1
|
Xu L, Sun J, Guo J, Guo S, Li J, Tang Y, Liu X. Transcriptional factor KLF9 overcomes 5-fluorouracil resistance in breast cancer via PTEN-dependent regulation of aerobic glycolysis. J Chemother 2024:1-12. [PMID: 39491797 DOI: 10.1080/1120009x.2024.2421701] [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: 07/23/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
The emergence of resistance to 5-Fluorouracil (5-FU) is a staple in breast cancer chemotherapy. This paper delves into the role of PTEN in breast cancer resistance to 5-FU and examines the underlying molecular pathways. PTEN expression was detected in bioinformatics databases and upstream transcription factors (TFs) were identified. PTEN mRNA and protein levels, aerobic glycolysis proteins, lactate production, glucose consumption, and cell viability were measured. Binding interactions were confirmed, and cell proliferation assessed. In breast cancer cells, PTEN expression was downregulated. PTEN overexpression counteracted 5-FU resistance through the suppression of aerobic glycolysis. KLF9, as a TF upstream of PTEN, enhanced the levels of PTEN. In conclusion, the TF KLF9 inhibits the aerobic glycolysis level of breast cancer cells by up-regulating PTEN expression, thereby reducing their resistance to 5-FU. The discovery of this mechanism provides a new theoretical basis for the treatment of breast cancer.
Collapse
Affiliation(s)
- Liang Xu
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Jing Sun
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Junlan Guo
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Shengnan Guo
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Jiangli Li
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Yijun Tang
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Xiaohui Liu
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| |
Collapse
|
2
|
Calì B, Troiani M, Bressan S, Attanasio G, Merler S, Moscarda V, Mosole S, Ricci E, Guo C, Yuan W, Gallagher L, Lundberg A, Bernett I, Figueiredo I, Arzola RA, Abreut EB, D'Ambrosio M, Bancaro N, Brina D, Zumerle S, Pasquini E, Maddalena M, Lai P, Colucci M, Pernigoni N, Rinaldi A, Minardi D, Morlacco A, Moro FD, Sabbadin M, Galuppini F, Fassan M, Rüschoff JH, Moch H, Rescigno P, Francini E, Saieva C, Modesti M, Theurillat JP, Gillessen S, Wilgenbus P, Graf C, Ruf W, de Bono J, Alimonti A. Coagulation factor X promotes resistance to androgen-deprivation therapy in prostate cancer. Cancer Cell 2024; 42:1676-1692.e11. [PMID: 39303726 DOI: 10.1016/j.ccell.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/13/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
Although hypercoagulability is commonly associated with malignancies, whether coagulation factors directly affect tumor cell proliferation remains unclear. Herein, by performing single-cell RNA sequencing (scRNA-seq) of the prostate tumor microenvironment (TME) of mouse models of castration-resistant prostate cancer (CRPC), we report that immunosuppressive neutrophils (PMN-MDSCs) are a key extra-hepatic source of coagulation factor X (FX). FX activation within the TME enhances androgen-independent tumor growth by activating the protease-activated receptor 2 (PAR2) and the phosphorylation of ERK1/2 in tumor cells. Genetic and pharmacological inhibition of factor Xa (FXa) antagonizes the oncogenic activity of PMN-MDSCs, reduces tumor progression, and synergizes with enzalutamide therapy. Intriguingly, F10high PMN-MDSCs express the surface marker CD84 and CD84 ligation enhances F10 expression. Elevated levels of FX, CD84, and PAR2 in prostate tumors associate with worse survival in CRPC patients. This study provides evidence that FXa directly promotes cancer and highlights additional targets for PMN-MDSCs for cancer therapies.
Collapse
Affiliation(s)
- Bianca Calì
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Martina Troiani
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Silvia Bressan
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
| | - Giuseppe Attanasio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Sara Merler
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Section of Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Viola Moscarda
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Section of Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Simone Mosole
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Elena Ricci
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Christina Guo
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Wei Yuan
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Lewis Gallagher
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Arian Lundberg
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Ilona Bernett
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Ines Figueiredo
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Rydell Alvarez Arzola
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Department of Immunoregulation, Immunology and Immunotherapy Division, Center of Molecular Immunology, La Habana 3GGH+C9G, Cuba
| | - Ernesto Bermudez Abreut
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Department of Immunoregulation, Immunology and Immunotherapy Division, Center of Molecular Immunology, La Habana 3GGH+C9G, Cuba
| | - Mariantonietta D'Ambrosio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Nicolò Bancaro
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Daniela Brina
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Sara Zumerle
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, University of Padova, 35121 Padova, Italy
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Martino Maddalena
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Ping Lai
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Manuel Colucci
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Nicolò Pernigoni
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Davide Minardi
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Alessandro Morlacco
- Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Fabrizio Dal Moro
- Urology Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | - Marianna Sabbadin
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Francesca Galuppini
- Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology Unit, University of Padova, 35121 Padova, Italy
| | - Jan Hendrik Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich (USZ), 8091 Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich (USZ), 8091 Zurich, Switzerland
| | | | - Edoardo Francini
- Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy
| | - Calogero Saieva
- Cancer Risk Factors and Lifestyle Epidemiology Unit - ISPRO, 50139 Florence, Italy
| | - Mikol Modesti
- Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland
| | - Jean-Philippe Theurillat
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland
| | - Silke Gillessen
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland
| | - Petra Wilgenbus
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Claudine Graf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Johann de Bono
- The Institute of Cancer Research, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, CH6900 Lugano, Switzerland; Medical Oncology Unit, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, CH6500 Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, 35129 Padova, Italy; Department of Medicine, University of Padova, 35121 Padova, Italy; Department of Health Sciences and Technology (D-HEST) ETH Zurich, 8092 Zurich, Switzerland.
| |
Collapse
|
3
|
Dunston K, Hunter MI, Johannesen E, Jung JS, Kim TH, Yoo JY, Jeong JW. ERBB2 Targeting Reveals a Significant Suppression of Tumorigenesis in Murine Endometrial Cancer with Pten Mutation. Reprod Sci 2024; 31:2458-2467. [PMID: 38637476 DOI: 10.1007/s43032-024-01546-3] [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: 10/09/2023] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Endometrial cancer is the most common gynecologic malignancy. PTEN is a negative regulator of PI3K signaling and is deficient in > 50% of primary human endometrial cancer. Amplification of ERBB2 promotes tumorigenesis and pathogenesis of several human cancers. However, the effect of ERBB2 targeting has not been studied in endometrial cancer with PTEN mutations. The murine model Pgrcre/+Erbb2f/fPtenf/f (Erbb2d/d Ptend/d) was developed to evaluate the effect of ERBB2 targeted therapy in endometrial cancer with PTEN deficiency. Histopathological and molecular analysis was performed for Ptend/d and Erbb2d/dPtend/d mice. Histopathological analysis revealed that Erbb2d/dPtend/d mice significantly reduced development and progression of endometrial cancer compared to Ptend/d mice. Furthermore, percentage of proliferative cells in Erbb2d/dPtend/d mice revealed anti-tumorigenic effect of Erbb2 ablation compared to Ptend/d mice. Our results demonstrate that Erbb2 ablation reveals a significant suppression of tumorigenesis on endometrial cancer of Ptend/d mice. Our results suggest that Erbb2 functions as an oncogene in endometrial cancer of Ptend/d mice implying that Erbb2 targeting can be used as an effective therapeutic approach for treatment of endometrial cancer with PTEN deficiency to hinder cancer development.
Collapse
Affiliation(s)
- Krystina Dunston
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Mark I Hunter
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Eric Johannesen
- Department of Pathology Medical Science Building (MSB), University of Missouri, Columbia, MO, USA
| | - Jin-Seok Jung
- Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, 1 Yonseidae-gil, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, 1 Yonseidae-gil, Wonju, Gangwon-do, 26493, Republic of Korea.
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, 1030 Hitt Street, Columbia, MO, 65211, USA.
| |
Collapse
|
4
|
Montero-Vallejo R, Maya-Miles D, Ampuero J, Martín F, Romero-Gómez M, Gallego-Durán R. Novel insights into metabolic-associated steatotic liver disease preclinical models. Liver Int 2024; 44:644-662. [PMID: 38291855 DOI: 10.1111/liv.15830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024]
Abstract
Metabolic-associated steatotic liver disease (MASLD) encompasses a wide spectrum of metabolic conditions associated with an excess of fat accumulation in the liver, ranging from simple hepatic steatosis to cirrhosis and hepatocellular carcinoma. Finding appropriate tools to study its development and progression is essential to address essential unmet therapeutic and staging needs. This review discusses advantages and shortcomings of different dietary, chemical and genetic factors that can be used to mimic this disease and its progression in mice from a hepatic and metabolic point of view. Also, this review will highlight some additional factors and considerations that could have a strong impact on the outcomes of our model to end up providing recommendations and a checklist to facilitate the selection of the appropriate MASLD preclinical model based on clinical aims.
Collapse
Affiliation(s)
- Rocío Montero-Vallejo
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
| | - Douglas Maya-Miles
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
| | - Javier Ampuero
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
- Digestive Diseases Unit, Hospital Universitario Virgen Del Rocío, Sevilla, Spain
| | - Franz Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, University Pablo Olavide-University of Seville-CSIC, Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Romero-Gómez
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
- Digestive Diseases Unit, Hospital Universitario Virgen Del Rocío, Sevilla, Spain
| | - Rocío Gallego-Durán
- SeLiver Group, Instituto de Biomedicina de Sevilla/CSIC/Hospital Virgen del Rocío, Sevilla, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Sevilla, Spain
| |
Collapse
|
5
|
Lorenz J, Richter S, Kirstein AS, Kolbig F, Nebe M, Schulze M, Kiess W, Spitzbarth I, Klöting N, Le Duc D, Baschant U, Garten A. Pten knockout in mouse preosteoblasts leads to changes in bone turnover and strength. JBMR Plus 2024; 8:ziad016. [PMID: 38505222 PMCID: PMC10945711 DOI: 10.1093/jbmrpl/ziad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 03/21/2024] Open
Abstract
Bone development and remodeling are controlled by the phosphoinositide-3-kinase (Pi3k) signaling pathway. We investigated the effects of downregulation of phosphatase and tensin homolog (Pten), a negative regulator of Pi3k signaling, in a mouse model of Pten deficiency in preosteoblasts. We aimed to identify mechanisms that are involved in the regulation of bone turnover and are linked to bone disorders. Femora, tibiae, and bone marrow stromal cells (BMSCs) isolated from mice with a conditional deletion of Pten (Pten cKO) in Osterix/Sp7-expressing osteoprogenitor cells were compared to Cre-negative controls. Bone phenotyping was performed by μCT measurements, bone histomorphometry, quantification of bone turnover markers CTX and procollagen type 1 N propeptide (P1NP), and three-point bending test. Proliferation of BMSCs was measured by counting nuclei and Ki-67-stained cells. In vitro, osteogenic differentiation capacity was determined by ALP staining, as well as by detecting gene expression of osteogenic markers. BMSCs from Pten cKO mice were functionally different from control BMSCs. Osteogenic markers were increased in BMSCs derived from Pten cKO mice, while Pten protein expression was lower and Akt phosphorylation was increased. We detected a higher trabecular bone volume and an altered cortical bone morphology in Pten cKO bones with a progressive decrease in bone and tissue mineral density. Pten cKO bones displayed fewer osteoclasts and more osteoblasts (P = .00095) per trabecular bone surface and a higher trabecular bone formation rate. Biomechanical analysis revealed a significantly higher bone strength (P = .00012 for males) and elasticity of Pten cKO femora. On the cellular level, both proliferation and osteogenic differentiation capacity of Pten cKO BMSCs were significantly increased compared to controls. Our findings suggest that Pten knockout in osteoprogenitor cells increases bone stability and elasticity by increasing trabecular bone mass and leads to increased proliferation and osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Judith Lorenz
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
| | - Sandy Richter
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
| | - Anna S Kirstein
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Florentien Kolbig
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
| | - Michèle Nebe
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
| | - Marco Schulze
- Saxon Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Wieland Kiess
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
| | - Ingo Spitzbarth
- Faculty of Veterinary Medicine, Institute of Veterinary Pathology, Leipzig University, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München, Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, Leipzig University, 04103 Leipzig, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, 01309 Dresden, Germany
| | - Antje Garten
- Pediatric Research Center, Leipzig University, University Hospital for Children and Adolescents, Department for Child and Adolescent Medicine, 04103 Leipzig, Germany
| |
Collapse
|
6
|
Mishra AB, Nishank SS. Therapeutic targeting approach on epithelial-mesenchymal plasticity to combat cancer metastasis. Med Oncol 2023; 40:190. [PMID: 37247000 DOI: 10.1007/s12032-023-02049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/06/2023] [Indexed: 05/30/2023]
Abstract
Epithelial-mesenchymal plasticity (EMP) is a process in which epithelial cells lose their characteristics and acquire mesenchymal properties, leading to increased motility and invasiveness, which are key factors in cancer metastasis. Targeting EMP has emerged as a promising therapeutic approach to combat cancer metastasis. Various strategies have been developed to target EMP, including inhibition of key signaling pathways, such as TGF-β, Wnt/β-catenin, and Notch, that regulate EMP, as well as targeting specific transcription factors, such as Snail, Slug, and Twist, that promote EMP. Additionally, targeting the tumor microenvironment, which plays a critical role in promoting EMP, has also shown promise. Several preclinical and clinical studies have demonstrated the efficacy of EMP-targeting therapies in inhibiting cancer metastasis. However, further research is needed to optimize these strategies and improve their clinical efficacy. Overall, therapeutic targeting of EMP represents a promising approach for the development of novel cancer therapies that can effectively inhibit metastasis, a major cause of cancer-related mortality.
Collapse
|
7
|
van Ree JH, Jeganathan KB, Fierro Velasco RO, Zhang C, Can I, Hamada M, Li H, Baker DJ, van Deursen JM. Hyperphosphorylated PTEN exerts oncogenic properties. Nat Commun 2023; 14:2983. [PMID: 37225693 PMCID: PMC10209192 DOI: 10.1038/s41467-023-38740-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
PTEN is a multifaceted tumor suppressor that is highly sensitive to alterations in expression or function. The PTEN C-tail domain, which is rich in phosphorylation sites, has been implicated in PTEN stability, localization, catalytic activity, and protein interactions, but its role in tumorigenesis remains unclear. To address this, we utilized several mouse strains with nonlethal C-tail mutations. Mice homozygous for a deletion that includes S370, S380, T382 and T383 contain low PTEN levels and hyperactive AKT but are not tumor prone. Analysis of mice containing nonphosphorylatable or phosphomimetic versions of S380, a residue hyperphosphorylated in human gastric cancers, reveal that PTEN stability and ability to inhibit PI3K-AKT depends on dynamic phosphorylation-dephosphorylation of this residue. While phosphomimetic S380 drives neoplastic growth in prostate by promoting nuclear accumulation of β-catenin, nonphosphorylatable S380 is not tumorigenic. These data suggest that C-tail hyperphosphorylation creates oncogenic PTEN and is a potential target for anti-cancer therapy.
Collapse
Affiliation(s)
- Janine H van Ree
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Masakazu Hamada
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Langdon CG. Nuclear PTEN's Functions in Suppressing Tumorigenesis: Implications for Rare Cancers. Biomolecules 2023; 13:biom13020259. [PMID: 36830628 PMCID: PMC9953540 DOI: 10.3390/biom13020259] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Phosphatase and tensin homolog (PTEN) encodes a tumor-suppressive phosphatase with both lipid and protein phosphatase activity. The tumor-suppressive functions of PTEN are lost through a variety of mechanisms across a wide spectrum of human malignancies, including several rare cancers that affect pediatric and adult populations. Originally discovered and characterized as a negative regulator of the cytoplasmic, pro-oncogenic phosphoinositide-3-kinase (PI3K) pathway, PTEN is also localized to the nucleus where it can exert tumor-suppressive functions in a PI3K pathway-independent manner. Cancers can usurp the tumor-suppressive functions of PTEN to promote oncogenesis by disrupting homeostatic subcellular PTEN localization. The objective of this review is to describe the changes seen in PTEN subcellular localization during tumorigenesis, how PTEN enters the nucleus, and the spectrum of impacts and consequences arising from disrupted PTEN nuclear localization on tumor promotion. This review will highlight the immediate need in understanding not only the cytoplasmic but also the nuclear functions of PTEN to gain more complete insights into how important PTEN is in preventing human cancers.
Collapse
Affiliation(s)
- Casey G. Langdon
- Department of Pediatrics, Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA; ; Tel.: +1-(843)-792-9289
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
9
|
Gao S, Liu S, Wei W, Qi Y, Meng F. Advances in targeting of miR‑10‑associated lncRNAs/circRNAs for the management of cancer (Review). Oncol Lett 2023; 25:89. [PMID: 36817057 PMCID: PMC9931999 DOI: 10.3892/ol.2023.13675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
With advancements in sequencing technologies, an increasing number of aberrantly expressed long-non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have been identified in various types of cancer. lncRNAs and circRNAs are now well-established tumor-influencing factors in cancer, driving not only tumor proliferation and invasion, but also cancer progression, drug resistance and metastatic recurrence. The majority of lncRNAs and circRNAs influence cancer progression by targeting microRNAs (miRNAs/miRs). miR-10a and miR-10b, key members of the miR-10 family, have been shown to play important regulatory roles in cell proliferation, differentiation to cancer progression, and development. Manual evaluation and grouping according to different types of competing endogenous RNA and tumor was performed. The review outlined the current state of knowledge on the regulation of miR-10 family-related lncRNAs and circRNAs. The involvement of lncRNAs and circRNAs in the biogenesis, maturation and function of malignant tumors through the miR-10 family, and the key gene targets and signaling cascades that lncRNAs and circRNAs regulate through the miR-10 family were summarized. Based on the findings of this review, it can be hypothesized that lncRNAs and circRNAs targeting the miR-10 family may serve as diagnostic/prognostic markers and/or therapeutic targets for the management of cancer.
Collapse
Affiliation(s)
- Shengyu Gao
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China,Department of General Surgery I, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Shuang Liu
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Weiwei Wei
- Department of General Surgery I, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Yanxiu Qi
- Department of General Surgery I, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China
| | - Fanshi Meng
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China,Department of General Surgery I, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154002, P.R. China,Correspondence to: Professor Fanshi Meng, Department of General Surgery I, The First Affiliated Hospital of Jiamusi University, 348 Dexiang Street, Jiamusi, Heilongjiang 154002, P.R. China, E-mail:
| |
Collapse
|
10
|
Mejía-Hernández JO, Keam SP, Saleh R, Muntz F, Fox SB, Byrne D, Kogan A, Pang L, Huynh J, Litchfield C, Caramia F, Lozano G, He H, You JM, Sandhu S, Williams SG, Haupt Y, Haupt S. Modelling aggressive prostate cancers of young men in immune-competent mice, driven by isogenic Trp53 alterations and Pten loss. Cell Death Dis 2022; 13:777. [PMID: 36075907 PMCID: PMC9465983 DOI: 10.1038/s41419-022-05211-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 01/21/2023]
Abstract
Understanding prostate cancer onset and progression in order to rationally treat this disease has been critically limited by a dire lack of relevant pre-clinical animal models. We have generated a set of genetically engineered mice that mimic human prostate cancer, initiated from the gland epithelia. We chose driver gene mutations that are specifically relevant to cancers of young men, where aggressive disease poses accentuated survival risks. An outstanding advantage of our models are their intact repertoires of immune cells. These mice provide invaluable insight into the importance of immune responses in prostate cancer and offer scope for studying treatments, including immunotherapies. Our prostate cancer models strongly support the role of tumour suppressor p53 in functioning to critically restrain the emergence of cancer pathways that drive cell cycle progression; alter metabolism and vasculature to fuel tumour growth; and mediate epithelial to mesenchymal-transition, as vital to invasion. Importantly, we also discovered that the type of p53 alteration dictates the specific immune cell profiles most significantly disrupted, in a temporal manner, with ramifications for disease progression. These new orthotopic mouse models demonstrate that each of the isogenic hotspot p53 amino acid mutations studied (R172H and R245W, the mouse equivalents of human R175H and R248W respectively), drive unique cellular changes affecting pathways of proliferation and immunity. Our findings support the hypothesis that individual p53 mutations confer their own particular oncogenic gain of function in prostate cancer.
Collapse
Affiliation(s)
- Javier Octavio Mejía-Hernández
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,Present Address: Telix Pharmaceuticals Ltd, Melbourne, VIC 3051 Australia
| | - Simon P. Keam
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1135.60000 0001 1512 2287Present Address: CSL Innovation, CSL Ltd, Melbourne, VIC 3052 Australia
| | - Reem Saleh
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Fenella Muntz
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Stephen B. Fox
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Pathology Department, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - David Byrne
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1055.10000000403978434Pathology Department, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Arielle Kogan
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Lokman Pang
- grid.1018.80000 0001 2342 0938Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084 Australia
| | - Jennifer Huynh
- grid.1018.80000 0001 2342 0938Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084 Australia
| | - Cassandra Litchfield
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Franco Caramia
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Guillermina Lozano
- grid.240145.60000 0001 2291 4776Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX USA ,grid.267308.80000 0000 9206 2401University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas, Houston, TX USA
| | - Hua He
- grid.240145.60000 0001 2291 4776Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - James M. You
- grid.267308.80000 0000 9206 2401University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas, Houston, TX USA ,grid.240145.60000 0001 2291 4776Department of Hematopathology, UT MD Anderson Cancer Center, Houston, TX USA
| | - Shahneen Sandhu
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC 3000 Australia
| | - Scott G. Williams
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Division of Radiation Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| | - Ygal Haupt
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,Present Address: Vittail Ltd, Melbourne, VIC 3146 Australia
| | - Sue Haupt
- grid.1055.10000000403978434Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia ,grid.1008.90000 0001 2179 088XSir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000 Australia
| |
Collapse
|
11
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15. [DOI: https:/doi.org/10.3389/fnmol.2022.910543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
|
12
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15:910543. [PMID: 35935338 PMCID: PMC9354928 DOI: 10.3389/fnmol.2022.910543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
Affiliation(s)
- Hao Wu
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Min Wei
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Yuping Li
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Qiang Ma
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Hengzhu Zhang
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
- *Correspondence: Hengzhu Zhang,
| |
Collapse
|
13
|
Vallabhaneni S, Liu J, Morel M, Wang J, DeMayo FJ, Long W. Conditional ERK3 overexpression cooperates with PTEN deletion to promote lung adenocarcinoma formation in mice. Mol Oncol 2021; 16:1184-1199. [PMID: 34719109 PMCID: PMC8895443 DOI: 10.1002/1878-0261.13132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 11/23/2022] Open
Abstract
ERK3, officially known as mitogen‐activated protein kinase 6 (MAPK6), is a poorly studied mitogen‐activated protein kinase (MAPK). Recent studies have revealed the upregulation of ERK3 expression in cancer and suggest an important role for ERK3 in promoting cancer cell growth and invasion in some cancers, in particular lung cancer. However, it is unknown whether ERK3 plays a role in spontaneous tumorigenesis in vivo. To determine the role of ERK3 in lung tumorigenesis, we created a conditional ERK3 transgenic mouse line in which ERK3 transgene expression is controlled by Cre recombinase. By crossing these transgenic mice with a mouse line harboring a lung tissue–specific Cre recombinase transgene driven by a club cell secretory protein gene promoter (CCSP‐iCre), we have found that conditional ERK3 overexpression cooperates with phosphatase and tensin homolog (PTEN) deletion to induce the formation of lung adenocarcinomas (LUADs). Mechanistically, ERK3 overexpression stimulates activating phosphorylations of erb‐b2 receptor tyrosine kinases 2 and 3 (ERBB2 and ERBB3) by upregulating Sp1 transcription factor (SP1)–mediated gene transcription of neuregulin 1 (NRG1), a potent ligand for ERBB2/ERBB3. Our study has revealed a bona fide tumor‐promoting role for ERK3 using genetically engineered mouse models. Together with previous findings showing the roles of ERK3 in cultured cells and in a xenograft lung tumor model, our findings corroborate that ERK3 acts as an oncoprotein in promoting LUAD development and progression.
Collapse
Affiliation(s)
- Sreeram Vallabhaneni
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Jian Liu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, 314400, China.,Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - Marion Morel
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Jixin Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, 314400, China.,Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - Francesco J DeMayo
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park (RTP), NC, USA
| | - Weiwen Long
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| |
Collapse
|
14
|
Liu F, Deng W, Wan Z, Xu D, Chen J, Yang X, Xu J. lncRNA MAGI2-AS3 overexpression had antitumor effect on Hepatic cancer via miRNA-23a-3p/PTEN axis. Food Sci Nutr 2021; 9:2517-2530. [PMID: 34026068 PMCID: PMC8116851 DOI: 10.1002/fsn3.2199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/02/2021] [Accepted: 02/06/2021] [Indexed: 11/16/2022] Open
Abstract
The present study aimed to evaluate the antitumor effects of MAGI2-AS3 and its mechanism in liver cancer. Cancer tissues and adjacent nontumor tissues were collected, and lncRNAs were analyzed via chip assay. The correlation between MAGEI2-AS3 and patient pathology and prognosis was then analyzed. Bel-7402 and Huh-7 cell lines were also used in our study. For the in vitro study, MTT assay, flow cytometry, transwell assay, and wound healing assay were conducted to evaluate hepatic cancer cell (Bel-7402 and Huh-7) proliferation, apoptosis, invasion, and migration. The relative mechanisms were evaluated by Western blot (WB) and cellular immunofluorescence. The correlation among MAGI2-AS3, miRNA-23a-3p, and PTEN was determined by a dual-luciferase reporter assay. The expression of lncRNA MAGI2-AS3 was significantly downregulated in tumor tissues. MAGI2-AS3 expression was closely correlation with HCC patient's clinicopathology and prognosis and prognosis. In the cell experiment, compared with the negative control (NC) group, MAGI2-AS3 overexpression reduced cell proliferation, invasion, and migration and increased cell apoptosis in Bel-7402 and Huh-7 cell lines. However, when Bel-7402 and Huh-7 cells were transfected with miRNA-23a-3p, their biological activities (proliferation, invasion, and migration) were significantly increased. Through WB assay, MAGI2-AS3 could increase PTEN and depress p-AKT and MMP-9 protein expressions via miRNA-23a-3p suppression. The dual-luciferase reporter assay revealed that MAGI2-AS3 directly targeted miRNA-23a-3p and that miRNA-23a-3p could target PTEN. MAGI2-AS3 might be a potential therapeutic target for liver cancer owing to its regulation by the miRNA-23a-3p/PTEN axis.
Collapse
Affiliation(s)
- Fei Liu
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Wenwen Deng
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Zhenda Wan
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Dajin Xu
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Jun Chen
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Xin Yang
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| | - Jianhua Xu
- Jiangxi Province Hospital of Integrated Chinese and Western MedicineNanchangChina
| |
Collapse
|
15
|
Bastos DC, Ribeiro CF, Ahearn T, Nascimento J, Pakula H, Clohessy J, Mucci L, Roberts T, Zanata SM, Zadra G, Loda M. Genetic ablation of FASN attenuates the invasive potential of prostate cancer driven by Pten loss. J Pathol 2020; 253:292-303. [PMID: 33166087 PMCID: PMC7898611 DOI: 10.1002/path.5587] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 09/23/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022]
Abstract
Loss of the tumor suppressor gene Pten in murine prostate recapitulates human carcinogenesis and causes stromal proliferation surrounding murine prostate intraepithelial neoplasia (mPIN), which is reactive to microinvasion. In turn, invasion has been shown to be regulated in part by de novo fatty acid synthesis in prostate cancer. We therefore investigated the effects of genetic ablation of Fasn on invasive potential in prostate‐specific Pten knockout mice. Combined genetic ablation of Fasn and Pten reduced the weight and volume of all the prostate lobes when compared to single knockouts. The stromal reaction to microinvasion and the cell proliferation that typically occurs in Pten knockout were largely abolished by Fasn knockout. To verify that Fasn knockout indeed results in decreased invasive potential, we show that genetic ablation and pharmacologic inhibition of FASN in prostate cancer cells significantly inhibit cellular motility and invasion. Finally, combined loss of PTEN with FASN overexpression was associated with lethality as assessed in 660 prostate cancer patients with 14.2 years of median follow‐up. Taken together, these findings show that de novo lipogenesis contributes to the aggressive phenotype induced by Pten loss in murine prostate and targeting Fasn may reduce the invasive potential of prostate cancer driven by Pten loss. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Débora C Bastos
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Oral Biosciences, University of Campinas, Piracicaba, Brazil
| | - Caroline F Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Thomas Ahearn
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jéssica Nascimento
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hubert Pakula
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY, USA
| | - John Clohessy
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lorelei Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Thomas Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Silvio M Zanata
- Departments of Basic Pathology and Cell Biology, Universidade Federal do Paraná, Curitiba, Brazil
| | - Giorgia Zadra
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY, USA.,New York Genome Center, New York, NY, USA.,The Broad Institute, Cambridge, MA, USA
| |
Collapse
|
16
|
Butler LM, Perone Y, Dehairs J, Lupien LE, de Laat V, Talebi A, Loda M, Kinlaw WB, Swinnen JV. Lipids and cancer: Emerging roles in pathogenesis, diagnosis and therapeutic intervention. Adv Drug Deliv Rev 2020; 159:245-293. [PMID: 32711004 PMCID: PMC7736102 DOI: 10.1016/j.addr.2020.07.013] [Citation(s) in RCA: 303] [Impact Index Per Article: 75.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/02/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
With the advent of effective tools to study lipids, including mass spectrometry-based lipidomics, lipids are emerging as central players in cancer biology. Lipids function as essential building blocks for membranes, serve as fuel to drive energy-demanding processes and play a key role as signaling molecules and as regulators of numerous cellular functions. Not unexpectedly, cancer cells, as well as other cell types in the tumor microenvironment, exploit various ways to acquire lipids and extensively rewire their metabolism as part of a plastic and context-dependent metabolic reprogramming that is driven by both oncogenic and environmental cues. The resulting changes in the fate and composition of lipids help cancer cells to thrive in a changing microenvironment by supporting key oncogenic functions and cancer hallmarks, including cellular energetics, promoting feedforward oncogenic signaling, resisting oxidative and other stresses, regulating intercellular communication and immune responses. Supported by the close connection between altered lipid metabolism and the pathogenic process, specific lipid profiles are emerging as unique disease biomarkers, with diagnostic, prognostic and predictive potential. Multiple preclinical studies illustrate the translational promise of exploiting lipid metabolism in cancer, and critically, have shown context dependent actionable vulnerabilities that can be rationally targeted, particularly in combinatorial approaches. Moreover, lipids themselves can be used as membrane disrupting agents or as key components of nanocarriers of various therapeutics. With a number of preclinical compounds and strategies that are approaching clinical trials, we are at the doorstep of exploiting a hitherto underappreciated hallmark of cancer and promising target in the oncologist's strategy to combat cancer.
Collapse
Affiliation(s)
- Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, SA 5005, Australia; South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Ylenia Perone
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine, London, UK
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Leslie E Lupien
- Program in Experimental and Molecular Medicine, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 037560, USA
| | - Vincent de Laat
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Massimo Loda
- Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - William B Kinlaw
- The Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven Cancer Institute, 3000 Leuven, Belgium.
| |
Collapse
|