38677532 DOI: 10.1016/j.phrs.2024.107195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Eukaryotic elongation factor 1A (eEF1A) is among the most abundant proteins in eukaryotic cells. Evolutionarily conserved across species, eEF1A is in charge of translation elongation for protein biosynthesis as well as a plethora of non-translational moonlighting functions for cellular homeostasis. In malignant cells, however, eEF1A becomes a pleiotropic driver of cancer progression via a broad diversity of pathways, which are not limited to hyperactive translational output. In the past decades, mounting studies have demonstrated the causal link between eEF1A and carcinogenesis, gaining deeper insights into its multifaceted mechanisms and corroborating its value as a prognostic marker in various cancers. On the other hand, an increasing number of natural and synthetic compounds were discovered as anticancer eEF1A-targeting inhibitors. Among them, plitidepsin was approved for the treatment of multiple myeloma whereas metarrestin was currently under clinical development. Despite significant achievements in these two interrelated fields, hitherto there lacks a systematic examination of the eEF1A protein in the context of cancer research. Therefore, the present work aims to delineate its clinical implications, molecular oncogenic mechanisms, and targeted therapeutic strategies as reflected in the ever expanding body of literature, so as to deepen mechanistic understanding of eEF1A-involved tumorigenesis and inspire the development of eEF1A-targeted chemotherapeutics and biologics.
Collapse
Affiliation(s)
- Weicheng Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| | - Jiyan Wang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Changliang Shan
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| |
Collapse
|
7
|
Kosinski J, Sechi A, Hain J, Villwock S, Ha SA, Hauschulz M, Rose M, Steib F, Ortiz‐Brüchle N, Heij L, Maas SL, van der Vorst EPC, Knoesel T, Altendorf‐Hofmann A, Simon R, Sauter G, Bednarsch J, Jonigk D, Dahl E. ITIH5 as a multifaceted player in pancreatic cancer suppression, impairing tyrosine kinase signaling, cell adhesion and migration. Mol Oncol 2024; 18:1486-1509. [PMID: 38375974 PMCID: PMC11161730 DOI: 10.1002/1878-0261.13609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
Inter-alpha-trypsin inhibitor heavy chain 5 (ITIH5) has been identified as a metastasis suppressor gene in pancreatic cancer. Here, we analyzed ITIH5 promoter methylation and protein expression in The Cancer Genome Atlas (TCGA) dataset and three tissue microarray cohorts (n = 618), respectively. Cellular effects, including cell migration, focal adhesion formation and protein tyrosine kinase activity, induced by forced ITIH5 expression in pancreatic cancer cell lines were studied in stable transfectants. ITIH5 promoter hypermethylation was associated with unfavorable prognosis, while immunohistochemistry demonstrated loss of ITIH5 in the metastatic setting and worsened overall survival. Gain-of-function models showed a significant reduction in migration capacity, but no alteration in proliferation. Focal adhesions in cells re-expressing ITIH5 exhibited a smaller and more rounded phenotype, typical for slow-moving cells. An impressive increase of acetylated alpha-tubulin was observed in ITIH5-positive cells, indicating more stable microtubules. In addition, we found significantly decreased activities of kinases related to focal adhesion. Our results indicate that loss of ITIH5 in pancreatic cancer profoundly affects its molecular profile: ITIH5 potentially interferes with a variety of oncogenic signaling pathways, including the PI3K/AKT pathway. This may lead to altered cell migration and focal adhesion formation. These cellular alterations may contribute to the metastasis-inhibiting properties of ITIH5 in pancreatic cancer.
Collapse
Affiliation(s)
- Jennifer Kosinski
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Antonio Sechi
- Department of Cell and Tumor BiologyRWTH Aachen UniversityGermany
| | - Johanna Hain
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Sophia Villwock
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Stefanie Anh Ha
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Maximilian Hauschulz
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Michael Rose
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Florian Steib
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Nadina Ortiz‐Brüchle
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Lara Heij
- Institute of PathologyUniversity Hospital EssenGermany
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
- Department of PathologyErasmus Medical Center RotterdamThe Netherlands
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityThe Netherlands
| | - Sanne L. Maas
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐University MunichGermany
| | - Thomas Knoesel
- Institute of PathologyLudwig‐Maximilians‐University MunichGermany
| | | | - Ronald Simon
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Guido Sauter
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Jan Bednarsch
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
| | - Danny Jonigk
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
- German Center for Lung Research (DZL), BREATHHanoverGermany
| | - Edgar Dahl
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
| |
Collapse
|
8
|
Tufail M, Wan WD, Jiang C, Li N. Targeting PI3K/AKT/mTOR signaling to overcome drug resistance in cancer. Chem Biol Interact 2024; 396:111055. [PMID: 38763348 DOI: 10.1016/j.cbi.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
This review comprehensively explores the challenge of drug resistance in cancer by focusing on the pivotal PI3K/AKT/mTOR pathway, elucidating its role in oncogenesis and resistance mechanisms across various cancer types. It meticulously examines the diverse mechanisms underlying resistance, including genetic mutations, feedback loops, and microenvironmental factors, while also discussing the associated resistance patterns. Evaluating current therapeutic strategies targeting this pathway, the article highlights the hurdles encountered in drug development and clinical trials. Innovative approaches to overcome resistance, such as combination therapies and precision medicine, are critically analyzed, alongside discussions on emerging therapies like immunotherapy and molecularly targeted agents. Overall, this comprehensive review not only sheds light on the complexities of resistance in cancer but also provides a roadmap for advancing cancer treatment.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
9
|
Gan X, Luo X, Chen J, Fang W, Nie M, Lu H, Liu Y, Wang X. Ilicicolin C suppresses the progression of prostate cancer by inhibiting PI3K/AKT/mTOR pathway. Mol Cell Biochem 2024:10.1007/s11010-024-05026-9. [PMID: 38801644 DOI: 10.1007/s11010-024-05026-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024]
Abstract
Aberrant activation of the PI3K/AKT pathway is a driving factor in the development of prostate cancer. Therefore, inhibiting the function of the PI3K/AKT signaling pathway is a strategy for the treatment of prostate cancer. Ilicicolin C is an ascochlorin derivative isolated from the coral-derived fungus Acremonium sclerotigenum GXIMD 02501. Which has anti-inflammatory activity, but its activity against prostate cancer has not yet been elucidated. MTT assay, plate clone-formation assay, flow cytometry and real-time cell analysis technology were used to detect the effects of ilicicolin C on cell viability, proliferation, apoptosis and migration of prostate cancer cells. Molecular docking software and surface plasmon resonance technology were used to analyze the interaction between ilicicolin C and PI3K/AKT proteins. Western blot assay was performed to examine the changes in protein expression. Finally, QikProp software was used to simulate the process of ilicicolin C in vivo, and a zebrafish xenograft model was used to further verify the anti-prostate cancer activity of ilicicolin C in vivo. Ilicicolin C showed cytotoxic effects on prostate cancer cells, with the most significant effect on PC-3 cells. Ilicicolin C inhibited proliferation and migration of PC-3 cells. It could also block the cell cycle and induce apoptosis in PC-3 cells. In addition, ilicicolin C could bind to PI3K/AKT proteins. Furthermore, ilicicolin C inhibited the expression of PI3K, AKT and mTOR proteins and could also regulate the expression of downstream proteins in the PI3K/AKT/mTOR signaling pathway. Moreover, the calculations speculated that ilicicolin C was well absorbed orally, and the zebrafish xenograft model confirmed the in vivo anti-prostate cancer effect of ilicicolin C. Ilicicolin C emerges as a promising marine compound capable of inducing apoptosis of prostate cancer cells by counteracting the aberrant activation of PI3K/AKT/mTOR, suggesting that ilicicolin C may be a viable candidate for anti-prostate cancer drug development. These findings highlight the potential of ilicicolin C against prostate cancer and shed light on its mechanism of action.
Collapse
Affiliation(s)
- Xia Gan
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Xiaowei Luo
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Jingqin Chen
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Wenxuan Fang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Mingyi Nie
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Humu Lu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Yonghong Liu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China.
| | - Xueni Wang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, Nanning, 530200, China.
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China.
| |
Collapse
|
10
|
Czekay RP, Higgins CE, Aydin HB, Samarakoon R, Subasi NB, Higgins SP, Lee H, Higgins PJ. SERPINE1: Role in Cholangiocarcinoma Progression and a Therapeutic Target in the Desmoplastic Microenvironment. Cells 2024; 13:796. [PMID: 38786020 PMCID: PMC11119900 DOI: 10.3390/cells13100796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
A heterogenous population of inflammatory elements, other immune and nonimmune cells and cancer-associated fibroblasts (CAFs) are evident in solid malignancies where they coexist with the growing tumor mass. In highly desmoplastic malignancies, CAFs are the prominent mesenchymal cell type in the tumor microenvironment (TME), where their presence and abundance signal a poor prognosis. CAFs play a major role in the progression of various cancers by remodeling the supporting stroma into a dense, fibrotic matrix while secreting factors that promote the maintenance of cancer stem-like characteristics, tumor cell survival, aggressive growth and metastasis and reduced sensitivity to chemotherapeutics. Tumors with high stromal fibrotic signatures are more likely to be associated with drug resistance and eventual relapse. Identifying the molecular underpinnings for such multidirectional crosstalk among the various normal and neoplastic cell types in the TME may provide new targets and novel opportunities for therapeutic intervention. This review highlights recent concepts regarding the complexity of CAF biology in cholangiocarcinoma, a highly desmoplastic cancer. The discussion focuses on CAF heterogeneity, functionality in drug resistance, contributions to a progressively fibrotic tumor stroma, the involved signaling pathways and the participating genes.
Collapse
Affiliation(s)
- Ralf-Peter Czekay
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA; (R.-P.C.); (C.E.H.); (R.S.); (S.P.H.)
| | - Craig E. Higgins
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA; (R.-P.C.); (C.E.H.); (R.S.); (S.P.H.)
| | - Hasan Basri Aydin
- Department of Pathology & Laboratory Medicine, Albany Medical College, Albany, NY 12208, USA; (H.B.A.); (N.B.S.); (H.L.)
| | - Rohan Samarakoon
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA; (R.-P.C.); (C.E.H.); (R.S.); (S.P.H.)
| | - Nusret Bekir Subasi
- Department of Pathology & Laboratory Medicine, Albany Medical College, Albany, NY 12208, USA; (H.B.A.); (N.B.S.); (H.L.)
| | - Stephen P. Higgins
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA; (R.-P.C.); (C.E.H.); (R.S.); (S.P.H.)
| | - Hwajeong Lee
- Department of Pathology & Laboratory Medicine, Albany Medical College, Albany, NY 12208, USA; (H.B.A.); (N.B.S.); (H.L.)
| | - Paul J. Higgins
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA; (R.-P.C.); (C.E.H.); (R.S.); (S.P.H.)
| |
Collapse
|
11
|
Ye Y, Lin M, Zhou G, Wang W, Yao Y, Su Y, Qi J, Zheng Y, Zhong C, Chen X, Huang M, Lu Y. Fuyuan decoction prevents nasopharyngeal carcinoma metastasis by inhibiting circulating tumor cells/ endothelial cells interplay and enhancing anti-cancer immune response. Front Pharmacol 2024; 15:1355650. [PMID: 38738179 PMCID: PMC11084272 DOI: 10.3389/fphar.2024.1355650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/14/2024] Open
Abstract
Distant metastasis is a major cause of treatment failure in cancer patients and a key challenge to improving cancer care today. We hypothesized that enhancing anti-cancer immune response and inhibiting circulating tumor cells (CTCs) adhesion and transendothelial migration through synergistic multi-target approaches may effectively prevent cancer metastasis. "Fuyuan Decoction" (FYD) is a traditional Chinese medicine compound that is widely used to prevent postoperative metastasis in cancer patients, but its underlying mechanism remains unclear. In this work, we systematically elucidated the underlying molecular mechanism by which FYD prevents cancer metastasis through multi-compound and multi-target synergies in vitro and in vivo. FYD significantly prevented cancer metastasis at non-cytotoxic concentrations by suppressing the adhesion of CTCs to endothelial cells and their subsequent transendothelial migration, as well as enhancing anti-cancer immune response. Mechanistically, FYD interrupts adhesion of CTCs to vascular endothelium by inhibiting TNF-α-induced CAMs expression via regulation of the NF-κB signaling pathway in endothelial cells. FYD inhibits invasion and migration of CTCs by suppressing EMT, PI3K/AKT and FAK signaling pathways. Moreover, FYD enhances the anti-cancer immune response by significantly increasing the population of Tc and NK cells in the peripheral immune system. In addition, the chemical composition of FYD was determined by UPLC-HRMS, and the results indicated that multiple compounds in FYD prevents cancer metastasis through multi-target synergistic treatment. This study provides a modern medical basis for the application of FYD in the prevention of cancer metastasis, and suggesting that multi-drug and multi-target synergistic therapy may be one of the most effective ways to prevent cancer metastasis.
Collapse
Affiliation(s)
- Yuying Ye
- Department of Otorhinolaryngology, Affiliated People’s Hospital (Fujian Provincial People’s Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mengting Lin
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Guiyu Zhou
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Weiyu Wang
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Yinyin Yao
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Yafei Su
- Department of Otorhinolaryngology, Fuzhou Second Hospital, Fuzhou, China
| | - Jianqiang Qi
- Center for Teaching of Clinical Skills, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yanfang Zheng
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Chunlian Zhong
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, China
| | - Xi Chen
- Department of Otorhinolaryngology, Affiliated People’s Hospital (Fujian Provincial People’s Hospital), Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mingqing Huang
- College of Pharmacy, Fujian Key Laboratory of Chinese Materia Medica, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yusheng Lu
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, China
- Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, China
| |
Collapse
|
12
|
Hirai Y, Kawaguchi Y, Kasahara C, Hirose H, Futaki S. Liquid Droplet-Mediated Formulation of Lipid Nanoparticles Encapsulating Immunoglobulin G for Cytosolic Delivery. Mol Pharm 2024; 21:1653-1661. [PMID: 38290425 DOI: 10.1021/acs.molpharmaceut.3c00868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Antibodies are promising biopharmaceuticals that offer new therapeutic options for diseases. Since antibodies are membrane impermeable, approaches that allow immunoglobulin Gs (IgGs) to access intracellular therapeutic targets would open new horizons in antibody therapies. Lipid nanoparticles (LNPs) are among the classes of vectors that deliver biopharmaceuticals into cells. Using liquid droplets formed by IgG and polyglutamate, we report here a unique approach to forming LNPs containing IgG via liquid droplets formed in the presence of polyglutamic acid (polyE). The addition of polyE promoted the formation of smaller LNPs with cationic lipids than in its absence, and the formed LNPs were much more efficient in cytosolic IgG delivery and targeting of cellular proteins. This approach also allows for the encapsulation of intact IgG without the need for chemical or sequence modification. The intracellularly delivered IgG retained its target binding ability, as demonstrated by labeling of nuclear pore complex and HRas-GFP and inhibition of antiapoptotic cell death by phosphorylated Akt protein in live cells.
Collapse
Affiliation(s)
- Yusuke Hirai
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yoshimasa Kawaguchi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Chisato Kasahara
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Hisaaki Hirose
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
13
|
Zhang Q, Yuan Y, Cao S, Kang N, Qiu F. Withanolides: Promising candidates for cancer therapy. Phytother Res 2024; 38:1104-1158. [PMID: 38176694 DOI: 10.1002/ptr.8090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Natural products have played a significant role throughout history in the prevention and treatment of numerous diseases, particularly cancers. As a natural product primarily derived from various medicinal plants in the Withania genus, withanolides have been shown in several studies to exhibit potential activities in cancer treatment. Consequently, understanding the molecular mechanism of withanolides could herald the discovery of new anticancer agents. Withanolides have been studied widely, especially in the last 20 years, and attracted the attention of numerous researchers. Currently, over 1200 withanolides have been classified, with approximately a quarter of them having been reported in the literature to be able to modulate the survival and death of cancer cells through multiple avenues. To what extent, though, has the anticancer effects of these compounds been studied? How far are they from being developed into clinical drugs? What are their potential, characteristic features, and challenges? In this review, we elaborate on the current knowledge of natural compounds belonging to this class and provide an overview of their natural sources, anticancer activity, mechanism of action, molecular targets, and implications for anticancer drug research. In addition, direct targets and clinical research to guide the design and implementation of future preclinical and clinical studies to accelerate the application of withanolides have been highlighted.
Collapse
Affiliation(s)
- Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - YongKang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shijie Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
14
|
Keesari PR, Jain A, Ganampet NR, Subhasri GSD, Edusa S, Muslehuddin Z, Theik NWY, Palisetti S, Salibindla D, Manaktala PS, Desai R. Association between prediabetes and breast cancer: a comprehensive meta-analysis. Breast Cancer Res Treat 2024; 204:1-13. [PMID: 38060076 DOI: 10.1007/s10549-023-07181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Breast cancer accounts for up to 30% of cancer cases in women in the US. Diabetes mellitus has been recognized as a risk factor for breast cancer. Some studies have suggested that prediabetes may also be associated with breast cancer whereas other studies have shown no or an inverse association; thus, we conducted a meta-analysis to assess the risk of breast cancer in prediabetes. METHODS We searched PubMed/Medline, EMBASE, Google Scholar, and Scopus to identify studies that reported breast cancer risks in patients having prediabetes compared to normoglycemic patients. Binary random-effects model was used to calculate a pooled odds ratio (OR) with 95% confidence intervals. I2 statistics were used to assess heterogeneity. Leave-one-out sensitivity analysis and subgroup analyses were performed. RESULTS We analyzed 7 studies with 24,586 prediabetic and 224,314 normoglycemic individuals (783 and 5739 breast cancer cases, respectively). Unadjusted odds ratio (OR) for breast cancer was 1.45 (95% CI = 1.14, 1.83); adjusted OR was 1.19 (95% CI = 1.07, 1.34) in prediabetes. Subgroup analysis revealed a higher breast cancer risk in individuals aged less than 60 years (OR = 1.86, 95% CI = 1.39, 2.49) than in those aged 60 years or more (OR = 1.07, 95% CI = 0.97, 1.18). Subgroup analysis by median follow-up length indicated a higher risk of breast cancer for follow-ups of less than or equal to 2 years (OR = 2.34, 95% CI = 1.85, 2.95) than in those of over 10 years (OR = 1.1, 95% CI = 0.99, 1.23) and 6 to 10 years (OR = 1.03, 95% CI = 0.88, 1.21). CONCLUSIONS In conclusion, individuals with prediabetes have higher risk of developing breast cancer than those with normoglycemia, especially younger prediabetes patients. These individuals may benefit from early identification, monitoring, and interventions to reverse prediabetes.
Collapse
Affiliation(s)
- Praneeth Reddy Keesari
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Akhil Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | | | | | - Samuel Edusa
- Department of Internal Medicine, Piedmont Athens Regional, Athens, GA, USA
| | - Zainab Muslehuddin
- Department of Internal Medicine, Detroit Medical Center, Wayne State University-Sinai Grace Hospital, Detroit, MI, USA
| | | | - Spandana Palisetti
- Department of Medicine, Jawaharlal Nehru Medical College, Belgaum, Karnataka, India
| | | | | | - Rupak Desai
- Independent Researcher, Outcomes Research, Atlanta, GA, USA
| |
Collapse
|
15
|
Bracho-Valdés I, Cervantes-Villagrana RD, Beltrán-Navarro YM, Olguín-Olguín A, Escobar-Islas E, Carretero-Ortega J, Olivares-Reyes JA, Reyes-Cruz G, Gutkind JS, Vázquez-Prado J. Akt Is Controlled by Bag5 through a Monoubiquitination to Polyubiquitination Switch. Int J Mol Sci 2023; 24:17531. [PMID: 38139359 PMCID: PMC10743781 DOI: 10.3390/ijms242417531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
The serine-threonine kinase Akt plays a fundamental role in cell survival, metabolism, proliferation, and migration. To keep these essential processes under control, Akt activity and stability must be tightly regulated; otherwise, life-threatening conditions might prevail. Although it is well understood that phosphorylation regulates Akt activity, much remains to be known about how its stability is maintained. Here, we characterize BAG5, a chaperone regulator, as a novel Akt-interactor and substrate that attenuates Akt stability together with Hsp70. BAG5 switches monoubiquitination to polyubiquitination of Akt and increases its degradation caused by Hsp90 inhibition and Hsp70 overexpression. Akt interacts with BAG5 at the linker region that joins the first and second BAG domains and phosphorylates the first BAG domain. The Akt-BAG5 complex is formed in serum-starved conditions and dissociates in response to HGF, coincident with BAG5 phosphorylation. BAG5 knockdown attenuated Akt degradation and facilitated its activation, whereas the opposite effect was caused by BAG5 overexpression. Altogether, our results indicate that Akt stability and signaling are dynamically regulated by BAG5, depending on growth factor availability.
Collapse
Affiliation(s)
- Ismael Bracho-Valdés
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
- Academic Department of Apparatus and Systems I, Deanship of Health Sciences, Universidad Autónoma de Guadalajara, Av. Patria 1201, Zapopan 45129, Mexico
| | - Rodolfo Daniel Cervantes-Villagrana
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
- Department of Pharmacology, Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, San Diego, CA 92093, USA
| | - Yarely Mabell Beltrán-Navarro
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
| | - Adán Olguín-Olguín
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
| | - Estanislao Escobar-Islas
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
| | - Jorge Carretero-Ortega
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
| | - J. Alberto Olivares-Reyes
- Department of Biochemistry, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico
| | - J. Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, San Diego, CA 92093, USA
| | - José Vázquez-Prado
- Department of Pharmacology, Cinvestav-IPN. Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Mexico City 07360, Mexico; (I.B.-V.)
| |
Collapse
|
16
|
Qi X, Chen Y, Liu S, Liu L, Yu Z, Yin L, Fu L, Deng M, Liang S, Lü M. Sanguinarine inhibits melanoma invasion and migration by targeting the FAK/PI3K/AKT/mTOR signalling pathway. PHARMACEUTICAL BIOLOGY 2023; 61:696-709. [PMID: 37092313 PMCID: PMC10128503 DOI: 10.1080/13880209.2023.2200787] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Sanguinarine (SAG) is the most abundant constituent of Macleaya cordata (Willd.) R. Br. (Popaceae). SAG has shown antimammary and colorectal metastatic effects in mice in vivo, suggesting its potential for cancer chemotherapy. OBJECTIVE To determine the antimetastatic effect and underlying molecular mechanisms of SAG on melanoma. MATERIALS AND METHODS CCK8 assay was used to determine the inhibition of SAG on the proliferation of A375 and A2058 cells. Network pharmacology analysis was applied to construct a compound-target network and select potential therapeutic targets of SAG against melanoma. Molecular docking simulation was conducted for further analysis of the selected targets. In vitro migration/invasion/western blot assay with 1, 1.5, 2 μM SAG and in vivo effect of 2, 4, 8 mg/kg SAG in xenotransplantation model in nude mice. RESULTS The key targets of SAG treatment for melanoma were mainly enriched in PI3K-AKT pathway, and the binding energy of SAG to PI3K, AKT, and mTOR were -6.33, -6.31, and -6.07 kcal/mol, respectively. SAG treatment inhibited the proliferation, migration, and invasion ability of A375 and A2058 cells (p < 0.05) with IC50 values of 2.378 μM and 2.719 μM, respectively. It also decreased the phosphorylation levels of FAK, PI3K, AKT, mTOR and protein expression levels of MMP2 and ICAM-2. In the nude mouse xenograft model, 2, 4, 8 mg/kg SAG was shown to be effective in inhibiting tumour growth. CONCLUSIONS Our research offered a theoretical foundation for the clinical antitumor properties of SAG, further suggesting its potential application in the clinic.
Collapse
Affiliation(s)
- Xiaoyi Qi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- The Public Platform of Advanced Detecting Instruments, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou, China
- Cardiovascular and Metabolic Diseases of Sichuan Key Laboratory, Luzhou, China
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang University, Hangzhou, China
| | - Yonglan Chen
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Sha Liu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Liu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zehui Yu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ling Yin
- The Public Platform of Advanced Detecting Instruments, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Lu Fu
- The Public Platform of Advanced Detecting Instruments, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Mingming Deng
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Sicheng Liang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- The Public Platform of Advanced Detecting Instruments, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou, China
- Cardiovascular and Metabolic Diseases of Sichuan Key Laboratory, Luzhou, China
- CONTACT Sicheng Liang Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China; The Public Platform of Advanced Detecting Instruments, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China; Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou, China; Cardiovascular and Metabolic Diseases of Sichuan Key Laboratory, Luzhou, China
| | - Muhan Lü
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou, China
- Cardiovascular and Metabolic Diseases of Sichuan Key Laboratory, Luzhou, China
- Muhan Lü Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Human Microecology and Precision Diagnosis and Treatment of Luzhou Key Laboratory, Luzhou, China; Cardiovascular and Metabolic Diseases of Sichuan Key Laboratory, Luzhou, China
| |
Collapse
|
17
|
Islam M, Jones S, Ellis I. Role of Akt/Protein Kinase B in Cancer Metastasis. Biomedicines 2023; 11:3001. [PMID: 38002001 PMCID: PMC10669635 DOI: 10.3390/biomedicines11113001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Metastasis is a critical step in the process of carcinogenesis and a vast majority of cancer-related mortalities result from metastatic disease that is resistant to current therapies. Cell migration and invasion are the first steps of the metastasis process, which mainly occurs by two important biological mechanisms, i.e., cytoskeletal remodelling and epithelial to mesenchymal transition (EMT). Akt (also known as protein kinase B) is a central signalling molecule of the PI3K-Akt signalling pathway. Aberrant activation of this pathway has been identified in a wide range of cancers. Several studies have revealed that Akt actively engages with the migratory process in motile cells, including metastatic cancer cells. The downstream signalling mechanism of Akt in cell migration depends upon the tumour type, sites, and intracellular localisation of activated Akt. In this review, we focus on the role of Akt in the regulation of two events that control cell migration and invasion in various cancers including head and neck squamous cell carcinoma (HNSCC) and the status of PI3K-Akt pathway inhibitors in clinical trials in metastatic cancers.
Collapse
Affiliation(s)
- Mohammad Islam
- Unit of Cell and Molecular Biology, School of Dentistry, University of Dundee, Park Place, Dundee DD1 4HR, UK; (S.J.); (I.E.)
| | | | | |
Collapse
|
18
|
Nukpook T, Kiyono T, Ekalaksananan T, Kasemsiri P, Teeramatwanich W, Vatanasapt P, Chaiwiriyakul S, Nakahara T, Pientong C. An in vitro model and the underlying pathways of sinonasal inverted papilloma development. Sci Rep 2023; 13:18456. [PMID: 37891239 PMCID: PMC10611779 DOI: 10.1038/s41598-023-45585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Recently, the specific association between Sinonasal inverted papilloma (SIP) and EGFR exon 20 mutations has been reported. To investigate the link between specific EGFR mutations and SIP development, we established organotypic raft culture system using nasal polyp-derived immortalized NP2 (iNP2) cells expressing EGFR exon 20 mutants or an exon 19 mutant, and SIP-derived iIP4 cells harboring P772_H773insPYNP mutation. In the raft culture, iIP4 cells showed the inverted growth pattern characteristic to SIP. Interestingly, iNP2 cells expressing EGFR exon 20 duplication mutants, S768_D770dup and N771_H773dup, but not of EGFR exon 19 mutant, E746_A750del, showed the inverted growth pattern. Enhanced activation of the PI3K/AKT signaling pathway was observed in iNP2_S768_D770dup and iIP4 cells, while increased MAPK signaling was found in iNP2_N771_H773dup. Increased cell migration and invasion were found in all cells carrying EGFR mutations when compared to iNP2 cells, and this effect was inhibited by either PI3K or MEK inhibitor. Notably, iNP2 cells expressing the N771_H773dup mutant showed the highest migration and invasion abilities. These results suggest that specific mutations in EGFR exon 20 play a crucial role in SIP development, partially though hyper-activation of the PI3K/AKT and MAPK signaling pathways. This study presents the first in vitro model for SIP development, which could facilitate further investigations into SIP pathogenesis and preclinical studies for new therapeutic agents.
Collapse
Affiliation(s)
- Thawaree Nukpook
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Tohru Kiyono
- Project for Prevention of HPV-Related Cancer, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Pornthep Kasemsiri
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watchareporn Teeramatwanich
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Patravoot Vatanasapt
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
- Department of Otorhinolaryngology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Tomomi Nakahara
- Division of Immune Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuoku, Tokyo, 104-0045, Japan
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
19
|
Dedigama-Arachchige PM, Acharige NPN, Zhang X, Bremer HJ, Yi Z, Pflum MKH. Identification of PP1c-PPP1R12A Substrates Using Kinase-Catalyzed Biotinylation to Identify Phosphatase Substrates. ACS OMEGA 2023; 8:35628-35637. [PMID: 37810667 PMCID: PMC10552495 DOI: 10.1021/acsomega.3c01944] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/21/2023] [Indexed: 10/10/2023]
Abstract
Protein phosphatase 1 regulatory subunit 12A (PPP1R12A) interacts with the catalytic subunit of protein phosphatase 1 (PP1c) to form the myosin phosphatase complex. In addition to a well-documented role in muscle contraction, the PP1c-PPP1R12A complex is associated with cytoskeleton organization, cell migration and adhesion, and insulin signaling. Despite the variety of biological functions, only a few substrates of the PP1c-PPP1R12A complex are characterized, which limit a full understanding of PP1c-PPP1R12A activities in muscle contraction and cytoskeleton regulation. Here, the chemoproteomics method Kinase-catalyzed Biotinylation to Identify Phosphatase Substrates (K-BIPS) was used to identify substrates of the PP1c-PPP1R12A complex in L6 skeletal muscle cells. K-BIPS enriched 136 candidate substrates with 14 high confidence hits. One high confidence hit, AKT1 kinase, was validated as a novel PP1c-PPP1R12A substrate. Given the previously documented role of AKT1 in PPP1R12A phosphorylation and cytoskeleton organization, the data suggest that PP1c-PPP1R12A regulates its own phosphatase activity through an AKT1-dependent feedback mechanism to influence cytoskeletal arrangement in muscle cells.
Collapse
Affiliation(s)
| | - Nuwan P N Acharige
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit 48202-3489, Michigan, United States
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, Wayne State University, 259 Mack Ave, Detroit 48201, Michigan, United States
| | - Hannah J Bremer
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit 48202-3489, Michigan, United States
| | - Zhengping Yi
- Department of Pharmaceutical Sciences, Wayne State University, 259 Mack Ave, Detroit 48201, Michigan, United States
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit 48202-3489, Michigan, United States
| |
Collapse
|
20
|
Shi Q, Gui J, Sun L, Song Y, Na J, Zhang J, Fan Y, Zheng L. Frizzled-9 triggers actin polymerization and activates mechano-transducer YAP to rescue simulated microgravity-induced osteoblast dysfunction. FASEB J 2023; 37:e23147. [PMID: 37585277 DOI: 10.1096/fj.202300977r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023]
Abstract
Long-term spaceflight can result in bone loss and osteoblast dysfunction. Frizzled-9 (Fzd9) is a Wnt receptor of the frizzled family that is vital for osteoblast differentiation and bone formation. In the present study, we elucidated whether Fzd9 plays a role in osteoblast dysfunction induced by simulated microgravity (SMG). After 1-7 days of SMG, osteogenic markers such as alkaline phosphatase (ALP), osteopontin (OPN), and Runt-related transcription factor 2 (RUNX2) were decreased, accompanied by a decrease in Fzd9 expression. Furthermore, Fzd9 expression decreased in the rat femur after 3 weeks of hindlimb unloading. In contrast, Fzd9 overexpression counteracted the decrease in ALP, OPN, and RUNX2 induced by SMG in osteoblasts. Moreover, SMG regulated phosphorylated glycogen synthase kinase-3β (pGSK3β) and β-catenin expression or sublocalization. However, Fzd9 overexpression did not affect pGSK3β and β-catenin expression or sublocalization induced by SMG. In addition, Fzd9 overexpression regulated protein kinase B also known as Akt and extracellular signal-regulated kinase (ERK) phosphorylation and induced F-actin polymerization to form the actin cap, press the nuclei, and increase nuclear pore size, thereby promoting the nuclear translocation of Yes-associated protein (YAP). Our study findings provide mechanistic insights into the role of Fzd9 in triggering actin polymerization and activating YAP to rescue SMG-induced osteoblast dysfunction and suggest that Fzd9 is a potential target to restore osteoblast function in individuals with bone diseases and after spaceflight.
Collapse
Affiliation(s)
- Qiusheng Shi
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jinpeng Gui
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lianwen Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yaxin Song
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Na
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jingyi Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
21
|
Silvestri R, Nicolì V, Gangadharannambiar P, Crea F, Bootman MD. Calcium signalling pathways in prostate cancer initiation and progression. Nat Rev Urol 2023; 20:524-543. [PMID: 36964408 DOI: 10.1038/s41585-023-00738-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/26/2023]
Abstract
Cancer cells proliferate, differentiate and migrate by repurposing physiological signalling mechanisms. In particular, altered calcium signalling is emerging as one of the most widespread adaptations in cancer cells. Remodelling of calcium signalling promotes the development of several malignancies, including prostate cancer. Gene expression data from in vitro, in vivo and bioinformatics studies using patient samples and xenografts have shown considerable changes in the expression of various components of the calcium signalling toolkit during the development of prostate cancer. Moreover, preclinical and clinical evidence suggests that altered calcium signalling is a crucial component of the molecular re-programming that drives prostate cancer progression. Evidence points to calcium signalling re-modelling, commonly involving crosstalk between calcium and other cellular signalling pathways, underpinning the onset and temporal progression of this disease. Discrete alterations in calcium signalling have been implicated in hormone-sensitive, castration-resistant and aggressive variant forms of prostate cancer. Hence, modulation of calcium signals and downstream effector molecules is a plausible therapeutic strategy for both early and late stages of prostate cancer. Based on this premise, clinical trials have been undertaken to establish the feasibility of targeting calcium signalling specifically for prostate cancer.
Collapse
Affiliation(s)
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Francesco Crea
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Martin D Bootman
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
22
|
Desjardins-Lecavalier N, Annis MG, Nowakowski A, Kiepas A, Binan L, Roy J, Modica G, Hébert S, Kleinman CL, Siegel PM, Costantino S. Migration speed of captured breast cancer subpopulations correlates with metastatic fitness. J Cell Sci 2023; 136:jcs260835. [PMID: 37313743 PMCID: PMC10657211 DOI: 10.1242/jcs.260835] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 06/02/2023] [Indexed: 06/15/2023] Open
Abstract
The genetic alterations contributing to migration proficiency, a phenotypic hallmark of metastatic cells required for colonizing distant organs, remain poorly defined. Here, we used single-cell magneto-optical capture (scMOCa) to isolate fast cells from heterogeneous human breast cancer cell populations, based on their migratory ability alone. We show that captured fast cell subpopulations retain higher migration speed and focal adhesion dynamics over many generations as a result of a motility-related transcriptomic profile. Upregulated genes in isolated fast cells encoded integrin subunits, proto-cadherins and numerous other genes associated with cell migration. Dysregulation of several of these genes correlates with poor survival outcomes in people with breast cancer, and primary tumors established from fast cells generated a higher number of circulating tumor cells and soft tissue metastases in pre-clinical mouse models. Subpopulations of cells selected for a highly migratory phenotype demonstrated an increased fitness for metastasis.
Collapse
Affiliation(s)
- Nicolas Desjardins-Lecavalier
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
- Institut de genie biomedical, University of Montreal, Pavillon Paul-G.-Desmarais, 2960, chemin de la Tour, Montréal, QC H3T 1J4, Canada
| | - Matthew G. Annis
- Goodman Cancer Institute, McGill University, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada
- Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Alexander Nowakowski
- Goodman Cancer Institute, McGill University, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada
- Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Alexander Kiepas
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health Bethesda, MA 20892-4370, USA
| | - Loïc Binan
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
| | - Joannie Roy
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
| | - Graziana Modica
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
| | - Steven Hébert
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E2, Canada
| | - Claudia L. Kleinman
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montréal, QC H3T 1E2, Canada
| | - Peter M. Siegel
- Goodman Cancer Institute, McGill University, 1160 Pine Avenue West, Montreal, QC H3A 1A3, Canada
- Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Santiago Costantino
- Maisonneuve-Rosemont Hospital Research Center, 5415, boulevard de l'Assomption, Montréal, QC H1T 2M4, Canada
- Department of Ophthalmology, University of Montreal, Pavillon Roger-Gaudry, Bureau S-700, 2900, boul. Édouard-Montpetit, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
23
|
Splitt RL, DeMali KA. Metabolic reprogramming in response to cell mechanics. Biol Cell 2023; 115:e202200108. [PMID: 36807920 PMCID: PMC10192020 DOI: 10.1111/boc.202200108] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/20/2023]
Abstract
Much attention has been dedicated to understanding how cells sense and respond to mechanical forces. The types of forces cells experience as well as the repertoire of cell surface receptors that sense these forces have been identified. Key mechanisms for transmitting that force to the cell interior have also emerged. Yet, how cells process mechanical information and integrate it with other cellular events remains largely unexplored. Here we review the mechanisms underlying mechanotransduction at cell-cell and cell-matrix adhesions, and we summarize the current understanding of how cells integrate information from the distinct adhesion complexes with cell metabolism.
Collapse
Affiliation(s)
- Rebecca L. Splitt
- Department of Biochemistry and Molecular Biology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242
| | - Kris A. DeMali
- Department of Biochemistry and Molecular Biology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242
| |
Collapse
|
24
|
Yang J, Dong C, Wu J, Liu D, Luo Q, Jin X. Fructose utilization enhanced by GLUT5 promotes lung cancer cell migration via activating glycolysis/AKT pathway. Clin Transl Oncol 2023; 25:1080-1090. [PMID: 36454516 DOI: 10.1007/s12094-022-03015-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/12/2022] [Indexed: 12/03/2022]
Abstract
PURPOSE Lung cancer is the leading cause of cancer-related mortalities worldwide, and metastasis contributes to a large number of deaths in lung carcinoma patients. New approaches for anti-metastatic treatment are urgently needed. Enhanced fructose metabolism mediated by GLUT5 directly contributes to cancer metastasis. However, the underlying mechanism remains to be elucidated, which we aimed to explore in this study. METHODS The overexpression and knockdown of SLC2A5, the encoding gene of GLUT5, were established by retrovirus system and CRISPR/Cas9 technology, respectively. Cell migration was conducted by trans-well assay. Western blotting assay was carried out to detect the expression of GLUT5, total AKT, phosphorylated AKT (pAKT-S473 and pAKT-T308) and LDHA. Lactate production was measured by colorimetric assay. Experimental lung metastasis model by tail vein injection was constructed to evaluate the metastatic potential of GLUT5 in vivo. RESULTS Overexpression of SLC2A5 promoted migration of lung cancer cells both in vitro and in vivo, and shortened the overall survival of mice. While, SLC2A5 deletion blocked the migration of lung cancer cells. GLUT5-mediated fructose utilization upregulated phosphorylated AKT, which was responsible for enhanced migration of lung cancer cells. Additionally, GLUT5-mediated fructose utilization boosted glycolysis with overproduction of lactate, resulting in upregulation of phosphorylated AKT. Moreover, lung cancer cell migration and AKT activation were restrained by glycolysis inhibitor 2-deoxy-D-glucose (2-DG) or GLUT5-specific inhibitor 2,5-anhydro-D-mannitol (2,5-AM). CONCLUSION Our study unveils glycolysis/lactate/AKT pathway is responsible for lung cancer cell migration induced by GLUT5-mediated fructose metabolism, providing a potential therapeutic avenue for lung cancer metastasis.
Collapse
Affiliation(s)
- Jing Yang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Changsheng Dong
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Jia Wu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Dan Liu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qin Luo
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Xing Jin
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
25
|
α-Hederin Saponin Augments the Chemopreventive Effect of Cisplatin against Ehrlich Tumors and Bioinformatic Approach Identifying the Role of SDF1/CXCR4/p-AKT-1/NFκB Signaling. Pharmaceuticals (Basel) 2023; 16:ph16030405. [PMID: 36986504 PMCID: PMC10056433 DOI: 10.3390/ph16030405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/10/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF1) and its C-X-C chemokine receptor type 4 receptor (CXCR4) are significant mediators for cancer cells’ proliferation, and we studied their expression in Ehrlich solid tumors (ESTs) grown in mice. α-Hederin is a pentacyclic triterpenoid saponin found in Hedera or Nigella species with biological activity that involves suppression of growth of breast cancer cell lines. The aim of this study was to explore the chemopreventive activity of α-hederin with/without cisplatin; this was achieved by measuring the reduction in tumor masses and the downregulation in SDF1/CXCR4/pAKT signaling proteins and nuclear factor kappa B (NFκB). Ehrlich carcinoma cells were injected in four groups of Swiss albino female mice (Group1: EST control group, Group2: EST + α-hederin group, Group3: EST + cisplatin group, and Group4: EST+α-hederin/cisplatin treated group). Tumors were dissected and weighed, one EST was processed for histopathological staining with hematoxylin and eosin (HE), and the second MC was frozen and processed for estimation of signaling proteins. Computational analysis for these target proteins interactions showed direct-ordered interactions. The dissected solid tumors revealed decreases in tumor masses (~21%) and diminished viable tumor regions with significant necrotic surrounds, particularly with the combination regimens. Immunohistochemistry showed reductions (~50%) in intratumoral NFκβ in the mouse group that received the combination therapy. The combination treatment lowered the SDF1/CXCR4/p-AKT proteins in ESTs compared to the control. In conclusion, α-hederin augmented the chemotherapeutic potential of cisplatin against ESTs; this effect was at least partly mediated through suppressing the chemokine SDF1/CXCR4/p-AKT/NFκB signaling. Further studies are recommended to verify the chemotherapeutic potential of α-hederin in other breast cancer models.
Collapse
|
26
|
Paccosi E, Balzerano A, Proietti-De-Santis L. Interfering with the Ubiquitin-Mediated Regulation of Akt as a Strategy for Cancer Treatment. Int J Mol Sci 2023; 24:ijms24032809. [PMID: 36769122 PMCID: PMC9917864 DOI: 10.3390/ijms24032809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The serine/threonine kinase Akt modulates the functions of numerous substrates, many of them being involved in cell proliferation and growth, metabolism, angiogenesis, resistance to hypoxia and migration. Akt is frequently deregulated in many types of human cancers, its overexpression or abnormal activation being associated with the increased proliferation and survival of cancer cells. A promising avenue for turning off the functionality of Akt is to either interfere with the K63-linked ubiquitination that is necessary for Akt membrane recruitment and activation or increase the K48-linked polyubiquitination that aims to target Akt to the proteasome for its degradation. Recent evidence indicates that targeting the ubiquitin proteasome system is effective for certain cancer treatments. In this review, the functions and roles of Akt in human cancer will be discussed, with a main focus on molecules and compounds that target various elements of the ubiquitination processes that regulate the activation and inactivation of Akt. Moreover, their possible and attractive implications for cancer therapy will be discussed.
Collapse
|
27
|
Han I, Song IS, Choi SA, Lee T, Yusupov M, Shaw P, Bogaerts A, Choi EH, Ryu JJ. Bioactive Nonthermal Biocompatible Plasma Enhances Migration on Human Gingival Fibroblasts. Adv Healthc Mater 2023; 12:e2200527. [PMID: 36373222 DOI: 10.1002/adhm.202200527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 10/18/2022] [Indexed: 11/16/2022]
Abstract
This study hypothesizes that the application of low-dose nonthermal biocompatible dielectric barrier discharge plasma (DBD-NBP) to human gingival fibroblasts (HGFs) will inhibit colony formation but not cell death and induce matrix metalloproteinase (MMP) expression, extracellular matrix (ECM) degradation, and subsequent cell migration, which can result in enhanced wound healing. HGFs treated with plasma for 3 min migrate to each other across the gap faster than those in the control and 5-min treatment groups on days 1 and 3. The plasma-treated HGFs show significantly high expression levels of the cell cycle arrest-related p21 gene and enhanced MMP activity. Focal adhesion kinase (FAK) mediated attenuation of wound healing or actin cytoskeleton rearrangement, and plasma-mediated reversal of this attenuation support the migratory effect of DBD-NBP. Further, this work performs computer simulations to investigate the effect of oxidation on the stability and conformation of the catalytic kinase domain (KD) of FAK. It is found that the oxidation of highly reactive amino acids (AAs) Cys427, Met442, Cys559, Met571, Met617, and Met643 changes the conformation and increases the structural flexibility of the FAK protein and thus modulates its function and activity. Low-dose DBD-NBP-induces host cell cycle arrest, ECM breakdown, and subsequent migration, thus contributing to the enhanced wound healing process.
Collapse
Affiliation(s)
- Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Kwangwoon University, Seoul, 01897, Republic of Korea.,Department of Plasma Bio-Display, Kwangwoon University, Seoul, 01897, Korea
| | - In-Seok Song
- Department of Dentistry, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, 03080, Republic of Korea
| | - Taebok Lee
- Confocal Core Facility, Center for Medical Innovation, Seoul National University Hospital, Seoul, 03082, Korea
| | - Maksudbek Yusupov
- Research group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Priyanka Shaw
- Research group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Annemie Bogaerts
- Research group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Jae Jun Ryu
- Department of Dentistry, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| |
Collapse
|
28
|
Acharekar A, Bachal K, Shirke P, Thorat R, Banerjee A, Gardi N, Majumder A, Dutt S. Substrate stiffness regulates the recurrent glioblastoma cell morphology and aggressiveness. Matrix Biol 2023; 115:107-127. [PMID: 36563706 DOI: 10.1016/j.matbio.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022]
Abstract
Recurrent glioblastoma is highly aggressive with currently no specific treatment regime. Therefore, to identify novel therapeutic targets for recurrent GBM, we used a cellular model developed in our lab from commercially available cell line U87MG and patient-derived cultures that allows the comparison between radiation naïve (Parent) and recurrent GBM cells generated after parent cells are exposed to lethal dose of radiation. Total RNA-seq of parent and recurrent population revealed significant upregulation of cell-ECM interactions pathway in the recurrent population. These results led us to hypothesize that the physical microenvironment contributes to the aggressiveness of recurrent GBM. To verify this, we cultured parent and recurrent GBM cells on collagen-coated polyacrylamide gels mimicking the stiffness of normal brain (Young's modulus E = 0.5kPa) or tumorigenic brain (E = 10kPa) and tissue culture plastic dishes (E ∼ 1 GPa). We found that compared to parent cells, recurrent cells showed higher proliferation, invasion, migration, and resistance to EGFR inhibitor. Using orthotopic GBM mouse model and resection model, we demonstrate that recurrent cells cultured on 0.5kPa had higher in vivo tumorigenicity and recurrent disease progression than parent cells, whereas these differences were insignificant when parent and recurrent cells were cultured on plastic substrates. Furthermore, recurrent cells on 0.5kPa showed high expression of ECM proteins like Collagen, MMP2 and MMP9. These proteins were also significantly upregulated in recurrent patient biopsies. Additionally, the brain of mice injected with recurrent cells grown on 0.5kPa showed higher Young's moduli suggesting the ability of these cells to make the surrounding ECM stiffer. Total RNA-seq of parent and recurrent cells grown on plastic and 0.5kpa identified PLEKHA7 significantly upregulated specifically in recurrent cells grown on 0.5 kPa substrate. PLEKHA7 was also found to be high in recurrent GBM patient biopsies. Accordingly, PLEKHA7 knockdown reduced invasion and survival of recurrent GBM cells. Together, these data provide an in vitro model system that captures the observed in vivo and clinical behavior of recurrent GBM by mimicking mechanical microenvironment and identifies PLEKHA7 as a novel potential target for recurrent GBM.
Collapse
Affiliation(s)
- Anagha Acharekar
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Ketaki Bachal
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Pallavi Shirke
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India
| | - Archisman Banerjee
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Nilesh Gardi
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Abhijit Majumder
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shilpee Dutt
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India..
| |
Collapse
|
29
|
Yun S, Cha SS, Kim JH. DJ-1 promotes cell migration by interacting with Mena, the mammalian homolog of Drosophila enabled. Adv Biol Regul 2022; 88:100943. [PMID: 36542983 DOI: 10.1016/j.jbior.2022.100943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/26/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
DJ-1 has gained extensive attention after being identified in 2003 as a protein implicated in the pathogenesis of early-onset Parkinson's disease. Since then, efforts have revealed versatile DJ-1 functions in reactive oxygen species (ROS) control, transcriptional regulation, chaperone function, fertility, and cell transformation. Herein, we report a novel function of DJ-1 in actin cytoskeletal rearrangements. DJ-1 was identified as a new binding partner of Mena, a protein of the Enah/VASP family, and it promoted cancer cell migration by Mena-dependent actin polymerization and filopodia formation. These results suggest a novel molecular mechanism for DJ-1-dependent cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae, 50834, Republic of Korea.
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, 50612, Yangsan, Republic of Korea.
| |
Collapse
|
30
|
Vidal S, Bouzaher YH, El Motiam A, Seoane R, Rivas C. Overview of the regulation of the class IA PI3K/AKT pathway by SUMO. Semin Cell Dev Biol 2022; 132:51-61. [PMID: 34753687 DOI: 10.1016/j.semcdb.2021.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022]
Abstract
The phosphatidylinositol-3-kinase (PI3K)/AKT pathway is a major regulator of metabolism, migration, survival, proliferation, and antiviral immunity. Both an overactivation and an inhibition of the PI3K/AKT pathway are related to different pathologies. Activation of this signaling pathway is tightly controlled through a multistep process and its deregulation can be associated with aberrant post-translational modifications including SUMOylation. Here, we review the complex modulation of the PI3K/AKT pathway by SUMOylation and we discuss its putative incvolvement in human disease.
Collapse
Affiliation(s)
- Santiago Vidal
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain
| | - Yanis Hichem Bouzaher
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain
| | - Ahmed El Motiam
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain; Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health Systems, Department of Ophthalmology and Vision Science, and Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Rocío Seoane
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain
| | - Carmen Rivas
- Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias (IDIS), 15706 Santiago de Compostela, Spain; Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
31
|
Safaeifard F, Goliaei B, Aref AR, Foroughmand-Araabi MH, Goliaei S, Lorch J, Jenkins RW, Barbie DA, Shariatpanahi SP, Rüegg C. Distinct Dynamics of Migratory Response to PD-1 and CTLA-4 Blockade Reveals New Mechanistic Insights for Potential T-Cell Reinvigoration following Immune Checkpoint Blockade. Cells 2022; 11:3534. [PMID: 36428963 PMCID: PMC9688893 DOI: 10.3390/cells11223534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/22/2022] [Accepted: 10/28/2022] [Indexed: 11/10/2022] Open
Abstract
Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1), two clinically relevant targets for the immunotherapy of cancer, are negative regulators of T-cell activation and migration. Optimizing the therapeutic response to CTLA-4 and PD-1 blockade calls for a more comprehensive insight into the coordinated function of these immune regulators. Mathematical modeling can be used to elucidate nonlinear tumor-immune interactions and highlight the underlying mechanisms to tackle the problem. Here, we investigated and statistically characterized the dynamics of T-cell migration as a measure of the functional response to these pathways. We used a previously developed three-dimensional organotypic culture of patient-derived tumor spheroids treated with anti-CTLA-4 and anti-PD-1 antibodies for this purpose. Experiment-based dynamical modeling revealed the delayed kinetics of PD-1 activation, which originates from the distinct characteristics of PD-1 and CTLA-4 regulation, and followed through with the modification of their contributions to immune modulation. The simulation results show good agreement with the tumor cell reduction and active immune cell count in each experiment. Our findings demonstrate that while PD-1 activation provokes a more exhaustive intracellular cascade within a mature tumor environment, the time-delayed kinetics of PD-1 activation outweighs its preeminence at the individual cell level and consequently confers a functional dominance to the CTLA-4 checkpoint. The proposed model explains the distinct immunostimulatory pattern of PD-1 and CTLA-4 blockade based on mechanisms involved in the regulation of their expression and may be useful for planning effective treatment schemes targeting PD-1 and CTLA-4 functions.
Collapse
Affiliation(s)
- Fateme Safaeifard
- Laboratory of Biophysics and Molecular Biology, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614411, Iran
| | - Bahram Goliaei
- Laboratory of Biophysics and Molecular Biology, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614411, Iran
| | - Amir R. Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Xsphera Biosciences Inc., Boston, MA 02210, USA
| | | | - Sama Goliaei
- Faculty of New Sciences & Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Jochen Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Russell W. Jenkins
- MassGeneral Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Seyed Peyman Shariatpanahi
- Laboratory of Biophysics and Molecular Biology, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417614411, Iran
| | - Curzio Rüegg
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology, and Immunology, Faculty of Sciences and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
32
|
Shi Q, Zheng L, Na J, Li X, Yang Z, Chen X, Song Y, Li C, Zhou L, Fan Y. Fluid shear stress promotes periodontal ligament cells proliferation via p38-AMOT-YAP. Cell Mol Life Sci 2022; 79:551. [DOI: 10.1007/s00018-022-04591-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022]
|
33
|
Guo L, Wu Z. FOXM1-mediated NUF2 expression confers temozolomide resistance to human glioma cells by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. Neuropathology 2022; 42:430-446. [PMID: 35701983 DOI: 10.1111/neup.12824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Glioma is the most common malignant tumor in the central nervous system and has a high mortality rate. Temozolomide (TMZ) is a widely used chemotherapeutic drug for glioma. NDC80 kinetochore complex (NUF2) is suggested to play a regulatory role in different cancers, but its specific function and mechanism in glioblastoma TMZ resistance remain unknown. NUF2, assessed by reverse transcription quantitative polymerase chain reaction (RT-qPCR), was highly expressed in glioma cell lines. TMZ was used to treat cells to establish a TMZ-resistant cell line. The potential functions of NUF2 in glioma were assessed using cell counting kit-8 (CCK-8) assays, colony formation assays, 5-Ethynyl-2'-deoxyuridine (EdU) assays, flow cytometry, Western blotting, and a tumor xenograft model. The results showed that NUF2 knockdown attenuated malignant phenotypes of TMZ-resistant cells and prevented tumor growth. Mechanistically, as luciferase reporter assays and chromatin immunoprecipitation (ChIP) as showed, Fox transcription factor M1 (FOXM1) had binding sites on the NUF2 promoter. Rescue assays demonstrated that FOXM1 upregulation counteracted the inhibitory effects of NUF2 depletion on the malignancies of TMZ-resistant cells. This study demonstrates that FOXM1-activated NUF2 promotes TMZ to human glioma cells by regulating proliferation, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Liang Guo
- Department of Neurosurgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhangyi Wu
- Department of Neurosurgery, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
34
|
Wang C, Chen J, Kuang Y, Cheng X, Deng M, Jiang Z, Hu X. A novel methylated cation channel TRPM4 inhibited colorectal cancer metastasis through Ca 2+/Calpain-mediated proteolysis of FAK and suppression of PI3K/Akt/mTOR signaling pathway. Int J Biol Sci 2022; 18:5575-5590. [PMID: 36147460 PMCID: PMC9461655 DOI: 10.7150/ijbs.70504] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
Colorectal cancer (CRC) is an aggressive malignancy with poor prognosis. It is imperative to elucidate the potential molecular mechanisms that regulate CRC cell aggressiveness. In present study, the transient receptor potential melastatin 4 (TRPM4), a calcium-activated nonselective cation channel, is downregulated in CRC as a novel methylated tumor suppressor gene (TSG). The reduced mRNA level of TRPM4 is due to the epigenetic methylation of its promoter CpG island (CGI). Moreover, ectopic expression of TRPM4 inhibited tumor growth and metastasis both in vitro and in vivo. Our experiments also demonstrate that TRPM4 restructures the CRC cytoskeleton and activates the Ca2+-mediated calpain pathway through enhancing calcium influx. The western blot analysis shows that the expression of focal adhesion kinase (FAK), a calpain-mediated proteolytic substrate, is markedly suppressed after ectopic overexpression of TRPM4, besides, Akt (also known as protein kinase B, PKB), phosphatidylinositol 3-kinase (PI3K) as well as its central target mTOR have significantly decreased expression accompanied by elevated E-cadherin and restrained matrix metalloproteinases (MMP2/MMP9) expression. The inhibition of protease calpain effectively relieves the retard of FAK/Akt signals and reverses the migration suppression of TRPM4. Taken together, TRPM4, identified as a novel methylated TSG, employs intracellular Ca2+ signals to activate calpain-mediated cleavage of FAK and impede CRC migration and invasion through modulating the PI3K/Akt/mTOR signaling cascade, providing the first evidence that TRPM4 is likely to be a significant biomarker and potential target for CRC therapy.
Collapse
Affiliation(s)
- Chan Wang
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, China.,Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Jiaxin Chen
- Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Yeye Kuang
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, China.,Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Xiaoqing Cheng
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Min Deng
- Department of Pathology, The First People's Hospital of Fuyang, Hangzhou 311400, China
| | - Zhinong Jiang
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, China
| | - Xiaotong Hu
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, Zhejiang, China.,Key Laboratory of Biotherapy of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
35
|
Wang W, Fu C, Lin M, Lu Y, Lian S, Xie X, Zhou G, Li W, Zhang Y, Jia L, Zhong C, Huang M. Fucoxanthin prevents breast cancer metastasis by interrupting circulating tumor cells adhesion and transendothelial migration. Front Pharmacol 2022; 13:960375. [PMID: 36160416 PMCID: PMC9500434 DOI: 10.3389/fphar.2022.960375] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Metastasis is the leading cause of cancer-related death and a critical challenge in improving cancer treatment today. Circulating tumor cells (CTCs) adhesion to and across the vascular endothelium are critical steps in the establishment of micrometastatic foci away from the primary tumor. Therefore, we believe that interrupting CTCs adhesion to endothelium and transendothelial migration may efficiently prevent cancer metastasis. Fucoxanthin (Fx) is an algal carotenoid widely distributed in brown algae, macroalgae, and diatoms. Previous studies have found that Fx has various pharmacological activities, including antidiabetic, antioxidant, anti-inflammatory, anti-obesity, antimalarial, anticancer, and so on. However, it remains unclear whether Fx has a preventive effect on cancer metastasis. Here, we found that Fx interrupts breast cancer cells MCF-7 adhesion to endothelium and transendothelial migration, thus inhibiting CTCs-based pulmonary metastasis in vivo. The hetero-adhesion assay showed that Fx significantly inhibited the expression of inflammatory factor-induced cell adhesion molecules (CAMs) and the resulting adhesion between MCF-7 cells and endothelial cells. The wound-healing and transwell assays showed that Fx significantly inhibited the motility, invasion, and transendothelial migration abilities of MCF-7 cells. However, the same concentration of Fx did not significantly alter the cell viability, cell cycle, apoptosis, and ROS of breast cancer cells, thus excluding the possibility that Fx inhibits MCF-7 cell adhesion and transendothelial migration through cytotoxicity. Mechanistically, Fx inhibits the expression of CAMs on endothelial cells by inhibiting the NF-кB signaling pathway by down-regulating the phosphorylation level of IKK-α/β, IкB-α, and NF-кB p65. Fx inhibits transendothelial migration of MCF-7 cells by inhibiting Epithelial-to-mesenchymal transition (EMT), PI3K/AKT, and FAK/Paxillin signaling pathways. Moreover, we demonstrated that Fx significantly inhibits the formation of lung micrometastatic foci in immunocompetent syngeneic mouse breast cancer metastasis models. We also showed that Fx enhances antitumor immune responses by substantially increasing the subsets of cytotoxic T lymphocytes in the peripheral immune system. This new finding provides a basis for the application of Fx in cancer metastatic chemoprevention and suggests that interruption of the CTCs adhesion to endothelium and transendothelial migration may serve as a new avenue for cancer metastatic chemoprevention.
Collapse
Affiliation(s)
- Weiyu Wang
- Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
| | - Chengbin Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Breast Surgery Institute, Fujian Medical University, Fuzhou, China
| | - Mengting Lin
- Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
| | - Yusheng Lu
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
- *Correspondence: Yusheng Lu, ; Chunlian Zhong, ; Mingqing Huang,
| | - Shu Lian
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
| | - Xiaodong Xie
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
| | - Guiyu Zhou
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
| | - Wulin Li
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
- College of Chemistry, Fuzhou University, Fuzhou, China
| | - Yiping Zhang
- Technical Innovation Center for Utilization of Marine Biological Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Lee Jia
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
| | - Chunlian Zhong
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Material and Chemical Engineering, Minjiang University, Fuzhou, Fujian, China
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China
- *Correspondence: Yusheng Lu, ; Chunlian Zhong, ; Mingqing Huang,
| | - Mingqing Huang
- Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- *Correspondence: Yusheng Lu, ; Chunlian Zhong, ; Mingqing Huang,
| |
Collapse
|
36
|
The In Vitro Effect of Psoralen on Glioma Based on Network Pharmacology and Potential Target Research. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1952891. [PMID: 36065261 PMCID: PMC9440786 DOI: 10.1155/2022/1952891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022]
Abstract
Glioma is an aggressive tumor, currently there is no satisfactory management available. Psoralen, as a natural product, has been found to have an effect of treating cancer in recent years, but its effect on glioma has not been explored. In this study, we investigated the in vitro inhibition effect and potential targets of psoralen on glioma through network pharmacology and in vitro glioma treatment experiments. First, we used network pharmacology to preliminarily predict the 21 core genes of psoralen in the treatment of glioma, including PIK3CA, PIK3CB, PIK3CG, and JAK2. The CCK-8 method was used to detect the effect of psoralen on the proliferation of glioma U87 and U251 cells, and the results showed that psoralen could significantly inhibit the proliferation of U87 and U251 cells. The flow cytometry was used to detect the apoptosis and cell cycle changes, and it was found that psoralen could significantly promote the early apoptosis of U87 and U251 cells and had a significant cycle arrest effect on the two cells. The cell scratch test showed that psoralen could significantly inhibit the migration of U87 and U251 cells. The relative expression levels of PIK3CA, PIK3CB, PIK3CG, and JAK2 were analyzed by Real-time Quantitative polymerase chain reaction (QT-PCR), and the results showed that psoralen could inhibit the gene expression of PIK3CA, PIK3CB, PIK3CG, and JAK2. Later, Western blotting (WB) experiments showed that psoralen could inhibit the protein expressions of PI3K and JAK2. This study has preliminarily explored and verified the antiglioma effect of psoralen in the form of inhibiting cell proliferation and migration, promoting cell apoptosis and organizing cell cycle in vitro. And may play a role by inhibiting the expression of PIK3CA, PIK3CB, PIK3CG, JAK2 gene and PI3K, JAK2 protein, psoralen has become a potential antiglioma drug.
Collapse
|
37
|
Chen Y, Chen C, Fang J, Su K, Yuan Q, Hou H, Xin H, Sun J, Huang C, Li S, Yuan Z, Luo S. Targeting the Akt/PI3K/mTOR signaling pathway for complete eradication of keloid disease by sunitinib. Apoptosis 2022; 27:812-824. [PMID: 35802302 DOI: 10.1007/s10495-022-01744-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2022] [Indexed: 11/24/2022]
Abstract
Keloid disease is a nodular lesion, tumor-like but not cancerous, and characterized of excessive proliferation of fibroblasts and deposition of extracellular matrix (ECM) components. This condition often causes itching, pain and cosmetic disfigurement, significantly reducing patient quality of life. To date, no universally effective therapies are available, possibly due to inadequate understanding of keloid pathogenesis. As an oral small-molecule inhibitor of certain tyrosine kinase receptors, sunitinib has shown significant therapeutic effects in renal cell carcinoma (RCC) and gastrointestinal stromal tumor (GIST). However, it has never been tested if keloid therapy can be effective for the management of keloids. This study thus aims to explore the potential of sunitinib for keloid treatment. Keloid-derived fibroblasts (KFs) were successfully isolated and demonstrated proliferative advantage to normal skin-derived fibroblasts (NFs). Additionally, sunitinib showed specific cytotoxicity and inhibition of invasion, and induced cell cycle arrest and significant apoptosis in KFs. These effects were accompanied by complete suppression of ECM component expression, including collagen types 1 and 3, upregulation of autophagy-associated LC3B and significant suppression of the Akt/PI3K/mTOR pathway. Moreover, a keloid explant culture model was successfully established and used to test the therapeutic efficacy of sunitinib on keloid formation in nude mice. Sunitinib was found to induce complete regression of keloid explant fragments in nude mice, showing significantly higher therapeutic efficacy than the most commonly used intralesional drug triamcinolone acetonide (TAC). These data suggest that sunitinib effectively inhibits keloid development through suppression of the Akt/PI3K/mTOR pathway and thus can be potentially developed as a monotherapy or combination therapy for the effective treatment of keloid disease.
Collapse
Affiliation(s)
- Yiqing Chen
- The Second School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China.,Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China
| | - Chunlin Chen
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China
| | - Junren Fang
- The Second School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China.,Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China
| | - Kui Su
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Qian Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Huan Hou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Huijuan Xin
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, 510317 Guangzhou, China
| | - Jianwu Sun
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Chaohong Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Shuyi Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, 510006, Guangzhou, China.
| | - Shengkang Luo
- The Second School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, China. .,Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 510317, Guangzhou, China.
| |
Collapse
|
38
|
Targeting Acid Ceramidase Inhibits Glioblastoma Cell Migration through Decreased AKT Signaling. Cells 2022; 11:cells11121873. [PMID: 35741006 PMCID: PMC9221433 DOI: 10.3390/cells11121873] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 01/12/2023] Open
Abstract
Glioblastoma (GBM) remains one of the most aggressive cancers, partially due to its ability to migrate into the surrounding brain. The sphingolipid balance, or the balance between ceramides and sphingosine-1-phosphate, contributes to the ability of GBM cells to migrate or invade. Of the ceramidases which hydrolyze ceramides, acid ceramidase (ASAH1) is highly expressed in GBM samples compared to non-tumor brain. ASAH1 expression also correlates with genes associated with migration and focal adhesion. To understand the role of ASAH1 in GBM migration, we utilized shRNA knockdown and observed decreased migration that did not depend upon changes in growth. Next, we inhibited ASAH1 using carmofur, a clinically utilized small molecule inhibitor. Inhibition of ASAH1 by carmofur blocks in vitro migration of U251 (GBM cell line) and GBM cells derived from patient-derived xenografts (PDXs). RNA-sequencing suggested roles for carmofur in MAPK and AKT signaling. We found that carmofur treatment decreases phosphorylation of AKT, but not of MAPK. The decrease in AKT phosphorylation was confirmed by shRNA knockdown of ASAH1. Our findings substantiate ASAH1 inhibition using carmofur as a potential clinically relevant treatment to advance GBM therapeutics, particularly due to its impact on migration.
Collapse
|
39
|
Ju Z, Thomas TN, Chiu YJ, Yamanouchi S, Yoshida Y, Abe JI, Takahashi A, Wang J, Fujiwara K, Hada M. Adaptation and Changes in Actin Dynamics and Cell Motility as Early Responses of Cultured Mammalian Cells to Altered Gravitational Vector. Int J Mol Sci 2022; 23:6127. [PMID: 35682810 PMCID: PMC9181735 DOI: 10.3390/ijms23116127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
Cultured mammalian cells have been shown to respond to microgravity (μG), but the molecular mechanism is still unknown. The study we report here is focused on molecular and cellular events that occur within a short period of time, which may be related to gravity sensing by cells. Our assumption is that the gravity-sensing mechanism is activated as soon as cells are exposed to any new gravitational environment. To study the molecular events, we exposed cells to simulated μG (SμG) for 15 min, 30 min, 1 h, 2 h, 4 h, and 8 h using a three-dimensional clinostat and made cell lysates, which were then analyzed by reverse phase protein arrays (RPPAs) using a panel of 453 different antibodies. By comparing the RPPA data from cells cultured at 1G with those of cells under SμG, we identified a total of 35 proteomic changes in the SμG samples and found that 20 of these changes took place, mostly transiently, within 30 min. In the 4 h and 8 h samples, there were only two RPPA changes, suggesting that the physiology of these cells is practically indistinguishable from that of cells cultured at 1 G. Among the proteins involved in the early proteomic changes were those that regulate cell motility and cytoskeletal organization. To see whether changes in gravitational environment indeed activate cell motility, we flipped the culture dish upside down (directional change in gravity vector) and studied cell migration and actin cytoskeletal organization. We found that compared with cells grown right-side up, upside-down cells transiently lost stress fibers and rapidly developed lamellipodia, which was supported by increased activity of Ras-related C3 botulinum toxin substrate 1 (Rac1). The upside-down cells also increased their migratory activity. It is possible that these early molecular and cellular events play roles in gravity sensing by mammalian cells. Our study also indicated that these early responses are transient, suggesting that cells appear to adapt physiologically to a new gravitational environment.
Collapse
Affiliation(s)
- Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Tamlyn N. Thomas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.N.T.); (J.-i.A.)
- Aab Cardiovascular Research Institute, University of Rochester Medical School, Rochester, NY 14642, USA;
| | - Yi-Jen Chiu
- Aab Cardiovascular Research Institute, University of Rochester Medical School, Rochester, NY 14642, USA;
| | - Sakuya Yamanouchi
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (S.Y.); (Y.Y.); (A.T.)
| | - Yukari Yoshida
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (S.Y.); (Y.Y.); (A.T.)
| | - Jun-ichi Abe
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.N.T.); (J.-i.A.)
| | - Akihisa Takahashi
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (S.Y.); (Y.Y.); (A.T.)
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Keigi Fujiwara
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.N.T.); (J.-i.A.)
| | - Megumi Hada
- Radiation Institute for Science & Engineering, Prairie View A&M University, Prairie View, TX 77446, USA;
| |
Collapse
|
40
|
Umetsu D. Cell mechanics and cell-cell recognition controls by Toll-like receptors in tissue morphogenesis and homeostasis. Fly (Austin) 2022; 16:233-247. [PMID: 35579305 PMCID: PMC9116419 DOI: 10.1080/19336934.2022.2074783] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Signal transduction by the Toll-like receptors (TLRs) is conserved and essential for innate immunity in metazoans. The founding member of the TLR family, Drosophila Toll-1, was initially identified for its role in dorsoventral axis formation in early embryogenesis. The Drosophila genome encodes nine TLRs that display dynamic expression patterns during development, suggesting their involvement in tissue morphogenesis and homeostasis. Recent progress on the developmental functions of TLRs beyond dorsoventral patterning has revealed not only their diverse functions in various biological processes, but also unprecedented molecular mechanisms in directly regulating cell mechanics and cell-cell recognition independent of the canonical signal transduction pathway involving transcriptional regulation of target genes. In this review, I feature and discuss the non-immune functions of TLRs in the control of epithelial tissue homeostasis, tissue morphogenesis, and cell-cell recognition between cell populations with different cell identities.
Collapse
Affiliation(s)
- Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
41
|
Sumislawski P, Rotermund R, Klose S, Lautenbach A, Wefers AK, Soltwedel C, Mohammadi B, Jacobsen F, Mawrin C, Flitsch J, Saeger W. ACTH-secreting pituitary carcinoma with TP53, NF1, ATRX and PTEN mutations Case report and review of the literature. Endocrine 2022; 76:228-236. [PMID: 35171439 PMCID: PMC8986667 DOI: 10.1007/s12020-021-02954-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/14/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Piotr Sumislawski
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr.52, 20246, Hamburg, Germany
| | - Roman Rotermund
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr.52, 20246, Hamburg, Germany
| | - Silke Klose
- Department of Internal Medicine/Endocrinology, Otto von Guericke Universität Magdeburg, Magdeburg, Germany
| | - Anne Lautenbach
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika K Wefers
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Celina Soltwedel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistr, Hamburg, Germany
| | - Christian Mawrin
- Institute of Neuropathology, University of Magdeburg, Magdeburg, Germany
| | - Jörg Flitsch
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistr.52, 20246, Hamburg, Germany
| | - Wolfgang Saeger
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
42
|
PIK3CA Gene Mutations in HNSCC: Systematic Review and Correlations with HPV Status and Patient Survival. Cancers (Basel) 2022; 14:cancers14051286. [PMID: 35267596 PMCID: PMC8909011 DOI: 10.3390/cancers14051286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/24/2022] Open
Abstract
PIK3CA mutations are believed to contribute to the pathogenesis of human papillomavirus (HPV)-associated head and neck squamous cell carcinomas (HNSCC). This study aims to establish the frequency of PIK3CA mutations in a Portuguese HNSCC cohort and to determine their association with the HPV status and patient survival. A meta-analysis of scientific literature also revealed widely different mutation rates in cohorts from different world regions and a trend towards improved prognosis among patients with PIK3CA mutations. DNA samples were available from 95 patients diagnosed with HNSCC at the Portuguese Institute of Oncology in Lisbon between 2010 and 2019. HPV status was established based on viral DNA detected using real-time PCR. The evaluation of PIK3CA gene mutations was performed by real-time PCR for four mutations (H1047L; E542K, E545K, and E545D). Thirty-seven cases were found to harbour PIK3CA mutations (39%), with the E545D mutation (73%) more frequently detected. There were no significant associations between the mutational status and HPV status (74% WT and 68% MUT were HPV (+); p = 0.489) or overall survival (OS) (3-year OS: WT 54% and MUT 65%; p = 0.090). HPV status was the only factor significantly associated with both OS and disease-free survival (DFS), with HPV (+) patients having consistently better outcomes (3-year OS: HPV (+) 65% and HPV (-) 36%; p = 0.007; DFS HPV (+) 83% and HPV (-) 43%; p = 0.001). There was a statistically significant interaction effect between HPV status and PIK3CA mutation regarding DFS (Interaction test: p = 0.026). In HPV (+) patients, PIK3CA wild-type is associated with a significant 4.64 times increase in the hazard of recurrence or death (HR = 4.64; 95% CI 1.02-20.99; p = 0.047). Overall, PIK3CA gene mutations are present in a large number of patients and may help define patient subsets who can benefit from therapies targeting the PI3K pathway. The systematic assessment of PIK3CA gene mutations in HNSCC patients will require further methodological standardisation.
Collapse
|
43
|
Weiss F, Lauffenburger D, Friedl P. Towards targeting of shared mechanisms of cancer metastasis and therapy resistance. Nat Rev Cancer 2022; 22:157-173. [PMID: 35013601 PMCID: PMC10399972 DOI: 10.1038/s41568-021-00427-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 02/07/2023]
Abstract
Resistance to therapeutic treatment and metastatic progression jointly determine a fatal outcome of cancer. Cancer metastasis and therapeutic resistance are traditionally studied as separate fields using non-overlapping strategies. However, emerging evidence, including from in vivo imaging and in vitro organotypic culture, now suggests that both programmes cooperate and reinforce each other in the invasion niche and persist upon metastatic evasion. As a consequence, cancer cell subpopulations exhibiting metastatic invasion undergo multistep reprogramming that - beyond migration signalling - supports repair programmes, anti-apoptosis processes, metabolic adaptation, stemness and survival. Shared metastasis and therapy resistance signalling are mediated by multiple mechanisms, such as engagement of integrins and other context receptors, cell-cell communication, stress responses and metabolic reprogramming, which cooperate with effects elicited by autocrine and paracrine chemokine and growth factor cues present in the activated tumour microenvironment. These signals empower metastatic cells to cope with therapeutic assault and survive. Identifying nodes shared in metastasis and therapy resistance signalling networks should offer new opportunities to improve anticancer therapy beyond current strategies, to eliminate both nodular lesions and cells in metastatic transit.
Collapse
Affiliation(s)
- Felix Weiss
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, Netherlands
| | - Douglas Lauffenburger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter Friedl
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, Netherlands.
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Genomics Center, Utrecht, Netherlands.
| |
Collapse
|
44
|
Yerramilli VS, Ross AH, Scarlata S, Gericke A. IQGAP1 scaffolding links phosphoinositide kinases to cytoskeletal reorganization. Biophys J 2022; 121:793-807. [PMID: 35077666 PMCID: PMC8943696 DOI: 10.1016/j.bpj.2022.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/24/2021] [Accepted: 01/21/2022] [Indexed: 11/02/2022] Open
Abstract
IQGAP1 is a multidomain scaffold protein that coordinates the direction and impact of multiple signaling pathways by scaffolding its various binding partners. However, the spatial and temporal resolution of IQGAP1 scaffolding remains unclear. Here, we use fluorescence imaging and correlation methods that allow for real-time live-cell changes in IQGAP1 localization and complex formation during signaling. We find that IQGAP1 and PIPKIγ interact on both the plasma membrane and in cytosol. Epidermal growth factor (EGF) stimulation, which can initiate cytoskeletal changes, drives the movement of the cytosolic pool toward the plasma membrane to promote cytoskeletal changes. We also observe that a significant population of cytosolic IQGAP1-PIPKIγ complexes localize to early endosomes, and in some instances form aggregated clusters which become highly mobile upon EGF stimulation. Our imaging studies show that PIPKIγ and PI3K bind simultaneously to IQGAP1, which may accelerate conversion of PI4P to PI(3,4,5)P3 that is required for cytoskeletal changes. Additionally, we find that IQGAP1 is responsible for PIPKIγ association with two proteins associated with cytoskeletal changes, talin and Cdc42, during EGF stimulation. These results directly show that IQGAP1 provides a physical link between phosphoinositides (through PIPKIγ), focal adhesion formation (through talin), and cytoskeletal reorganization (through Cdc42) upon EGF stimulation. Taken together, our results support the importance of IQGAP1 in regulating cell migration by linking phosphoinositide lipid signaling with cytoskeletal reorganization.
Collapse
Affiliation(s)
- V. Siddartha Yerramilli
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Alonzo H. Ross
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts,Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Arne Gericke
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts.
| |
Collapse
|
45
|
Kilbas PO, Can ND, Kizilboga T, Ezberci F, Doganay HL, Arisan ED, Dinler Doganay G. CRISPR/Cas9-mediated Bag-1 knockout increased mesenchymal characteristics of MCF-7 cells via Akt hyperactivation-mediated actin cytoskeleton remodeling. PLoS One 2022; 17:e0261062. [PMID: 34995286 PMCID: PMC8741009 DOI: 10.1371/journal.pone.0261062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/23/2021] [Indexed: 11/18/2022] Open
Abstract
Bag-1 protein is a crucial target in cancer to increase the survival and proliferation of cells. The Bag-1 expression is significantly upregulated in primary and metastatic cancer patients compared to normal breast tissue. Overexpression of Bag-1 decreases the efficiency of conventional chemotherapeutic drugs, whereas Bag-1 silencing enhances the apoptotic efficiency of therapeutics, mostly in hormone-positive breast cancer subtypes. In this study, we generated stable Bag-1 knockout (KO) MCF-7 breast cancer cells to monitor stress-mediated cellular alterations in comparison to wild type (wt) and Bag-1 overexpressing (Bag-1 OE) MCF-7 cells. Validation and characterization studies of Bag-1 KO cells showed different cellular morphology with hyperactive Akt signaling, which caused stress-mediated actin reorganization, focal adhesion decrease and led to mesenchymal characteristics in MCF-7 cells. A potent Akt inhibitor, MK-2206, suppressed mesenchymal transition in Bag-1 KO cells. Similar results were obtained following the recovery of Bag-1 isoforms (Bag-1S, M, or L) in Bag-1 KO cells. The findings of this study emphasized that Bag-1 is a mediator of actin-mediated cytoskeleton organization through regulating Akt activation.
Collapse
Affiliation(s)
- Pelin Ozfiliz Kilbas
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Istanbul, Turkey
| | - Nisan Denizce Can
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
| | - Tugba Kizilboga
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
| | - Fikret Ezberci
- Department of General Surgery, Umraniye Teaching and Research Hospital, Istanbul, Turkey
| | - Hamdi Levent Doganay
- Genomic Laboratory (GLAB), Umraniye Teaching And Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Elif Damla Arisan
- Institute of Biotechnology, Gebze Technical University, Kocaeli, Turkey
- * E-mail: (EDA); (GDD)
| | - Gizem Dinler Doganay
- Department of Molecular Biology Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
- * E-mail: (EDA); (GDD)
| |
Collapse
|
46
|
D’Aloia A, Arrigoni E, Costa B, Berruti G, Martegani E, Sacco E, Ceriani M. RalGPS2 Interacts with Akt and PDK1 Promoting Tunneling Nanotubes Formation in Bladder Cancer and Kidney Cells Microenvironment. Cancers (Basel) 2021; 13:cancers13246330. [PMID: 34944949 PMCID: PMC8699646 DOI: 10.3390/cancers13246330] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Cell-to-cell communication in the tumor microenvironment is a crucial process to orchestrate the different components of the tumoral infrastructure. Among the mechanisms of cellular interplay in cancer cells, tunneling nanotubes (TNTs) are dynamic connections that play an important role. The mechanism of the formation of TNTs among cells and the molecules involved in the process remain to be elucidated. In this study, we analyze several bladder cancer cell lines, representative of tumors at different stages and grades. We demonstrate that TNTs are formed only by mid or high-stage cell lines that show muscle-invasive properties and that they actively transport mitochondria and proteins. The formation of TNTs is triggered by stressful conditions and starts with the assembly of a specific multimolecular complex. In this study, we characterize some of the protein components of the TNTs complex, as they are potential novel molecular targets for future therapies aimed at counteracting tumor progression. Abstract RalGPS2 is a Ras-independent Guanine Nucleotide Exchange Factor for RalA GTPase that is involved in several cellular processes, including cytoskeletal organization. Previously, we demonstrated that RalGPS2 also plays a role in the formation of tunneling nanotubes (TNTs) in bladder cancer 5637 cells. In particular, TNTs are a novel mechanism of cell–cell communication in the tumor microenvironment, playing a central role in cancer progression and metastasis formation. However, the molecular mechanisms involved in TNTs formation still need to be fully elucidated. Here we demonstrate that mid and high-stage bladder cancer cell lines have functional TNTs, which can transfer mitochondria. Moreover, using confocal fluorescence time-lapse microscopy, we show in 5637 cells that TNTs mediate the trafficking of RalA protein and transmembrane MHC class III protein leukocyte-specific transcript 1 (LST1). Furthermore, we show that RalGPS2 is essential for nanotubes generation, and stress conditions boost its expression both in 5637 and HEK293 cell lines. Finally, we prove that RalGPS2 interacts with Akt and PDK1, in addition to LST1 and RalA, leading to the formation of a complex that promotes nanotubes formation. In conclusion, our findings suggest that in the tumor microenvironment, RalGPS2 orchestrates the assembly of multimolecular complexes that drive the formation of TNTs.
Collapse
Affiliation(s)
- Alessia D’Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Edoardo Arrigoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Barbara Costa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
| | - Giovanna Berruti
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy;
| | - Enzo Martegani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, 20126 Milan, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy; (A.D.); (E.A.); (B.C.); (E.M.); (E.S.)
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Piazza dell’Ateneo Nuovo 1, 20126 Milano, Italy
- Correspondence: ; Tel.: +39-0264483544
| |
Collapse
|
47
|
Upadhyay P, Ghosh A, Basu A, Pranati PA, Gupta P, Das S, Sarker S, Bhattacharjee M, Bhattacharya S, Ghosh S, Chattopadhyay S, Adhikary A. Delivery of gefitinib in synergism with thymoquinone via transferrin-conjugated nanoparticle sensitizes gefitinib-resistant non-small cell lung carcinoma to control metastasis and stemness. Biomater Sci 2021; 9:8285-8312. [PMID: 34766965 DOI: 10.1039/d1bm01148k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Epidermal growth factor receptor (EGFR) normally over-expresses in non-small cell lung cancer (NSCLC) cells. Its mutations act as oncogenic drivers in the cellular signal transduction pathway, and induce the downstream activation of numerous key cellular events involved in cellular proliferation and survival. EGFR tyrosine kinase inhibitors (EGFR-TK inhibitors), such as gefitinib and erlotinib, have been used for a long time in the treatment of NSCLC. However, they fail to overcome the EGFR-TK mutation due to the acquisition of drug resistance. It is strongly believed that the epithelial-to-mesenchymal transition (EMT) is a key player for acquired resistance and consequent limitation of the clinical efficiency of EGFR-TKIs. Therefore, a new strategy needs to be developed to overcome the resistance in NSCLC. In this current study, we have disclosed for the first time the efficiency of transferrin-modified PLGA-thymoquinone-nanoparticles in combination with gefitinib (NP-dual-1, NP-dual-2 and NP-dual-3) towards gefitinib-resistant A549 cells. The gefitinib-resistant A549 cells (A549/GR) showed 12.3-fold more resistance to gefitinib in comparison to non-resistant A549 cells. The phenotypic alteration resembling spindle-cell shape and increased pseudopodia integuments featured the EMT phenomena in A549/GR cells. EMT in A549/GR was later coupled with the loss of Ecad and expansion of Ncad, along with upregulated vimentin expression, as compared to the control A549 cells. Moreover, the invasive nature and migration potential are more amplified in A549/GR cells. Pre-incubation of A549 cells with TGFβ1 also initiated EMT, leading to drug resistance. Conversely, treatment of A549 or A549/GR cells with NP-dual-3 effectively retrieved the sensitivity to gefitinib, restricted the EMT phenomenon, and impaired the TGFβ1-induced EMT. On unveiling the underlying mechanism of therapeutic action, we found that STAT3 and miR-21 were individually overexpressed in the A549/GR cells by transfection, and followed by treatment with NP-dual-3. Simultaneously, NP-dual-3 fragmented HIF1-α induced EMT in A549/GR cells and reduced the CSCs markers, viz., Oct-4, Sox-2, Nanog, and Aldh1. These data are self-sufficient to suggest that NP-dual-3 re-sensitizes the drug-resistant A549/GR cells to gefitinib, possibly by retrieving MET phenomena via modulation of STAT3/mir-21/Akt/PTEN/HIF1-α axis. Thus, TQ nanoparticles combined with TKI gefitinib may provide an effective platform to treat NSCLC.
Collapse
Affiliation(s)
- Priyanka Upadhyay
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Avijit Ghosh
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Arijita Basu
- Department of Polymer Science and Technology, University of Calcutta, 92 Acharya Prafulla ChandraRoad, Kolkata-700009, West Bengal, India
| | - P A Pranati
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Payal Gupta
- Department of Physiology, University of Calcutta, 92 Acharya Prafulla Chandra Road, Kolkata-700009, West Bengal, India
| | - Shaswati Das
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Sushmita Sarker
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Mousumi Bhattacharjee
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Saurav Bhattacharya
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| | - Swatilekha Ghosh
- Amity Institute of Biotechnology, Amity University, Rajarhat, New Town, Kolkata-700156, West Bengal, India
| | - Sreya Chattopadhyay
- Department of Physiology, University of Calcutta, 92 Acharya Prafulla Chandra Road, Kolkata-700009, West Bengal, India
| | - Arghya Adhikary
- Center for Research in Nanoscience and Nanotechnology, Technology Campus, University of Calcutta, JD-2, Sector-III, Salt Lake, Kolkata-700106, West Bengal, India.
| |
Collapse
|
48
|
Zhao YT, Fallas JA, Saini S, Ueda G, Somasundaram L, Zhou Z, Xavier Raj I, Xu C, Carter L, Wrenn S, Mathieu J, Sellers DL, Baker D, Ruohola-Baker H. F-domain valency determines outcome of signaling through the angiopoietin pathway. EMBO Rep 2021; 22:e53471. [PMID: 34698433 DOI: 10.15252/embr.202153471] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
Angiopoietins 1 and 2 (Ang1 and Ang2) regulate angiogenesis through their similar F-domains by activating Tie2 receptors on endothelial cells. Despite the similarity in the underlying receptor-binding interaction, the two angiopoietins have opposite effects: Ang1 induces phosphorylation of AKT, strengthens cell-cell junctions, and enhances endothelial cell survival while Ang2 can antagonize these effects, depending on cellular context. To investigate the molecular basis for the opposing effects, we examined the phenotypes of a series of computationally designed protein scaffolds presenting the Ang1 F-domain in a wide range of valencies and geometries. We find two broad phenotypic classes distinguished by the number of presented F-domains: Scaffolds presenting 3 or 4 F-domains have Ang2-like activity, upregulating pFAK and pERK but not pAKT, while scaffolds presenting 6, 8, 12, 30, or 60 F-domains have Ang1-like activity, upregulating pAKT and inducing migration and vascular stability. The scaffolds with 6 or more F-domains display super-agonist activity, producing stronger phenotypes at lower concentrations than Ang1. Tie2 super-agonist nanoparticles reduced blood extravasation and improved blood-brain barrier integrity four days after a controlled cortical impact injury.
Collapse
Affiliation(s)
- Yan Ting Zhao
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
| | - Jorge A Fallas
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Shally Saini
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - George Ueda
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Logeshwaran Somasundaram
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Ziben Zhou
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Infencia Xavier Raj
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Chunfu Xu
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samuel Wrenn
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Julie Mathieu
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Drew L Sellers
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Protein Design, University of Washington, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
49
|
Lv J, Zhang S, Liu Y, Li C, Guo T, Zhang S, Li Z, Jiao Z, Sun H, Zhang Y, Xu L. NR2F1-AS1/miR-190a/PHLDB2 Induces the Epithelial-Mesenchymal Transformation Process in Gastric Cancer by Promoting Phosphorylation of AKT3. Front Cell Dev Biol 2021; 9:688949. [PMID: 34746118 PMCID: PMC8569557 DOI: 10.3389/fcell.2021.688949] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/07/2021] [Indexed: 01/09/2023] Open
Abstract
The median survival time of patients with advanced gastric cancer (GC) who received radiotherapy and chemotherapy was <1 year. Epithelial-mesenchymal transformation (EMT) gives GC cells the ability to invade, which is an essential biological mechanism in the progression of GC. The long non-coding RNA (lncRNA)-based competitive endogenous RNA (ceRNA) system has been shown to play a key role in the GC-related EMT process. Although the AKT pathway is essential for EMT in GC, the relationship between AKT3 subtypes and EMT in GC is unclear. Here, we evaluated the underlying mechanism of ceRNA involving NR2F1-AS1/miR-190a/PHLDB2 in inducing EMT by promoting the expression and phosphorylation of AKT3. The results of bioinformatics analysis showed that the expression of NR2F1-AS1/miR-190a/PHLDB2 in GC was positively associated with the pathological features, staging, poor prognosis, and EMT process. We performed cell transfection, qRT-PCR, western blot, cell viability assay, TUNEL assay, Transwell assay, cell morphology observation, and double luciferase assay to confirm the regulation of NR2F1-AS1/miR-190a/PHLDB2 and its effect on EMT transformation. Finally, GSEA and GO/KEGG enrichment analysis identified that PI3K/AKT pathway was positively correlated to NR2F1-AS1/miR-190a/PHLDB2 expression. AKT3 knockout cells were co-transfected with PHLDB2-OE, and the findings revealed that AKT3 expression and phosphorylation were essential for the PHLDB2-mediated EMT process. Thus, our results showed that NR2F1-AS1/miR-190a/PHLDB2 promoted the phosphorylation of AKT3 to induce EMT in GC cells. This study provides a comprehensive understanding of the underlying mechanism involved in the EMT process as well as the identification of new EMT markers.
Collapse
Affiliation(s)
- Jinqi Lv
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Simeng Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Yang Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Tianshu Guo
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Shuairan Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Zenan Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Zihan Jiao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Haina Sun
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Ling Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, China
| |
Collapse
|
50
|
Zheng F, Kelly MR, Ramms DJ, Heintschel ML, Tao K, Tutuncuoglu B, Lee JJ, Ono K, Foussard H, Chen M, Herrington KA, Silva E, Liu S, Chen J, Churas C, Wilson N, Kratz A, Pillich RT, Patel DN, Park J, Kuenzi B, Yu MK, Licon K, Pratt D, Kreisberg JF, Kim M, Swaney DL, Nan X, Fraley SI, Gutkind JS, Krogan NJ, Ideker T. Interpretation of cancer mutations using a multiscale map of protein systems. Science 2021; 374:eabf3067. [PMID: 34591613 PMCID: PMC9126298 DOI: 10.1126/science.abf3067] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A major goal of cancer research is to understand how mutations distributed across diverse genes affect common cellular systems, including multiprotein complexes and assemblies. Two challenges—how to comprehensively map such systems and how to identify which are under mutational selection—have hindered this understanding. Accordingly, we created a comprehensive map of cancer protein systems integrating both new and published multi-omic interaction data at multiple scales of analysis. We then developed a unified statistical model that pinpoints 395 specific systems under mutational selection across 13 cancer types. This map, called NeST (Nested Systems in Tumors), incorporates canonical processes and notable discoveries, including a PIK3CA-actomyosin complex that inhibits phosphatidylinositol 3-kinase signaling and recurrent mutations in collagen complexes that promote tumor proliferation. These systems can be used as clinical biomarkers and implicate a total of 548 genes in cancer evolution and progression. This work shows how disparate tumor mutations converge on protein assemblies at different scales.
Collapse
Affiliation(s)
- Fan Zheng
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Marcus R. Kelly
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Dana J. Ramms
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Marissa L. Heintschel
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Kai Tao
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Beril Tutuncuoglu
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - John J. Lee
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Keiichiro Ono
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Helene Foussard
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Michael Chen
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kari A. Herrington
- Department of Biochemistry and Biophysics Center for Advanced Light Microscopy at UCSF, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Erica Silva
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Sophie Liu
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jing Chen
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Christopher Churas
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicholas Wilson
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Anton Kratz
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Rudolf T. Pillich
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Devin N. Patel
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Jisoo Park
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Brent Kuenzi
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Michael K. Yu
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Katherine Licon
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Dexter Pratt
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jason F. Kreisberg
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| | - Minkyu Kim
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Danielle L. Swaney
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Xiaolin Nan
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - J. Silvio Gutkind
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - Nevan J. Krogan
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Cancer Cell Map Initiative (CCMI), La Jolla and San Francisco, CA, USA
| |
Collapse
|