1
|
Marrone MT, Reuss JE, Crawford A, Neelon B, Liu JO, Brahmer JR, Platz EA. Statin Use With Immune Checkpoint Inhibitors and Survival in Nonsmall Cell Lung Cancer. Clin Lung Cancer 2025; 26:201-209. [PMID: 39818516 PMCID: PMC12037305 DOI: 10.1016/j.cllc.2024.12.008] [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: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
OBJECTIVE To determine the association between concurrent statin use with immune checkpoint inhibitors (ICIs) and lung cancer-specific and overall mortality in patients with nonsmall cell lung cancer (NSCLC). MATERIALS AND METHODS SEER-Medicare was used to conduct a retrospective study of Medicare beneficiaries ≥65 years of age diagnosed with NSCLC between 2007 and 2017 treated with an ICI. Patients were followed from date of first ICI claim until death, 1 month from last ICI claim, or 12/31/2018, whichever came first. Associations for time-updated statin use and lung cancer-specific mortality, and overall mortality were estimated using Cox models adjusted for demographic, pathological, treatment-related factors, and a propensity score for statin use. RESULTS Among 1,401 patients, concurrent statin use with any ICI was associated with 41% lower risk of lung-cancer specific mortality compared to patients receiving ICI not using a statin (HR = 0.59; 95% CI = 0.35-0.99). Statin use was associated with a similarly lower risk of overall mortality (HR = 0.62; 95% CI = 0.41-0.94). Consistent inverse associations were observed when restricting to PD-1 inhibitors and by statin type. Limited anti-PD-L1 treatment prevented analysis in this subgroup. CONCLUSION Concurrent statin use with ICIs was associated with lower risk of lung cancer-specific and overall mortality in a population-based sample of older patients with NSCLC. Future work is needed to confirm these findings in prospective studies and randomized trials, including evaluating concurrent statin use with frontline ICIs, deciphering the underlying mechanism, and determining the optimal statin-ICI combination that maximize clinical benefit.
Collapse
Affiliation(s)
- Michael T Marrone
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC.
| | - Joshua E Reuss
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Anna Crawford
- Hollings Cancer Center Biostatistics Shared Resource, Medical University of South Carolina, Charleston, SC
| | - Brian Neelon
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC; Hollings Cancer Center Biostatistics Shared Resource, Medical University of South Carolina, Charleston, SC
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Julie R Brahmer
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
2
|
Azizi L, Hausman H, Meyer AK, Wong M, Pajonk F. The Mevalonate Pathway in the Radiation Response of Cancer. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00278-0. [PMID: 40194746 DOI: 10.1016/j.ijrobp.2025.03.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/14/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
The mevalonate (MVA) pathway plays a critical role in cholesterol biosynthesis, protein prenylation, and metabolic reprogramming, all of which contribute to cancer progression and therapy resistance. Targeting the MVA pathway with statins and other inhibitors has shown promise in preclinical studies; however, clinical outcomes remain controversial, raising concerns about translating these findings into effective treatments. Additionally, the interaction between the MVA pathway and radiation therapy (RT) is not yet fully understood, as RT upregulates the pathway, which can enhance tumor cell survival. This review summarizes the current literature on MVA pathway inhibition in cancer therapy, focusing on its potential to enhance the efficacy of RT. A better understanding of the pathway's role in radiation responses will be essential to translate combination therapies that target this pathway.
Collapse
Affiliation(s)
- Linda Azizi
- Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California.
| | - Hannah Hausman
- Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Alexandra K Meyer
- Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Matthew Wong
- Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California; Department of Neurosurgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California; Jonsson Comprehensive Cancer Center at University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
3
|
Salek F, Guest A, Johnson C, Kastelic JP, Thundathil J. Factors Affecting the Success of Ovum Pick-Up, In Vitro Production and Cryopreservation of Embryos in Cattle. Animals (Basel) 2025; 15:344. [PMID: 39943114 PMCID: PMC11815730 DOI: 10.3390/ani15030344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/26/2024] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Increasing global demand for animal proteins warrants improved productivity by genetic selection of superior cattle and faster dissemination of genetics. Availability of more progeny for genomic selection should maximize chances of identifying animals with desirable traits and increase selection pressure. OPU and IVP of embryos using these oocytes will substantially increase calves produced compared to conventional embryo transfer (ET). The OPU-IVP technology not only supports genetic improvement but also contributes to reducing environmental impacts of livestock production systems by improving efficiency and optimizing resources, aligning with the Sustainable Development Goals of the United Nations. However, there are several factors influencing the success of OPU-IVP. This review is focused on these factors and the impacts of in vitro culture conditions on the lipid content of embryos and potential role of L-carnitine, a lipolytic agent, on developmental competence of IVP embryos. The documented effects of L-carnitine and current knowledge regarding regulation of the Hippo signaling pathway suggest that supplementation of embryo culture media with L-carnitine will increase post-thaw survival of IVP embryos and their subsequent developmental competence by regulating lipid metabolism, production of reactive oxygen species, and Hippo signaling. Therefore, this review highlights current advancements in the field of OPU-IVP and potential areas for refining culture conditions to yield developmentally competent embryos that survive cryopreservation procedures.
Collapse
Affiliation(s)
| | | | | | | | - Jacob Thundathil
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada (C.J.); (J.P.K.)
| |
Collapse
|
4
|
Zhang J, Zhang M, Qiu A, Li C, Chen Q, Li J, Zeng Y, Zhu J, Huang JA, Zhang X, Liu Z. N 6-methyladenosine Reader IGF2BP2-modified HMMR Promotes Non-small Cell Lung Cancer Metastasis via Interaction with MAP4K4. Int J Biol Sci 2025; 21:1391-1409. [PMID: 39990663 PMCID: PMC11844294 DOI: 10.7150/ijbs.104097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/10/2025] [Indexed: 02/25/2025] Open
Abstract
Globally, lung cancer represents the leading cause of cancer-related mortality, with 85% of cases attributable to non-small cell lung cancer (NSCLC). Metastatic progression remains a major challenge in treating advanced lung cancer, resulting in a dismal five-year survival rate of 20-30%. Hyaluronan mediated motility receptor (HMMR) has been identified as a novel oncogene in NSCLC. However, its exact role and mechanisms in NSCLC and metastasis are yet to be fully understood. Elevated mRNA and protein levels of HMMR were observed in human NSCLC tumors in comparison with normal adjacent tissues. Increased HMMR expression was associated with poorer prognosis, with multivariate Cox regression analysis also identifying it as an independent prognostic factor. HMMR knockdown inhibited tumor cell migration and invasion, while its overexpression enhanced these processes. Mechanistically, HMMR promotes tumor metastasis by binding to mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), which activates the p-JNK/p-c-JUN/MMP1 signaling cascade. The effects of HMMR overexpression on metastatic potential and JNK signaling were confirmed by MAP4K4 knockdown or GNE-495 treatment. Additionally, insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) was found to bind to the N6-methyladenosine (m6A) site of HMMR, increasing mRNA stability and HMMR expression levels. In a mouse model, the MAP4K4 inhibitor GNE-495 successfully suppressed lung metastasis induced by HMMR overexpression. These results offer valuable insights into HMMR's biological functions while suggesting potential avenues for novel treatments.
Collapse
Affiliation(s)
- Jiansheng Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, The Affiliated Hospital of Jiangsu Vocational College of Medicine, Yancheng, 224000, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Mengzhu Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Aimin Qiu
- The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, The Affiliated Hospital of Jiangsu Vocational College of Medicine, Yancheng, 224000, China
| | - Chang Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Qiongju Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jianjun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Jian-an Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| | - Xiuqin Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
- Suzhou Key Laboratory for Respiratory Diseases, Suzhou, 215006, China
| |
Collapse
|
5
|
Aria H, Azizi M, Nazem S, Mansoori B, Darbeheshti F, Niazmand A, Daraei A, Mansoori Y. Competing endogenous RNAs regulatory crosstalk networks: The messages from the RNA world to signaling pathways directing cancer stem cell development. Heliyon 2024; 10:e35208. [PMID: 39170516 PMCID: PMC11337742 DOI: 10.1016/j.heliyon.2024.e35208] [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: 02/10/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Cancer stem cells (CSCs) are one of the cell types that account for cancer heterogeneity. The cancer cells arrest in G0 and generate non-CSC progeny through self-renewal and pluripotency, resulting in tumor recurrence, metastasis, and resistance to chemotherapy. They can stimulate tumor relapse and re-grow a metastatic tumor. So, CSCs is a promising target for eradicating tumors, and developing an anti-CSCs therapy has been considered. In recent years competing endogenous RNA (ceRNA) has emerged as a significant class of post-transcriptional regulators that affect gene expression via competition for microRNA (miRNA) binding. Furthermore, aberrant ceRNA expression is associated with tumor progression. Recent findings show that ceRNA network can cause tumor progression through the effect on CSCs. To overcome therapeutic resistance due to CSCs, we need to improve our current understanding of the mechanisms by which ceRNAs are implicated in CSC-related relapse. Thus, this review was designed to discuss the role of ceRNAs in CSCs' function. Targeting ceRNAs may open the path for new cancer therapeutic targets and can be used in clinical research.
Collapse
Affiliation(s)
- Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Nazem
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Mansoori
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzaneh Darbeheshti
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
6
|
Pan C, Hao X, Deng X, Lu F, Liu J, Hou W, Xu T. The roles of Hippo/YAP signaling pathway in physical therapy. Cell Death Discov 2024; 10:197. [PMID: 38670949 PMCID: PMC11053014 DOI: 10.1038/s41420-024-01972-x] [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: 01/24/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Cellular behavior is regulated by mechanical signals within the cellular microenvironment. Additionally, changes of temperature, blood flow, and muscle contraction also affect cellular state and the development of diseases. In clinical practice, physical therapy techniques such as ultrasound, vibration, exercise, cold therapy, and hyperthermia are commonly employed to alleviate pain and treat diseases. However, the molecular mechanism about how these physiotherapy methods stimulate local tissues and control gene expression remains unknow. Fortunately, the discovery of YAP filled this gap, which has been reported has the ability to sense and convert a wide variety of mechanical signals into cell-specific programs for transcription, thereby offering a fresh perspective on the mechanisms by which physiotherapy treat different diseases. This review examines the involvement of Hippo/YAP signaling pathway in various diseases and its role in different physical therapy approaches on diseases. Furthermore, we explore the potential therapeutic implications of the Hippo/YAP signaling pathway and address the limitations and controversies surrounding its application in physiotherapy.
Collapse
Affiliation(s)
- Chunran Pan
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjie Hou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
7
|
Noom A, Sawitzki B, Knaus P, Duda GN. A two-way street - cellular metabolism and myofibroblast contraction. NPJ Regen Med 2024; 9:15. [PMID: 38570493 PMCID: PMC10991391 DOI: 10.1038/s41536-024-00359-x] [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: 07/24/2023] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
Tissue fibrosis is characterised by the high-energy consumption associated with myofibroblast contraction. Although myofibroblast contraction relies on ATP production, the role of cellular metabolism in myofibroblast contraction has not yet been elucidated. Studies have so far only focused on myofibroblast contraction regulators, such as integrin receptors, TGF-β and their shared transcription factor YAP/TAZ, in a fibroblast-myofibroblast transition setting. Additionally, the influence of the regulators on metabolism and vice versa have been described in this context. However, this has so far not yet been connected to myofibroblast contraction. This review focuses on the known and unknown of how cellular metabolism influences the processes leading to myofibroblast contraction and vice versa. We elucidate the signalling cascades responsible for myofibroblast contraction by looking at FMT regulators, mechanical cues, biochemical signalling, ECM properties and how they can influence and be influenced by cellular metabolism. By reviewing the existing knowledge on the link between cellular metabolism and the regulation of myofibroblast contraction, we aim to pinpoint gaps of knowledge and eventually help identify potential research targets to identify strategies that would allow switching tissue fibrosis towards tissue regeneration.
Collapse
Affiliation(s)
- Anne Noom
- Julius Wolff Institute (JWI), Berlin Institute of Health and Center for Musculoskeletal Surgery at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Birgit Sawitzki
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt University of Berlin, 13353, Berlin, Germany
- Center of Immunomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Petra Knaus
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute (JWI), Berlin Institute of Health and Center for Musculoskeletal Surgery at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| |
Collapse
|
8
|
Atmane MI, Vigneau AL, Beaudry F, Rico C, Boerboom D, Paquet M. Therapeutic trial of fluvastatin in a cell line xenograft model of canine mammary gland cancer. Vet Comp Oncol 2023; 21:634-645. [PMID: 37709554 DOI: 10.1111/vco.12926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 09/16/2023]
Abstract
The Hippo signalling pathway is involved in breast cancer and canine mammary tumour (CMT). This study sought to evaluate the efficacy of fluvastatin on the Hippo pathway and its main effectors, YAP and TAZ, in vivo in a murine CMT cell line xenograft model. On treatment day 1, mice were divided into four groups: vehicle, fluvastatin, doxorubicin or a combination therapy. Tumour volumes were monitored with callipers and tissues harvested on day 28th of treatment. Histopathological examination of tumour tissues and major organs was performed as well as tumour evaluation of necrosis, apoptosis, cellular proliferation, expression of YAP, TAZ and the mRNA levels of four of their target genes (CTGF, CYR61, ANKRD1 and RHAMM2). Results showed a statistically significant variation in tumour volumes only for the combination therapy and final tumour weight only for the doxorubicin group compared to control. There was no significant difference in tumour necrosis, expression of CC3, ki-67, YAP and TAZ measured by immunohistochemistry and in the mRNA levels of the target genes. Unexpectedly, lung metastases were found in the control group (9) and not in the fluvastatin treated group (7). In addition, mass spectrometry-based quantification of fluvastatin reveals concentrations comparable to levels reported to exert therapeutic effects. This study shows that fluvastatin tumours concentration reached therapeutic levels without having an effect on the hippo pathway or various tumour parameters. Interestingly, only the control group had lung metastases. This study is the first to explore the repurposing of statins for cancer treatment in veterinary medicine.
Collapse
Affiliation(s)
- Mohamed Issam Atmane
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Anne-Laurence Vigneau
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Francis Beaudry
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Centre de recherche sur le cerveau et l'apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada
| | - Charlène Rico
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Derek Boerboom
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Centre de recherche en reproduction et fertilité (CRRF), Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Marilène Paquet
- Département de Pathologie et de Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
- Centre de recherche en reproduction et fertilité (CRRF), Université de Montréal, Saint-Hyacinthe, Québec, Canada
| |
Collapse
|
9
|
Benhammou JN, Qiao B, Ko A, Sinnett-Smith J, Pisegna JR, Rozengurt E. Lipophilic statins inhibit YAP coactivator transcriptional activity in HCC cells through Rho-mediated modulation of actin cytoskeleton. Am J Physiol Gastrointest Liver Physiol 2023; 325:G239-G250. [PMID: 37366601 PMCID: PMC10511177 DOI: 10.1152/ajpgi.00089.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of liver-related death. Lipophilic statins have been associated with a decrease in HCC incidence, raising the possibility of their use as chemoprevention agents. The Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) have emerged as an important pro-oncogenic mechanism in HCC. Statins modulate YAP/TAZ in other solid tumors, but few studies have assessed their mechanisms in HCC. We aimed to delineate how lipophilic statins regulate YAP protein localization by interrogating the mevalonate pathway in a stepwise manner using pharmacological and genetical approaches in HCC cells. Huh7 and Hep3B HCC cells were treated with the lipophilic statins cerivastatin and atorvastatin. YAP protein localization was determined using quantitative immunofluorescence (IF) imaging. The gene expression of CTGF and CYR61, known YAP/TEA-domain DNA-binding factor (TEAD)-regulated genes, was measured using quantitative real-time PCR. Rescue experiments were conducted using metabolites of the mevalonate pathway including mevalonic acid and geranylgeranyl pyrophosphate (GG-PP). The cellular cytoskeleton was assessed using F-actin IF staining. YAP protein was extruded from the nucleus to the cytoplasm with statin treatment. Consistently, CTGF and CYR61 mRNA expression significantly decreased with statins. Cytoskeletal structure was also compromised with statins. Gene expression, YAP protein localization, and cytoskeletal structure were all restored to baseline with exogenous GG-PP but not with other metabolites of the mevalonate pathway. Direct Rho GTPase inhibitor treatment mirrored the statin effects on YAP. YAP protein localization is regulated by lipophilic statins via Rho GTPases, causing cytoskeletal structural changes and is independent of cholesterol metabolites.NEW & NOTEWORTHY Statins are widely used for the treatment of cardiovascular diseases. Recently, their use has been associated with a decrease in the incidence of hepatocellular carcinoma (HCC); however, their mechanism(s) has remained elusive. In this study, we delineate the mechanism by which statins affect the Yes-associated protein (YAP), which has emerged as a key oncogenic pathway in HCC. We investigate each step of the mevalonate pathway and demonstrate that statins regulate YAP via Rho GTPases.
Collapse
Affiliation(s)
- Jihane N Benhammou
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, United States
| | - Bo Qiao
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
| | - Arthur Ko
- Center for Genetic Medicine Research, Childrens National Research Institute, Washington, District of Columbia, United States
| | - James Sinnett-Smith
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
| | - Joseph R Pisegna
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, United States
| | - Enrique Rozengurt
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, California, United States
- Division of Gastroenterology, Hepatology and Parental Nutrition, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, United States
| |
Collapse
|
10
|
Yoo H, Singh A, Li H, Strat AN, Bagué T, Ganapathy PS, Herberg S. Simvastatin Attenuates Glucocorticoid-Induced Human Trabecular Meshwork Cell Dysfunction via YAP/TAZ Inactivation. Curr Eye Res 2023; 48:736-749. [PMID: 37083467 PMCID: PMC10524554 DOI: 10.1080/02713683.2023.2206067] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/22/2023] [Accepted: 04/18/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE Impairment of the trabecular meshwork (TM) is the principal cause of increased outflow resistance in the glaucomatous eye. Yes-associated protein (YAP) and transcriptional coactivator with PDZ binding motif (TAZ) are emerging as potential mediators of TM cell/tissue dysfunction. Furthermore, YAP/TAZ activity was recently found to be controlled by the mevalonate pathway in non-ocular cells. Clinically used statins block the mevalonate cascade and were shown to improve TM cell pathobiology; yet, the link to YAP/TAZ signaling was not investigated. In this study, we hypothesized that simvastatin attenuates glucocorticoid-induced human TM (HTM) cell dysfunction via YAP/TAZ inactivation. METHODS Primary HTM cells were seeded atop or encapsulated within bioengineered extracellular matrix (ECM) hydrogels. Dexamethasone was used to induce a pathologic phenotype in HTM cells in the absence or presence of simvastatin. Changes in YAP/TAZ activity, actin cytoskeletal organization, phospho-myosin light chain levels, hydrogel contraction/stiffness, and fibronectin deposition were assessed. RESULTS Simvastatin potently blocked pathologic YAP/TAZ nuclear localization/activity, actin stress fiber formation, and myosin light chain phosphorylation in HTM cells. Importantly, simvastatin co-treatment significantly attenuated dexamethasone-induced ECM contraction/stiffening and fibronectin mRNA and protein levels. Sequential treatment was similarly effective but did not match clinically-used Rho kinase inhibition. CONCLUSIONS YAP/TAZ inactivation with simvastatin attenuates HTM cell pathobiology in a tissue-mimetic ECM microenvironment. Our data may help explain the association of statin use with a reduced risk of developing glaucoma via indirect YAP/TAZ inhibition as a proposed regulatory mechanism.
Collapse
Affiliation(s)
- Hannah Yoo
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Ayushi Singh
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Haiyan Li
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Ana N. Strat
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Tyler Bagué
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
11
|
Han Q, Qiu S, Hu H, Li W, Dang X, Li X. The relationship between the Hippo signaling pathway and bone metastasis of breast cancer. Front Oncol 2023; 13:1188310. [PMID: 37256184 PMCID: PMC10225633 DOI: 10.3389/fonc.2023.1188310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
Bone is the most common site of metastasis from breast cancer, which is the most prevalent cancer affecting women globally. Bone metastasis from breast cancer severely affects the quality of life of patients and increases mortality. The molecular mechanisms of metastasis, colonization, and proliferation of breast cancer cells in bone are complex and involve the interaction between breast cancer cells and the bone microenvironment. However, the precise mechanism is not clear at present. In recent years, the Hippo signaling pathway has attracted much attention due to its important role in regulating the expression of major effector molecules during tumor development. In particular, studies have found that the mutation and aberrant expression of the core components of the Hippo signaling pathway affect breast cancer cell migration and invasion, indicating that this pathway plays a role in bone metastasis, although the molecular mechanism of this pathway in breast cancer metastasis has not been fully elucidated. In this review, we discuss the function of the Hippo signaling pathway, introducing its role in breast cancer metastasis, especially bone metastasis of breast cancer, so as to lay a solid theoretical foundation for further research and for the development of effective targeted therapeutic agents.
Collapse
Affiliation(s)
- Qinyu Han
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Shi Qiu
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Huiwen Hu
- Department of The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Li
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Xiangguo Dang
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Xiangqi Li
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| |
Collapse
|
12
|
Bergen J, Karasova M, Bileck A, Pignitter M, Marko D, Gerner C, Del Favero G. Exposure to dietary fatty acids oleic and palmitic acid alters structure and mechanotransduction of intestinal cells in vitro. Arch Toxicol 2023; 97:1659-1675. [PMID: 37117602 PMCID: PMC10182945 DOI: 10.1007/s00204-023-03495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/04/2023] [Indexed: 04/30/2023]
Abstract
Intestinal cells are continuously exposed to food constituents while adapting to peristaltic movement and fluid shear stress. Oleic acid (OA) and palmitic acid (PA) are among the most prevalent fatty acids with respect to dietary lipids. Despite the central importance of dietary lipids for a balanced diet, awareness about potential detrimental effects related to excessive consumption is increasing; this includes toxicity, metabolic deregulation, and, particularly for cancer cells, a benefit from the uptake of fatty acids related to promotion of metastasis. Expanding on this, we started elucidating the effects of OA and PA (25-500 µM) on non-transformed human intestinal epithelial cells (HCEC-1CT) in comparison to colon carcinoma cells (HCT116), with regard to the mechanosensory apparatus. Hence, intestinal cells' motility is on the one side essential to ensure adaption to peristaltic movement and barrier function, but also to enable metastatic progression. Incubation with both OA and PA (≥ 25 µM) significantly decreased membrane fluidity of HCT116 cells, whereas the effect on HCEC-1CT was more limited. Application of rhodamine-labelled PA demonstrated that the fatty acid is incorporated into the plasma membrane of HCT116, which could not be observed in the non-tumorigenic cell line. Down-streaming into the intracellular compartment, a pronounced rearrangement of actin cytoskeleton was evident in both cell lines (OA and PA; 25 and 100 µM). This was accompanied by a variation of translocation efficiency of the mechanosensitive co-transcription factor YAP1, albeit with a stronger effect seen for PA and the cancer cells. Untargeted proteomic analysis confirmed that exposure to OA and PA could alter the response capacity of HCT116 cells to fluid shear stress. Taken together, OA and PA were able to functionally modulate the mechanosensory apparatus of intestinal cells, implying a novel role for dietary fatty acids in the regulation of intestinal pathophysiology.
Collapse
Affiliation(s)
- Janice Bergen
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
| | - Martina Karasova
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Medical University of Vienna, Vienna, Austria
| | - Marc Pignitter
- Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Josef-Holaubek-Platz 2, 1090, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria
- Joint Metabolome Facility, University of Vienna, Medical University of Vienna, Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria.
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Währingerstr. 38-42, 1090, Vienna, Austria.
| |
Collapse
|
13
|
Messelodi D, Strocchi S, Bertuccio SN, Baden P, Indio V, Giorgi FM, Taddia A, Serravalle S, Valente S, di Fonzo A, Frattini E, Bernardoni R, Pession A, Grifoni D, Deleidi M, Astolfi A, Pession A. Neuronopathic Gaucher disease models reveal defects in cell growth promoted by Hippo pathway activation. Commun Biol 2023; 6:431. [PMID: 37076591 PMCID: PMC10115838 DOI: 10.1038/s42003-023-04813-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/05/2023] [Indexed: 04/21/2023] Open
Abstract
Gaucher Disease (GD), the most common lysosomal disorder, arises from mutations in the GBA1 gene and is characterized by a wide spectrum of phenotypes, ranging from mild hematological and visceral involvement to severe neurological disease. Neuronopathic patients display dramatic neuronal loss and increased neuroinflammation, whose molecular basis are still unclear. Using a combination of Drosophila dGBA1b loss-of-function models and GD patient-derived iPSCs differentiated towards neuronal precursors and mature neurons we showed that different GD- tissues and neuronal cells display an impairment of growth mechanisms with an increased cell death and reduced proliferation. These phenotypes are coupled with the downregulation of several Hippo transcriptional targets, mainly involved in cells and tissue growth, and YAP exclusion from nuclei. Interestingly, Hippo knock-down in the GBA-KO flies rescues the proliferative defect, suggesting that targeting the Hippo pathway can be a promising therapeutic approach to neuronopathic GD.
Collapse
Affiliation(s)
- Daria Messelodi
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Silvia Strocchi
- Laboratory of Translational Research, USL-IRCCS of Reggio Emilia, 42123, Reggio Emilia, Italy
| | | | - Pascale Baden
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, 72076, Germany
- Hertie Institut for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Valentina Indio
- Department of Veterinary Medical Sciences, University of Bologna, 40064, Ozzano dell'Emilia (BO), Italy
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Alberto Taddia
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Salvatore Serravalle
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Sabrina Valente
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Alessio di Fonzo
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Emanuele Frattini
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Roberto Bernardoni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | | | - Daniela Grifoni
- Department of Life, Health and Environmental Sciences (MeSVA), University of L'Aquila, 67100, L'Aquila, Italy.
| | - Michela Deleidi
- Hertie Institut for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
- Institut Imagine, INSERM UMR1163, 75015, Paris, France
| | - Annalisa Astolfi
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Andrea Pession
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| |
Collapse
|
14
|
Anwar T, Sinnett-Smith J, Jin YP, Reed EF, Rozengurt E. Lipophilic Statins Inhibit YAP Nuclear Localization, Coactivator Activity, and Migration in Response to Ligation of HLA Class I Molecules in Endothelial Cells: Role of YAP Multisite Phosphorylation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1134-1145. [PMID: 36881871 PMCID: PMC10073314 DOI: 10.4049/jimmunol.2200568] [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: 08/05/2022] [Accepted: 02/10/2023] [Indexed: 03/09/2023]
Abstract
Solid-organ transplant recipients exhibiting HLA donor-specific Abs are at risk for graft loss due to chronic Ab-mediated rejection. HLA Abs bind HLA molecules expressed on the surface of endothelial cells (ECs) and induce intracellular signaling pathways, including the activation of the transcriptional coactivator yes-associated protein (YAP). In this study, we examined the impact of lipid-lowering drugs of the statin family on YAP localization, multisite phosphorylation, and transcriptional activity in human ECs. Exposure of sparse cultures of ECs to cerivastatin or simvastatin induced striking relocalization of YAP from the nucleus to the cytoplasm and inhibited the expression of the YAP/TEA domain DNA-binding transcription factor-regulated genes connective tissue growth factor and cysteine-rich angiogenic inducer 61. In dense cultures of ECs, statins prevented YAP nuclear import and expression of connective tissue growth factor and cysteine-rich angiogenic inducer 61 stimulated by the mAb W6/32 that binds HLA class I. Exposure of ECs to either cerivastatin or simvastatin completely blocked the migration of ECs stimulated by ligation of HLA class I. Exogenously supplied mevalonic acid or geranylgeraniol reversed the inhibitory effects of statins on YAP localization either in low-density ECs or high-density ECs challenged with W6/32. Mechanistically, cerivastatin increased the phosphorylation of YAP at Ser127, blunted the assembly of actin stress fiber, and inhibited YAP phosphorylation at Tyr357 in ECs. Using mutant YAP, we substantiated that YAP phosphorylation at Tyr357 is critical for YAP activation. Collectively, our results indicate that statins restrain YAP activity in EC models, thus providing a plausible mechanism underlying their beneficial effects in solid-organ transplant recipients.
Collapse
Affiliation(s)
- Tarique Anwar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - James Sinnett-Smith
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- VA Greater Los Angeles Health System
| | - Yi-Ping Jin
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- VA Greater Los Angeles Health System
| |
Collapse
|
15
|
Feng H, Liu X, Zhou C, Gu Q, Li Y, Chen J, Teng J, Zheng P. CCDC115 inhibits autophagy-mediated degradation of YAP to promote cell proliferation. FEBS Lett 2023; 597:618-630. [PMID: 36650560 DOI: 10.1002/1873-3468.14575] [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: 10/18/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 01/19/2023]
Abstract
Autophagy and Hippo signalling pathways both play important roles in cell homeostasis and are often involved in tumourigenesis. However, the crosstalk between these two signal pathways in response to stress conditions, such as nutrient deficiency, is incompletely understood. Here, we show that vesicular localised coiled-coil domain containing 115 (CCDC115) inhibits autophagy as well as Hippo signalling pathway under starvation. Moreover, we show that CCDC115 interacts with the HOPS complex. This interaction competes with STX17, thus inhibiting the fusion of autophagosomes with lysosomes. Hence, CCDC115 inhibits the autophagic degradation of yes-associated protein (YAP), thereby promoting cell proliferation in nutrient-restricted situation.
Collapse
Affiliation(s)
- Hui Feng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Department of Biotechnology, Beijing Polytechnic, China
| | - Xiao Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Chenqian Zhou
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Qiuchen Gu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- School of Life Sciences, Beijing Normal University, China
| | - Ye Li
- Department of Biotechnology, Beijing Polytechnic, China
| | - Jianguo Chen
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Junlin Teng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Pengli Zheng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
16
|
Abstract
Deregulation of transcription factors is critical to hallmarks of cancer. Genetic mutations, gene fusions, amplifications or deletions, epigenetic alternations, and aberrant post-transcriptional modification of transcription factors are involved in the regulation of various stages of carcinogenesis, including cancer initiation, progression, and metastasis. Thus, targeting the dysfunctional transcription factors may lead to new cancer therapeutic strategies. However, transcription factors are conventionally considered as "undruggable." Here, we summarize the recent progresses in understanding the regulation of transcription factors in cancers and strategies to target transcription factors and co-factors for preclinical and clinical drug development, particularly focusing on c-Myc, YAP/TAZ, and β-catenin due to their significance and interplays in cancer.
Collapse
Affiliation(s)
- Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
17
|
Sileo P, Simonin C, Melnyk P, Chartier-Harlin MC, Cotelle P. Crosstalk between the Hippo Pathway and the Wnt Pathway in Huntington's Disease and Other Neurodegenerative Disorders. Cells 2022; 11:cells11223631. [PMID: 36429058 PMCID: PMC9688160 DOI: 10.3390/cells11223631] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
The Hippo pathway consists of a cascade of kinases that controls the phosphorylation of the co-activators YAP/TAZ. When unphosphorylated, YAP and TAZ translocate into the nucleus, where they mainly bind to the TEAD transcription factor family and activate genes related to cell proliferation and survival. In this way, the inhibition of the Hippo pathway promotes cell survival, proliferation, and stemness fate. Another pathway can modulate these processes, namely the Wnt/β-catenin pathway that is indeed involved in cellular functions such as proliferation and cell survival, as well as apoptosis, growth, and cell renewal. Wnt signaling can act in a canonical or noncanonical way, depending on whether β-catenin is involved in the process. In this review, we will focus only on the canonical Wnt pathway. It has emerged that YAP/TAZ are components of the β-catenin destruction complex and that there is a close relationship between the Hippo pathway and the canonical Wnt pathway. Furthermore, recent data have shown that both of these pathways may play a role in neurodegenerative diseases, such as Huntington's disease, Alzheimer's disease, or Amyotrophic Lateral Sclerosis. Thus, this review analyzes the Hippo pathway and the Wnt pathway, their crosstalk, and their involvement in Huntington's disease, as well as in other neurodegenerative disorders. Altogether, these data suggest possible therapeutic approaches targeting key players of these pathways.
Collapse
Affiliation(s)
- Pasquale Sileo
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
| | - Clémence Simonin
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
- Centre de Référence Maladie de Huntington, CHU Lille, F-59000 Lille, France
| | - Patricia Melnyk
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
| | - Marie-Christine Chartier-Harlin
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
- Correspondence: (M.-C.C.-H.); (P.C.)
| | - Philippe Cotelle
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
- ENSCL-Centrale Lille, CS 90108, F-59652 Villeneuve d’Ascq, France
- Correspondence: (M.-C.C.-H.); (P.C.)
| |
Collapse
|
18
|
Koinis F, Chantzara E, Samarinas M, Xagara A, Kratiras Z, Leontopoulou V, Kotsakis A. Emerging Role of YAP and the Hippo Pathway in Prostate Cancer. Biomedicines 2022; 10:2834. [PMID: 36359354 PMCID: PMC9687800 DOI: 10.3390/biomedicines10112834] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/13/2022] [Accepted: 11/02/2022] [Indexed: 09/05/2023] Open
Abstract
The Hippo pathway regulates and contributes to several hallmarks of prostate cancer (PCa). Although the elucidation of YAP function in PCa is in its infancy, emerging studies have shed light on the role of aberrant Hippo pathway signaling in PCa development and progression. YAP overexpression and nuclear localization has been linked to poor prognosis and resistance to treatment, highlighting a therapeutic potential that may suggest innovative strategies to treat cancer. This review aimed to summarize available data on the biological function of the dysregulated Hippo pathway in PCa and identify knowledge gaps that need to be addressed for optimizing the development of YAP-targeted treatment strategies in patients likely to benefit.
Collapse
Affiliation(s)
- Filippos Koinis
- Department of Medical Oncology, University General Hospital of Larissa, 41221 Larissa, Greece
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Evangelia Chantzara
- Department of Medical Oncology, University General Hospital of Larissa, 41221 Larissa, Greece
| | - Michael Samarinas
- Department of Urology, General Hospital “Koutlibanio”, 41221 Larissa, Greece
| | - Anastasia Xagara
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Zisis Kratiras
- 3rd Urology Department University of Athens, “Attikon” University General Hospital, 12462 Chaidari, Greece
| | - Vasiliki Leontopoulou
- Department of Medical Oncology, University General Hospital of Larissa, 41221 Larissa, Greece
| | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, 41221 Larissa, Greece
- Laboratory of Oncology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| |
Collapse
|
19
|
Sun X, Zhang J, Dong J, Liu L, Li X, Xing P, Ying J, Che Y, Li J, Yang L. Prognostic significance of YAP1 expression and its association with neuroendocrine markers in resected pulmonary large cell neuroendocrine carcinoma (LCNEC). Transl Oncol 2022; 25:101538. [PMID: 36103754 PMCID: PMC9478447 DOI: 10.1016/j.tranon.2022.101538] [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: 07/24/2022] [Revised: 08/28/2022] [Accepted: 09/03/2022] [Indexed: 11/18/2022] Open
Abstract
It is the first study to determine the prognostic relevance of YAP1 in pulmonary LCNEC. And we found YAP1 is a prognostic factor for worse survival, especially for DFS. We investigated the relationship between YAP1 and NE markers (INSM1, DLL3, NeuroD1) and found that YAP1 expression was negatively correlated with INSM1 and DLL3, but not significantly correlated with NeuroD1. Our sample size is large and the clinical data is complete. The exploration of the prognostic mechanism of LCNEC is of great significance to its subtype classification and stratification of treatment and prognosis.
Background YAP1 (Yes-associated protein 1), an important effector of the Hippo pathway, acts as an oncogene and is overexpressed in various malignant tumors. However, the function and expression pattern of YAP1 in pulmonary large cell neuroendocrine carcinoma (LCNEC) have not been systematically established. This study aimed to explore the relationship between YAP1 expression and neuroendocrine differentiation markers and their prognostic significance in LCNEC. Materials and methods YAP1 protein and neuroendocrine markers (INSM1, NeuroD1 and DLL3) expression were examined by immunohistochemical (IHC) staining in 80 resected pulmonary LCNEC cases. The possible association between these markers and clinicopathological features was evaluated and survival analyses were performed. Results YAP1 was highly expressed in 25% LCNECs (20/80) , especially at a relatively higher T stage (p = 0.015). YAP1 expression was negatively correlated with INSM1 (χ2=11.53, p = 0.001) and DLL3(χ2=8.55, p = 0.004), but not with NeuroD1 (p = 0.482). For survival analyses, YAP1 expression was associated with worse disease-free survival (DFS) and overall survival (OS) (median DFS: 13 months vs. not reached (NR), p = 0.0096; median OS: not reached, NR vs. NR, p = 0.038), and was an unfavorable prognostic factor for DFS (HR:3.285; 95%CI: 1.526-7.071, p = 0.002) and OS (HR: 2.864, 95% CI: 0.932-8.796, p = 0.066). Conclusions YAP1 was found to be conversely correlated with neuroendocrine markers and a prognostic factor for worse survival in resected LCNEC patients, and mechanisms need to be further investigated.
Collapse
Affiliation(s)
- Xujie Sun
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jinyao Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiyan Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Li Liu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xue Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yiqun Che
- Center for Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing 100050, China
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
20
|
Targeting the Hippo Pathway in Gastric Cancer and Other Malignancies in the Digestive System: From Bench to Bedside. Biomedicines 2022; 10:biomedicines10102512. [PMID: 36289774 PMCID: PMC9599207 DOI: 10.3390/biomedicines10102512] [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: 07/25/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
The Hippo pathway is an evolutionally conserved signaling cascade that controls organ size and tissue regeneration under physiological conditions, and its aberrations have been well studied to promote tumor initiation and progression. Dysregulation of the Hippo tumor suppressor signaling frequently occurs in gastric cancer (GC) and other solid tumors and contributes to cancer development through modulating multiple aspects, including cell proliferation, survival, metastasis, and oncotherapy resistance. In the clinic, Hippo components also possess diagnostic and prognostic values for cancer patients. Considering its crucial role in driving tumorigenesis, targeting the Hippo pathway may greatly benefit developing novel cancer therapies. This review summarizes the current research progress regarding the core components and regulation of the Hippo pathway, as well as the mechanism and functional roles of their dysregulation in gastrointestinal malignancies, especially in GC, and discusses the therapeutic potential of targeting the Hippo pathway against cancers.
Collapse
|
21
|
Krasniqi E, Di Lisa FS, Di Benedetto A, Barba M, Pizzuti L, Filomeno L, Ercolani C, Tinari N, Grassadonia A, Santini D, Minelli M, Montemurro F, Fabbri MA, Mazzotta M, Gamucci T, D’Auria G, Botti C, Pelle F, Cavicchi F, Cappelli S, Cappuzzo F, Sanguineti G, Tomao S, Botticelli A, Marchetti P, Maugeri-Saccà M, De Maria R, Ciliberto G, Sperati F, Vici P. The Impact of the Hippo Pathway and Cell Metabolism on Pathological Complete Response in Locally Advanced Her2+ Breast Cancer: The TRISKELE Multicenter Prospective Study. Cancers (Basel) 2022; 14:cancers14194835. [PMID: 36230758 PMCID: PMC9563553 DOI: 10.3390/cancers14194835] [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: 09/07/2022] [Accepted: 09/28/2022] [Indexed: 12/02/2022] Open
Abstract
The Hippo pathway and its two key effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), are consistently altered in breast cancer. Pivotal regulators of cell metabolism such as the AMP-activated protein kinase (AMPK), Stearoyl-CoA-desaturase 1 (SCD1), and HMG-CoA reductase (HMGCR) are relevant modulators of TAZ/YAP activity. In this prospective study, we measured the tumor expression of TAZ, YAP, AMPK, SCD1, and HMGCR by immunohistochemistry in 65 Her2+ breast cancer patients who underwent trastuzumab-based neoadjuvant treatment. The aim of the study was to assess the impact of the immunohistochemical expression of the Hippo pathway transducers and cell metabolism regulators on pathological complete response. Low expression of cytoplasmic TAZ, both alone and in the context of a composite signature identified by machine learning including also low nuclear levels of YAP and HMGCR and high cytoplasmic levels of SCD1, was a predictor of residual disease in the univariate logistic regression. This finding was not confirmed in the multivariate model including estrogen receptor > 70% and body mass index > 20. However, our findings were concordant with overall survival data from the TCGA cohort. Our results, possibly affected by the relatively small sample size of this study population, deserve further investigation in adequately sized, ad hoc prospective studies.
Collapse
Affiliation(s)
- Eriseld Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Francesca Sofia Di Lisa
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Anna Di Benedetto
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence: or (M.B.); (C.E.); Tel.: +39-0652666762 (M.B.); +39-0652666134 (C.E.)
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Lorena Filomeno
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Cristiana Ercolani
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence: or (M.B.); (C.E.); Tel.: +39-0652666762 (M.B.); +39-0652666134 (C.E.)
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology (CAST), G. D’Annunzio University, 66100 Chieti, Italy
| | - Antonino Grassadonia
- Department of Innovative Technologies in Medicine and Dentistry, Centre for Advanced Studies and Technology (CAST), G. D’Annunzio University, 66100 Chieti, Italy
| | - Daniele Santini
- “Sapienza” University of Rome, Polo Pontino, 04011 Aprilia, Italy
| | - Mauro Minelli
- Division of Oncology, San Giovanni Hospital, 00184 Rome, Italy
| | - Filippo Montemurro
- Breast Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico), 10060 Candiolo, Italy
| | | | - Marco Mazzotta
- Medical Oncology Unit, Belcolle Hospital, 01100 Viterbo, Italy
| | - Teresa Gamucci
- Medical Oncology, Sandro Pertini Hospital, 00157 Rome, Italy
| | | | - Claudio Botti
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Fabio Pelle
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Flavia Cavicchi
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Sonia Cappelli
- Department of Surgery, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Federico Cappuzzo
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Andrea Botticelli
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Paolo Marchetti
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00167 Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Clinical Trial Center, Biostatistics and Bioinformatics, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Ruggero De Maria
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico), 00168 Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Francesca Sperati
- Clinical Trial Center, Biostatistics and Bioinformatics, San Gallicano Dermatological Institute IRCCS, 00144 Rome, Italy
| | - Patrizia Vici
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
22
|
Hinneh JA, Gillis JL, Moore NL, Butler LM, Centenera MM. The role of RHAMM in cancer: Exposing novel therapeutic vulnerabilities. Front Oncol 2022; 12:982231. [PMID: 36033439 PMCID: PMC9400171 DOI: 10.3389/fonc.2022.982231] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Receptor for hyaluronic acid-mediated motility (RHAMM) is a cell surface receptor for hyaluronic acid that is critical for cell migration and a cell cycle protein involved in microtubule assembly and stability. These functions of RHAMM are required for cellular stress responses and cell cycle progression but are also exploited by tumor cells for malignant progression and metastasis. RHAMM is often overexpressed in tumors and is an independent adverse prognostic factor for a number of cancers such as breast and prostate. Interestingly, pharmacological or genetic inhibition of RHAMM in vitro and in vivo ablates tumor invasiveness and metastatic spread, implicating RHAMM as a potential therapeutic target to restrict tumor growth and improve patient survival. However, RHAMM’s pro-tumor activity is dependent on its subcellular distribution, which complicates the design of RHAMM-directed therapies. An alternative approach is to identify downstream signaling pathways that mediate RHAMM-promoted tumor aggressiveness. Herein, we discuss the pro-tumoral roles of RHAMM and elucidate the corresponding regulators and signaling pathways mediating RHAMM downstream events, with a specific focus on strategies to target the RHAMM signaling network in cancer cells.
Collapse
Affiliation(s)
- Josephine A. Hinneh
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Freemason’s Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Joanna L. Gillis
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Nicole L. Moore
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Lisa M. Butler
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Freemason’s Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Lisa M. Butler, ; Margaret M. Centenera,
| | - Margaret M. Centenera
- South Australian Immunogenomics Cancer Institute and Adelaide Medical School, Adelaide, SA, Australia
- Freemason’s Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, SA, Australia
- Precision Cancer Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Lisa M. Butler, ; Margaret M. Centenera,
| |
Collapse
|
23
|
Pongjantarasatian S, Nowwarote N, Rotchanakitamnuai V, Srirodjanakul W, Saehun R, Janebodin K, Manokawinchoke J, Fournier BPJ, Osathanon T. A γ-Secretase Inhibitor Attenuates Cell Cycle Progression and Invasion in Human Oral Squamous Cell Carcinoma: An In Vitro Study. Int J Mol Sci 2022; 23:8869. [PMID: 36012128 PMCID: PMC9408752 DOI: 10.3390/ijms23168869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 12/13/2022] Open
Abstract
Notch signaling is associated with many human malignancies, including oral squamous cell carcinoma (OSCC). However, the exact function of Notch signaling in OSCC remains unclear. Here, we investigated the effect of Notch signaling inhibition using a γ-secretase inhibitor (DAPT) on OSCC behaviours in vitro. Bioinformatic analysis of public-available gene expression profiles revealed the dysregulation of the Notch signaling pathway in OSCC compared with normal tissues, indicating the role of Notch signaling in OSCC regulation. RNA sequencing analysis of DAPT-treated human OSCC cells revealed the dysregulation of genes related to cell cycle-related pathways. Blocking Notch signaling significantly inhibited cell proliferation. DAPT-induced G0/G1 cell cycle arrest induced cell apoptosis. Furthermore, cell migration and invasion were also reduced in DAPT-treated cells. These findings indicate that Notch signaling activation participates in OSCC regulation by promoting cell growth, cell cycle progression, cell migration, and invasion. These mechanisms could facilitate OSCC progression. These results imply the potential use of Notch signaling inhibitors as a candidate adjuvant treatment in OSCC patients.
Collapse
Affiliation(s)
- Sarai Pongjantarasatian
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nunthawan Nowwarote
- Department of Oral Biology, Faculty of Dentistry, Universite Paris Cite, 75006 Paris, France
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Universite Paris Cite, Sorbonne Universite, 75006 Paris, France
| | - Varumporn Rotchanakitamnuai
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watcharee Srirodjanakul
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ritmongkol Saehun
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kajohnkiart Janebodin
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Jeeranan Manokawinchoke
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Benjamin P. J. Fournier
- Department of Oral Biology, Faculty of Dentistry, Universite Paris Cite, 75006 Paris, France
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Universite Paris Cite, Sorbonne Universite, 75006 Paris, France
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
24
|
Benhammou JN, Sinnett-Smith J, Pisegna JR, Rozengurt EJ. Interplay Between Fatty Acids, Stearoyl-Co-A Desaturase, Mechanistic Target of Rapamycin, and Yes-Associated Protein/Transcriptional Coactivator With PDZ-Binding Motif in Promoting Hepatocellular Carcinoma. GASTRO HEP ADVANCES 2022; 2:232-241. [PMID: 39132609 PMCID: PMC11308718 DOI: 10.1016/j.gastha.2022.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/17/2022] [Indexed: 08/13/2024]
Abstract
Nonalcoholic fatty liver disease has reached pandemic proportions with one of its most consequential complications being hepatocellular carcinoma (HCC). Nonalcoholic fatty liver disease-related HCC is becoming the leading indication for liver transplantation in the United States. Given the scarcity of available organs, early detection and prevention remain key in prevention and management of the disease. Over the years, the yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) pathway emerged as a key signal transduction pathway in the pathogenesis of HCC. In this review, we explore the interplay between the YAP/TAZ pathway as a point of convergence in HCC pathogenesis. We review the evidence of how lipid reprogramming and key lipid pathways, saturated and monounsaturated fatty acids (through the rate-limiting enzyme stearoyl Co-A desaturase), the mevalonic acid pathway (the role of statins), and mechanistic target of rapamycin all play critical roles in intricate and complex networks that tightly regulate the YAP/TAZ pro-oncogenic pathway.
Collapse
Affiliation(s)
- Jihane N. Benhammou
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
| | - Jim Sinnett-Smith
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Joseph R. Pisegna
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
| | - Enrique J. Rozengurt
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
| |
Collapse
|
25
|
Senigagliesi B, Samperi G, Cefarin N, Gneo L, Petrosino S, Apollonio M, Caponnetto F, Sgarra R, Collavin L, Cesselli D, Casalis L, Parisse P. Triple negative breast cancer-derived small extracellular vesicles as modulator of biomechanics in target cells. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 44:102582. [PMID: 35817390 DOI: 10.1016/j.nano.2022.102582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 12/14/2022]
Abstract
Extracellular vesicle (EV) mediated communication has recently been proposed as one of the pivotal routes in the development of cancer metastasis. EVs are nano-sized vesicles swapped between cells, carrying a biologically active content that can promote tumor-induced immune suppression, metastasis and angiogenesis. Thus, EVs constitute a potential target in cancer therapy. However, their role in triggering the premetastatic niche and in tumor spreading is still unclear. Here, we focused on the EV ability to modulate the biomechanical properties of target cells, known to play a crucial role in metastatic spreading. To this purpose, we isolated and thoroughly characterized triple-negative breast cancer (TNBC)-derived small EVs. We then evaluated variations in the mechanical properties (cell stiffness, cytoskeleton/nuclear/morphology and Yap activity rearrangements) of non-metastatic breast cancer MCF7 cells upon EV treatment. Our results suggest that TNBC-derived small EVs are able to directly modify MCF7 cells by inducing a decrease in cell stiffness, rearrangements in cytoskeleton, focal adhesions and nuclear/cellular morphology, and an increase in Yap downstream gene expression. Testing the biomechanical response of cells after EV addition might represent a new functional assay in metastatic cancer framework that can be exploited for future application both in diagnosis and in therapy.
Collapse
Affiliation(s)
- Beatrice Senigagliesi
- Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy; Elettra-Sincrotrone Trieste S.C.p.A, Trieste, Italy.
| | | | - Nicola Cefarin
- Istituto Officina dei Materiali Consiglio Nazionale delle Ricerche, Trieste, Italy
| | | | - Sara Petrosino
- Telethon Institute of Genetics and Medicine, Naples, Italy.
| | - Mattia Apollonio
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| | - Licio Collavin
- Department of Life Sciences, University of Trieste, Trieste, Italy.
| | - Daniela Cesselli
- Pathology Department, University Hospital of Udine, Udine, Italy; Department of Medicine, University of Udine, Udine, Italy.
| | | | - Pietro Parisse
- Elettra-Sincrotrone Trieste S.C.p.A, Trieste, Italy; Istituto Officina dei Materiali Consiglio Nazionale delle Ricerche, Trieste, Italy.
| |
Collapse
|
26
|
Mutant p53, the Mevalonate Pathway and the Tumor Microenvironment Regulate Tumor Response to Statin Therapy. Cancers (Basel) 2022; 14:cancers14143500. [PMID: 35884561 PMCID: PMC9323637 DOI: 10.3390/cancers14143500] [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: 05/30/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor cells have the ability to co-opt multiple metabolic pathways, enhance glucose uptake and utilize aerobic glycolysis to promote tumorigenesis, which are characteristics constituting an emerging hallmark of cancer. Mutated tumor suppressor and proto-oncogenes are frequently responsible for enhanced metabolic pathway signaling. The link between mutant p53 and the mevalonate (MVA) pathway has been implicated in the advancement of various malignancies, with tumor cells relying heavily on increased MVA signaling to fuel their rapid growth, metastatic spread and development of therapy resistance. Statin drugs inhibit HMG-CoA reductase, the pathway’s rate-limiting enzyme, and as such, have long been studied as a potential anti-cancer therapy. However, whether statins provide additional anti-cancer properties is worthy of debate. Here, we examine retrospective, prospective and pre-clinical studies involving the use of statins in various cancer types, as well as potential issues with statins’ lack of efficacy observed in clinical trials and future considerations for upcoming clinical trials.
Collapse
|
27
|
YAP1 induces invadopodia formation by transcriptionally activating TIAM1 through enhancer in breast cancer. Oncogene 2022; 41:3830-3845. [PMID: 35773411 PMCID: PMC9337990 DOI: 10.1038/s41388-022-02344-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 11/12/2022]
Abstract
Yes-associated protein 1 (YAP1), a central component of the Hippo pathway, plays an important role in tumor metastasis; however, the underlying mechanism remains to be elucidated. Invadopodia are actin-rich protrusions containing multiple proteases and have been widely reported to promote cell invasiveness by degrading the extracellular matrix. In the present study, we report that YAP1 induces invadopodia formation and promotes tumor metastasis in breast cancer cells. We also identify TIAM1, a guanine nucleotide exchange factor, as a target of the YAP1–TEAD4 complex. Our results demonstrate that YAP1 could promote TEAD4 binding to the enhancer region of TIAM1, which activates TIAM1 expression, subsequently increasing RAC1 activity and inducing invadopodia formation. These findings reveal the functional role of Hippo signaling in the regulation of invadopodia and provide potential molecular targets for preventing tumor metastasis in breast cancer.
Collapse
|
28
|
Vahala D, Choi YS. Modelling the Tumor Microenvironment: Recapitulating Nano- and Micro-Scale Properties that Regulate Tumor Progression. Front Cell Dev Biol 2022; 10:908799. [PMID: 35800896 PMCID: PMC9254080 DOI: 10.3389/fcell.2022.908799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
Breast cancer remains a significant burden with 1 in 8 women affected and metastasis posing a significant challenge for patient survival. Disease progression involves remodeling of the extracellular matrix (ECM). In breast cancer, tissue stiffness increases owing to an increase in collagen production by recruited cancer-associated fibroblasts (CAFs). These stromal modifications are notable during primary tumor growth and have a dualistic action by creating a hard capsule to prevent penetration of anti-cancer therapies and forming a favorable environment for tumor progression. Remodeling of the tumor microenvironment immediately presented to cells can include changes in protein composition, concentration and structural arrangement and provides the first mechanical stimuli in the metastatic cascade. Not surprisingly, metastatic cancer cells possess the ability to mechanically adapt, and their adaptability ensures not only survival but successful invasion within altered environments. In the past decade, the importance of the microenvironment and its regulatory role in diseases have gained traction and this is evident in the shift from plastic culture to the development of novel biomaterials that mimic in vivo tissue. With these advances, elucidations can be made into how ECM remodeling and more specifically, altered cell-ECM adhesions, regulate tumor growth and cancer cell plasticity. Such enabling tools in mechanobiology will identify fundamental mechanisms in cancer progression that eventually help develop preventative and therapeutic treatment from a clinical perspective. This review will focus on current platforms engineered to mimic the micro and nano-properties of the tumor microenvironment and subsequent understanding of mechanically regulated pathways in cancer.
Collapse
|
29
|
Karalis T, Skandalis SS. Hyaluronan network: a driving force in cancer progression. Am J Physiol Cell Physiol 2022; 323:C145-C158. [PMID: 35649255 DOI: 10.1152/ajpcell.00139.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyaluronan is one of the most abundant macromolecules of the extracellular matrix and regulates several physiological cell and tissue properties. However, hyaluronan has been shown to accumulate together with its receptors in various cancers. In tumors, accumulation of hyaluronan system components (hyaluronan synthesizing/degrading enzymes and interacting proteins) associates with poor outcomes of the patients. In this article, we review the main roles of hyaluronan in normal physiology and cancer, and further discuss the targeting of hyaluronan system as an applicable therapeutic strategy.
Collapse
Affiliation(s)
- Theodoros Karalis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
30
|
Zhang Y, Wang X, Zhou X. Functions of Yes-association protein (YAP) in cancer progression and anticancer therapy resistance. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The Hippo pathway, a highly conserved kinase cascade, regulates cell proliferation, apoptosis, organ size, and tissue homeostasis. Dysregulation of this pathway reportedly plays an important role in the progression of various human cancers. Yes-association protein (YAP), the Hippo pathway’s core effector, is considered a marker for cancer therapy and patient prognosis. In addition, studies have indicated that YAP is involved in promoting anticancer drug resistance. This review summarizes current knowledge on YAP’s role in cancer progression, anticancer drug resistance, and advances in the development of YAP-targeting drugs. A thorough understanding of the complex interactions among molecular, cellular, and environmental factors concerning YAP function in cancer progression may provide new insight into the underlying mechanism of anticancer drug resistance. It might lead to improved prognosis through novel combined therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- These authors contributed equally to this work
| | - Xiang Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- These authors contributed equally to this work
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
31
|
She Q, Chen Y, Liu H, Tan J, Li Y. A high level of the long non-coding RNA MCF2L-AS1 is associated with poor prognosis in breast cancer and MCF2L-AS1 activates YAP transcriptional activity to enhance breast cancer proliferation and metastasis. Bioengineered 2022; 13:13437-13451. [PMID: 36700469 PMCID: PMC9276029 DOI: 10.1080/21655979.2022.2074108] [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] [Indexed: 02/01/2023] Open
Abstract
Breast cancer (BC) is one of the most prevalent gynecologic malignant tumors with a poor prognosis and the second leading cause of cancer-related deaths in women worldwide. In recent years, it has been shown that long non-coding RNA (lncRNA) plays an important role in the development of breast cancer (BC). An antisense lncRNA from the MCF2 cell line (MCF2L-AS1) has been discovered recently and has been shown to function in a variety of malignancies. However, its function as a regulator of BC development has yet to be determined. Herein, the bioinformatics study analysis showed that MCF2L-AS1 was frequently highly expressed in BC tumors, and this overexpression was associated with worse patient outcomes. BC cells' proliferation, migration, and invasion are inhibited when MCF2L-AS1 is silenced, whereas the inverse is evident when MCF2L-AS1 is overexpressed. It was also observed that MCF2L-AS1 knockdown decreased carcinogenesis in xenograft tumor models. Furthermore, we discovered that MCF2L-AS1 could bind to and improve the transcription activity of the yes-associated protein (YAP). However, following YAP knockdown, this lncRNA's ability to drive BC malignancy was considerably reduced. In conclusion, MCF2L-AS1 may represent a potential predictive biomarker in BC patients, as well as a key regulator of BC cell proliferation. It works through positive feedback processes involving direct YAP binding and subsequent modulation of intracellular gene expression. Our findings add to our understanding of MCF2L-AS1 regulation and its potential as a therapeutic target in patients with this fatal cancer type.
Collapse
Affiliation(s)
- Qing She
- Department of Breast Surgery, Baoji Municipal Central Hospital, Baoji, China
| | - Yuanyuan Chen
- Department of Breast Surgery, Baoji Municipal Central Hospital, Baoji, China
| | - Hong Liu
- Department of Breast Surgery, Baoji Municipal Central Hospital, Baoji, China
| | - Jichao Tan
- Department of Breast Surgery, Baoji Municipal Central Hospital, Baoji, China
| | - Youhuai Li
- Department of Breast Surgery, Baoji Municipal Central Hospital, Baoji, China,CONTACT Youhuai Li Department of Breast Surgery, Baoji Municipal Central Hospital, 8 Jiangtan Road, Weibin District, Baoji, Shaanxi721008, China
| |
Collapse
|
32
|
Lázaro-Mixteco PE, González-Coronel JM, Hernández-Padilla L, Martínez-Alcantar L, Martínez-Carranza E, López-Bucio JS, Guevara-García ÁA, Campos-García J. Transcriptomics Reveals the Mevalonate and Cholesterol Pathways Blocking as Part of the Bacterial Cyclodipeptides Cytotoxic Effects in HeLa Cells of Human Cervix Adenocarcinoma. Front Oncol 2022; 12:790537. [PMID: 35359411 PMCID: PMC8964019 DOI: 10.3389/fonc.2022.790537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 11/23/2022] Open
Abstract
The incidence of human cervix adenocarcinoma (CC) caused by papillomavirus genome integration into the host chromosome is the third most common cancer among women. Bacterial cyclodipeptides (CDPs) exert cytotoxic effects in human cervical cancer HeLa cells, primarily by blocking the PI3K/Akt/mTOR pathway, but downstream responses comprising gene expression remain unstudied. Seeking to understand the cytotoxic and anti-proliferative effects of CDPs in HeLa cells, a global RNA-Seq analysis was performed. This strategy permitted the identification of 151 differentially expressed genes (DEGs), which were either up- or down-regulated in response to CDPs exposure. Database analysis, including Gene Ontology (COG), and the Kyoto Encyclopedia of Genes and Genomes (KEGG), revealed differential gene expression on cancer transduction signals, and metabolic pathways, for which, expression profiles were modified by the CDPs exposure. Bioinformatics confirmed the impact of CDPs in the differential expression of genes from signal transduction pathways such as PI3K-Akt, mTOR, FoxO, Wnt, MAPK, P53, TGF-β, Notch, apoptosis, EMT, and CSC. Additionally, the CDPs exposure modified the expression of cancer-related transcription factors involved in the regulation of processes such as epigenetics, DNA splicing, and damage response. Interestingly, transcriptomic analysis revealed the participation of genes of the mevalonate and cholesterol biosynthesis pathways; in agreement with this observation, total cholesterol diminished, confirming the blockage of the cholesterol synthesis by the exposure of HeLa cells to CDPs. Interestingly, the expression of some genes of the mevalonate and cholesterol synthesis such as HMGS1, HMGCR, IDI1, SQLE, MSMO1, SREBF1, and SOAT1 was up-regulated by CDPs exposure. Accordingly, metabolites of the mevalonate pathway were accumulated in cultures treated with CDPs. This finding further suggests that the metabolism of cholesterol is crucial for the occurrence of CC, and the blockade of the sterol synthesis as an anti-proliferative mechanism of the bacterial CDPs, represents a reasonable chemotherapeutic drug target to explore. Our transcriptomic study supports the anti-neoplastic effects of bacterial CDPs in HeLa cells shown previously, providing new insights into the transduction signals, transcription factors and metabolic pathways, such as mevalonate and cholesterol that are impacted by the CDPs and highlights its potential as anti-neoplastic drugs.
Collapse
Affiliation(s)
- Pedro E Lázaro-Mixteco
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - José M González-Coronel
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Laura Hernández-Padilla
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Lorena Martínez-Alcantar
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Enrique Martínez-Carranza
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Jesús Salvador López-Bucio
- CONACYT-UMSNH, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Ángel A Guevara-García
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Jesús Campos-García
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
33
|
Wei J, Xia S, Sun A, Qu Y, Gao J, Shao G, Yang W, Lin Q. Geranylgeranylation signaling promotes breast cancer cell mitosis via the YAP-activated transcription of kinetochore/centromere genes. Am J Cancer Res 2022; 12:1143-1155. [PMID: 35411228 PMCID: PMC8984885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 02/15/2022] [Indexed: 06/14/2023] Open
Abstract
Geranylgeranylation signaling plays an important role in cancer cell proliferation. Our previous studies have shown that the YAP is one of the geranylgeranylation signal transducers in breast cancer cells (Mi W, et al., Oncogene. 2015; 34(24): 3095-3106). However, the downstream effectors that mediate the promoting effect of the geranylgeranylation/YAP signal axis on breast cancer cell proliferation remain elusive. In this report, we investigated the pathway that mediates the effect of the geranylgeranylation on breast cancer cell proliferation. The results have shown that inhibition of geranylgeranyl biosynthesis inactivates transcription of a set of kinetochore/centromere genes. Further biochemical and cell biological studies demonstrated that inhibition of geranylgeranyl biosynthesis significantly reduced the level of key kinetochore/centromere proteins, thus caused a defect in mitosis. Knockdown of YAP caused similar inhibitory effects on the kinetochore/centromere gene expression and mitosis to that of inhibition of geranylgeranyl biosynthesis. Furthermore, we found that E2F1, the gene coding for E2F1 that is known to activate expression of cell cycle genes, is a target gene of YAP. Knockdown of E2F1 also reduced expression of the kinetochore/centromere genes, suggesting that the activation effect of YAP on expression of the kinetochore/centromere genes may be mediated by E2F1. Our studies have proposed a novel geranylgeranylation-dependent cancer cell proliferation signaling pathway in which geranylgeranylation signaling promotes cancer cell mitosis via the YAP-activated transcription of kinetochore/centromere genes.
Collapse
Affiliation(s)
- Jing Wei
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Song Xia
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Aiqin Sun
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Yaping Qu
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Jinyi Gao
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Genbao Shao
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Wannian Yang
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Qiong Lin
- School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
34
|
The Hippo pathway in cancer: YAP/TAZ and TEAD as therapeutic targets in cancer. Clin Sci (Lond) 2022; 136:197-222. [PMID: 35119068 PMCID: PMC8819670 DOI: 10.1042/cs20201474] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023]
Abstract
Tumorigenesis is a highly complex process, involving many interrelated and cross-acting signalling pathways. One such pathway that has garnered much attention in the field of cancer research over the last decade is the Hippo signalling pathway. Consisting of two antagonistic modules, the pathway plays an integral role in both tumour suppressive and oncogenic processes, generally via regulation of a diverse set of genes involved in a range of biological functions. This review discusses the history of the pathway within the context of cancer and explores some of the most recent discoveries as to how this critical transducer of cellular signalling can influence cancer progression. A special focus is on the various recent efforts to therapeutically target the key effectors of the pathway in both preclinical and clinical settings.
Collapse
|
35
|
Li M, Rao X, Cui Y, Zhang L, Li X, Wang B, Zheng Y, Teng L, Zhou T, Zhuo W. The keratin 17/YAP/IL6 axis contributes to E-cadherin loss and aggressiveness of diffuse gastric cancer. Oncogene 2022; 41:770-781. [PMID: 34845376 DOI: 10.1038/s41388-021-02119-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022]
Abstract
DGC is a particular aggressive malignancy with poor prognosis. Recent omics studies characterized DGC with CDH1/E-cadherin loss and EMT-signatures. However, the underlying mechanisms for maintaining the aggressive behavior and molecular features of DGC remain unclear. Here, we find that intermediate filaments KRT17 is significantly lower in DGC tissues than that in intestinal gastric cancer tissues and associated with poor prognosis of DGC. We demonstrate that downregulation of KRT17 induces E-cadherin loss, EMT changes, and metastasis behaviors of GC cells. Mechanistically, the loss of intermediate filaments KRT17 induces reorganization of cytoskeleton, further activates YAP signaling, and increases IL6 expression, which contributes to the enhanced metastasis ability of GC cells. Together, these results indicate that KRT17/YAP/IL6 axis contributes to maintaining E-cadherin loss, EMT feature, and metastasis of DGC, providing a new insight into the role of aberrant intermediate filaments in DGC malignancy.
Collapse
Affiliation(s)
- Mengjie Li
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Xianping Rao
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Yun Cui
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Lu Zhang
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Xiang Li
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Boya Wang
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yijun Zheng
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Lisong Teng
- Cancer Center, Zhejiang University, Hangzhou, China
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Tianhua Zhou
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Wei Zhuo
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
36
|
Beyond Lipid-Lowering: Effects of Statins on Cardiovascular and Cerebrovascular Diseases and Cancer. Pharmaceuticals (Basel) 2022; 15:ph15020151. [PMID: 35215263 PMCID: PMC8877351 DOI: 10.3390/ph15020151] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, also known as statins, are administered as first-line therapy for hypercholesterolemia, both as primary and secondary prevention. Besides the lipid-lowering effect, statins have been suggested to inhibit the development of cardiovascular disease through anti-inflammatory, antioxidant, vascular endothelial function-improving, plaque-stabilizing, and platelet aggregation-inhibiting effects. The preventive effect of statins on atherothrombotic stroke has been well established, but statins can influence other cerebrovascular diseases. This suggests that statins have many neuroprotective effects in addition to lowering cholesterol. Furthermore, research suggests that statins cause pro-apoptotic, growth-inhibitory, and pro-differentiation effects in various malignancies. Preclinical and clinical evidence suggests that statins inhibit tumor growth and induce apoptosis in specific cancer cell types. The pleiotropic effects of statins on cardiovascular and cerebrovascular diseases have been well established; however, the effects of statins on cancer patients have not been fully elucidated and are still controversial. This review discusses the recent evidence on the effects of statins on cardiovascular and cerebrovascular diseases and cancer. Additionally, this study describes the pharmacological action of statins, focusing on the aspect of ‘beyond lipid-lowering’.
Collapse
|
37
|
Xiao Y, Liu Q, Peng N, Li Y, Qiu D, Yang T, Kang R, Usmani A, Amadasu E, Borlongan CV, Yu G. Lovastatin Inhibits RhoA to Suppress Canonical Wnt/β-Catenin Signaling and Alternative Wnt-YAP/TAZ Signaling in Colon Cancer. Cell Transplant 2022; 31:9636897221075749. [PMID: 35168393 PMCID: PMC8855423 DOI: 10.1177/09636897221075749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/25/2021] [Accepted: 01/08/2022] [Indexed: 12/14/2022] Open
Abstract
Statins are first-line drugs used to control patient lipid levels, but there is recent evidence that statin treatment can lower colorectal cancer (CRC) incidence by 50% and prolong CRC patient survival through mechanisms that are poorly understood. In this study, we found that the treatment of APCmin mice by the mevalonate pathway inhibitor lovastatin significantly reduced the number of colonic masses and improved hypersplenism and peripheral anemia. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) analysis of colonic mass tissues showed a potent inhibitory effect in both Wnt/β-catenin signaling and YAP/TAZ signaling in the lovastatin treatment group. The results of our transcriptomic analyses in RKO indicated that lovastatin regulated several proliferation-related signaling pathways. Moreover, lovastatin suppressed important genes and proteins related to the canonical Wnt/β-catenin and alternative Wnt-YAP/TAZ signaling pathways in RKO and SW480 cells, and these effects were rescued by mevalonic acid (MVA), as confirmed through a series of Western blotting, RT-PCR, and reporter assays. Given that statins suppress oncogenic processes primarily through the inhibition of Rho GTPase in the mevalonate pathway, we speculate that lovastatin can inhibit certain Rho GTPases to suppress both canonical Wnt/β-catenin signaling and alternative Wnt-YAP/TAZ signaling. In RKO cells, lovastatin showed similar inhibitory properties as the RhoA inhibitor CCG1423, being able to inhibit β-catenin, TAZ, and p-LATS1 protein activity. Our results revealed that lovastatin inhibited RhoA activity, thereby suppressing the downstream canonical Wnt/β-catenin and alternative Wnt-YAP/TAZ pathways in colon cancer cells. These inhibitory properties suggest the promise of statins as a treatment for CRC. Altogether, the present findings support the potential clinical use of statins in non-cardiovascular contexts and highlight novel targets for anticancer treatments.
Collapse
Affiliation(s)
- Yi Xiao
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Qin Liu
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Nanyin Peng
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Yuzhang Li
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Danyang Qiu
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Tianlun Yang
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| | - Richard Kang
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Ahsan Usmani
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Efosa Amadasu
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Cesario V. Borlongan
- Department of Neurosurgery and Brain
Repair, University of South Florida Morsani College of Medicine, Tampa, FL,
USA
| | - Guolong Yu
- Division of Cardiovascular, Xiangya
Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
Vigneau AL, Rico C, Boerboom D, Paquet M. Statins downregulate YAP and TAZ and exert anti-cancer effects in canine mammary tumour cells. Vet Comp Oncol 2021; 20:437-448. [PMID: 34881506 DOI: 10.1111/vco.12789] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022]
Abstract
Canine mammary tumours (CMTs) are the most common neoplasms in intact bitches, and few chemotherapeutic options are available for highly invasive and metastatic tumours. Recent studies have shown the potential involvement of dysregulated Hippo signalling in CMT development and progression. Statins can activate the Hippo pathway by blocking protein geranylgeranylation (GGylation), resulting in decreased expression and activity of the transcriptional co-activators YAP and TAZ. In this study, we therefore sought to determine if statins could exert anti-cancer effects in CMT cells. Our results demonstrate that Atorvastatin and Fluvastatin are cytotoxic to two CMT cell lines (CMT9 and CMT47), with ED50 values ranging from 0.95 to 23.5 μM. Both statins acted to increase apoptosis and promote cell cycle arrest. Both statins also decreased YAP and TAZ expression and reduced the mRNA levels of key Hippo transcriptional target genes known to be involved in breast cancer progression and chemoresistance (CYR61, CTGF and RHAMM). Moreover, both statins effectively inhibited cell migration and anchorage independent growth, but did not influence matrix invasion. Taken together, our results demonstrate for the first time that statins act upon the Hippo pathway in CMT cells to counteract several molecular and cellular hallmarks of cancer. These findings suggest that targeting the Hippo pathway with statins represents a novel and promising approach for the treatment canine mammary gland cancers.
Collapse
Affiliation(s)
- Anne-Laurence Vigneau
- Département de Pathologie et de Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Charlène Rico
- Département de Biomédecine Vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Derek Boerboom
- Département de Biomédecine Vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marilène Paquet
- Département de Pathologie et de Microbiologie, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
39
|
Yucer N, Ahdoot R, Workman MJ, Laperle AH, Recouvreux MS, Kurowski K, Naboulsi DJ, Liang V, Qu Y, Plummer JT, Gayther SA, Orsulic S, Karlan BY, Svendsen CN. Human iPSC-derived fallopian tube organoids with BRCA1 mutation recapitulate early-stage carcinogenesis. Cell Rep 2021; 37:110146. [PMID: 34965417 PMCID: PMC9000920 DOI: 10.1016/j.celrep.2021.110146] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/09/2021] [Accepted: 11/27/2021] [Indexed: 12/28/2022] Open
Abstract
Germline pathogenic mutations in BReast CAncer (BRCA1) genes are thought to drive normal fallopian tube epithelial (FTE) cell transformation to high-grade serous ovarian cancer. No human models capture the sequence of events for disease initiation and progression. Here, we generate induced pluripotent stem cells (iPSCs) from healthy individuals and young ovarian cancer patients with germline pathogenic BRCA1 mutations (BRCA1mut). Following differentiation into FTE organoids, BRCA1mut lines exhibit cellular abnormalities consistent with neoplastic transformation compared to controls. BRCA1mut organoids show an increased production of cancer-specific proteins and survival following transplantation into mice. Organoids from women with the most aggressive ovarian cancer show the greatest pathology, indicating the potential value to predict clinical severity prior to disease onset. These human FTE organoids from BRCA1mut carriers provide a faithful physiological in vitro model of FTE lesion generation and early carcinogenesis. This platform can be used for personalized mechanistic and drug screening studies. Yucer et al. generate a human BRCA1 mutant iPSC-derived fallopian tube organoid model, which recapitulates BRCA1 mutant ovarian carcinogenesis in vitro and shows tumors in vivo. This model provides a biologically relevant platform to validate drugs and a basis for personalized early detection and preventative strategies for women carrying BRCA1 mutations.
Collapse
|
40
|
Tang YP, Yin YX, Xie MZ, Liang XQ, Li JL, Li KZ, Hu BL. Systematic Analysis of the Clinical Significance of Hyaluronan-Mediated Motility Receptor in Colorectal Cancer. Front Mol Biosci 2021; 8:733271. [PMID: 34765644 PMCID: PMC8575689 DOI: 10.3389/fmolb.2021.733271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/23/2021] [Indexed: 01/01/2023] Open
Abstract
Background: The role of hyaluronan-mediated motility receptor (HMMR) in colorectal cancer (CRC) remains unclear. The present study aimed to explore the association of HMMR with the development and prognosis of CRC using sequence datasets, clinical tissues, blood samples, and cell lines. Methods: CRC datasets were downloaded from TCGA and GEO databases. Forty CRC tissue samples, 120 CRC blood samples, and 100 healthy controls were collected. Four CRC cell lines (HCT116, HT-29, LoVo, and SW480) and one normal human colon mucosal epithelial cell line (NCM460) were cultured. RT-qPCR was used to determine the expression of HMMR in the tissues and cell lines. ELISA was used to measure HMMR levels in the blood samples. Results: The expression of HMMR was significantly increased in CRC tissues than in corresponding adjacent tissues based on TCGA and GEO datasets, and clinical CRC tissues. No associations were found between the expression of HMMR and the TNM stage or other clinical parameters. The expression of HMMR varied in different CRC cell lines. The blood levels of HMMR tended to be higher in patients with CRC than in healthy controls. TCGA and GEO datasets showed inconsistent results regarding the association of HMMR expression with the survival of patients with CRC. Conclusion: The expression of HMMR is increased in CRC tissues but not in the blood. The expression of HMMR is independent of CRC development and has no prognostic significance in patients with CRC.
Collapse
Affiliation(s)
- Yan-Ping Tang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yi-Xin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ming-Zhi Xie
- Department of Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xin-Qiang Liang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ji-Lin Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ke-Zhi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bang-Li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
41
|
Targeting the ILK/YAP axis by LFG-500 blocks epithelial-mesenchymal transition and metastasis. Acta Pharmacol Sin 2021; 42:1847-1859. [PMID: 33879841 PMCID: PMC8563739 DOI: 10.1038/s41401-021-00655-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/14/2021] [Indexed: 02/02/2023]
Abstract
Metastasis is the main cause of mortality in patients with cancer. Epithelial-mesenchymal transition (EMT), a crucial process in cancer metastasis, is an established target for antimetastatic drug development. LFG-500, a novel synthetic flavonoid, has been revealed as a potential antitumor agent owing to its various activities, including modulation of EMT in the inflammatory microenvironment. Here, using a transforming growth factor beta (TGF-β)-induced EMT models, we found that LFG-500 inhibited EMT-associated migration and invasion in human breast cancer, MCF-7, and lung adenocarcinoma, A549, cell lines, consistent with the observed downregulation of YAP activity. Further studies demonstrated that LGF-500-induced suppression of YAP activation was mediated by integrin-linked kinase (ILK), suggesting that the ILK/YAP axis might be feasible target for anti-EMT and antimetastatic treatments, which was verified by a correlation analysis with clinical data and tumor specimens. Hence, our data support the use of LGF-500 as an antimetastatic drug in cancer therapy and provide evidence that the ILK/YAP axis is a feasible biomarker of cancer progression and a promising target for repression of EMT and metastasis in cancer therapy.
Collapse
|
42
|
Rozengurt E, Eibl G. Crosstalk between KRAS, SRC and YAP Signaling in Pancreatic Cancer: Interactions Leading to Aggressive Disease and Drug Resistance. Cancers (Basel) 2021; 13:5126. [PMID: 34680275 PMCID: PMC8533944 DOI: 10.3390/cancers13205126] [Citation(s) in RCA: 21] [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: 09/15/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the predominant form of pancreatic cancer, remains a devastating disease. The purpose of this review is to highlight recent literature on mechanistic and translational developments that advance our understanding of a complex crosstalk between KRAS, YAP and Src tyrosine kinase family (SFK) in PDAC development and maintenance. We discuss recent studies indicating the importance of RAS dimerization in signal transduction and new findings showing that the potent pro-oncogenic members of the SFK phosphorylate and inhibit RAS function. These surprising findings imply that RAS may not play a crucial role in maintaining certain subtypes of PDAC. In support of this interpretation, current evidence indicates that the survival of the basal-like subtype of PDAC is less dependent on RAS but relies, at least in part, on the activity of YAP/TAZ. Based on current evidence, we propose that SFK propels PDAC cells to a state of high metastasis, epithelial-mesenchymal transition (EMT) and reduced dependence on KRAS signaling, salient features of the aggressive basal-like/squamous subtype of PDAC. Strategies for PDAC treatment should consider the opposite effects of tyrosine phosphorylation on KRAS and SFK/YAP in the design of drug combinations that target these novel crosstalk mechanisms and overcome drug resistance.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| |
Collapse
|
43
|
Mehraj U, Ganai RA, Macha MA, Hamid A, Zargar MA, Bhat AA, Nasser MW, Haris M, Batra SK, Alshehri B, Al-Baradie RS, Mir MA, Wani NA. The tumor microenvironment as driver of stemness and therapeutic resistance in breast cancer: New challenges and therapeutic opportunities. Cell Oncol (Dordr) 2021; 44:1209-1229. [PMID: 34528143 DOI: 10.1007/s13402-021-00634-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Breast cancer (BC), the second most common cause of cancer-related deaths, remains a significant threat to the health and wellness of women worldwide. The tumor microenvironment (TME), comprising cellular components, such as cancer-associated fibroblasts (CAFs), immune cells, endothelial cells and adipocytes, and noncellular components such as extracellular matrix (ECM), has been recognized as a critical contributor to the development and progression of BC. The interplay between TME components and cancer cells promotes phenotypic heterogeneity, cell plasticity and cancer cell stemness that impart tumor dormancy, enhanced invasion and metastasis, and the development of therapeutic resistance. While most previous studies have focused on targeting cancer cells with a dismal prognosis, novel therapies targeting stromal components are currently being evaluated in preclinical and clinical studies, and are already showing improved efficacies. As such, they may offer better means to eliminate the disease effectively. CONCLUSIONS In this review, we focus on the evolving concept of the TME as a key player regulating tumor growth, metastasis, stemness, and the development of therapeutic resistance. Despite significant advances over the last decade, several clinical trials focusing on the TME have failed to demonstrate promising effectiveness in cancer patients. To expedite clinical efficacy of TME-directed therapies, a deeper understanding of the TME is of utmost importance. Secondly, the efficacy of TME-directed therapies when used alone or in combination with chemo- or radiotherapy, and the tumor stage needs to be studied. Likewise, identifying molecular signatures and biomarkers indicating the type of TME will help in determining precise TME-directed therapies.
Collapse
Affiliation(s)
- Umar Mehraj
- Department of Bioresources, University of Kashmir, Srinagar, Jammu & Kashmir, India
| | - Rais A Ganai
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology , Awantipora, Jammu & Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology , Awantipora, Jammu & Kashmir, India
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Mohammed A Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Ajaz A Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohammad Haris
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory of Animal Research, Qatar University, Doha, Qatar
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska, Lincoln, NE, USA.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Raid Saleem Al-Baradie
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Manzoor A Mir
- Department of Bioresources, University of Kashmir, Srinagar, Jammu & Kashmir, India.
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India.
| |
Collapse
|
44
|
Strepkos D, Markouli M, Papavassiliou KA, Papavassiliou AG, Piperi C. Emerging roles for the YAP/TAZ transcriptional regulators in brain tumour pathology and targeting options. Neuropathol Appl Neurobiol 2021; 48:e12762. [PMID: 34409639 DOI: 10.1111/nan.12762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
The transcriptional co-activators Yes-associated protein 1/transcriptional co-activator with PDZ-binding motif (YAP/TAZ) have emerged as significant regulators of a wide variety of cellular and organ functions with impact in early embryonic development, especially during the expansion of the neural progenitor cell pool. YAP/TAZ signalling regulates organ size development, tissue homeostasis, wound healing and angiogenesis by participating in a complex network of various pathways. However, recent evidence suggests an association of these physiologic regulatory effects of YAP/TAZ with pro-oncogenic activities. Herein, we discuss the physiological functions of YAP/TAZ as well as the extensive network of signalling pathways that control their expression and activity, leading to brain tumour development and progression. Furthermore, we describe current targeting approaches and drug options including direct YAP/TAZ and YAP-TEA domain transcription factor (TEAD) interaction inhibitors, G-protein coupled receptors (GPCR) signalling modulators and kinase inhibitors, which may be used to successfully attack YAP/TAZ-dependent tumours.
Collapse
Affiliation(s)
- Dimitrios Strepkos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Mariam Markouli
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas A Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
45
|
Cellular feedback dynamics and multilevel regulation driven by the hippo pathway. Biochem Soc Trans 2021; 49:1515-1527. [PMID: 34374419 PMCID: PMC8421037 DOI: 10.1042/bst20200253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
The Hippo pathway is a dynamic cellular signalling nexus that regulates differentiation and controls cell proliferation and death. If the Hippo pathway is not precisely regulated, the functionality of the upstream kinase module is impaired, which increases nuclear localisation and activity of the central effectors, the transcriptional co-regulators YAP and TAZ. Pathological YAP and TAZ hyperactivity consequently cause cancer, fibrosis and developmental defects. The Hippo pathway controls an array of fundamental cellular processes, including adhesion, migration, mitosis, polarity and secretion of a range of biologically active components. Recent studies highlight that spatio-temporal regulation of Hippo pathway components are central to precisely controlling its context-dependent dynamic activity. Several levels of feedback are integrated into the Hippo pathway, which is further synergized with interactors outside of the pathway that directly regulate specific Hippo pathway components. Likewise, Hippo core kinases also ‘moonlight’ by phosphorylating multiple substrates beyond the Hippo pathway and thereby integrates further flexibility and robustness in the cellular decision-making process. This topic is still in its infancy but promises to reveal new fundamental insights into the cellular regulation of this therapeutically important pathway. We here highlight recent advances emphasising feedback dynamics and multilevel regulation of the Hippo pathway with a focus on mitosis and cell migration, as well as discuss potential productive future research avenues that might reveal novel insights into the overall dynamics of the pathway.
Collapse
|
46
|
Wessels DJ, Pujol C, Pradhan N, Lusche DF, Gonzalez L, Kelly SE, Martin EM, Voss ER, Park YN, Dailey M, Sugg SL, Phadke S, Bashir A, Soll DR. Directed movement toward, translocation along, penetration into and exit from vascular networks by breast cancer cells in 3D. Cell Adh Migr 2021; 15:224-248. [PMID: 34338608 PMCID: PMC8331046 DOI: 10.1080/19336918.2021.1957527] [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] [Indexed: 11/07/2022] Open
Abstract
We developed a computer-assisted platform using laser scanning confocal microscopy to 3D reconstruct in real-time interactions between metastatic breast cancer cells and human umbilical vein endothelial cells (HUVECs). We demonstrate that MB-231 cancer cells migrate toward HUVEC networks, facilitated by filopodia, migrate along the network surfaces, penetrate into and migrate within the HUVEC networks, exit and continue migrating along network surfaces. The system is highly amenable to 3D reconstruction and computational analyses, and assessments of the effects of potential anti-metastasis monoclonal antibodies and other drugs. We demonstrate that an anti-RHAMM antibody blocks filopodium formation and all of the behaviors that we found take place between MB-231 cells and HUVEC networks.
Collapse
Affiliation(s)
- Deborah J Wessels
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Claude Pujol
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Nikash Pradhan
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Daniel F Lusche
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Luis Gonzalez
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Sydney E Kelly
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Elizabeth M Martin
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Edward R Voss
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Yang-Nim Park
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Michael Dailey
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| | - Sonia L Sugg
- Department of Surgery, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Sneha Phadke
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Amani Bashir
- Department of Pathology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - David R Soll
- Developmental Studies Hybridoma Bank and W.M. Keck Dynamic Image Analysis Facility, Department of Biology, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
47
|
Nguyen-Lefebvre AT, Selzner N, Wrana JL, Bhat M. The hippo pathway: A master regulator of liver metabolism, regeneration, and disease. FASEB J 2021; 35:e21570. [PMID: 33831275 DOI: 10.1096/fj.202002284rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/04/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
The liver is the only visceral organ in the body with a tremendous capacity to regenerate in response to insults that induce inflammation, cell death, and injury. Liver regeneration is a complicated process involving a well-orchestrated activation of non-parenchymal cells in the injured area and proliferation of undamaged hepatocytes. Furthermore, the liver has a Hepatostat, defined as adjustment of its volume to that required for homeostasis. Understanding the mechanisms that control different steps of liver regeneration is critical to informing therapies for liver repair, to help patients with liver disease. The Hippo signaling pathway is well known for playing an essential role in the control and regulation of liver size, regeneration, stem cell self-renewal, and liver cancer. Thus, the Hippo pathway regulates dynamic cell fates in liver, and in absence of its downstream effectors YAP and TAZ, liver regeneration is severely impaired, and the proliferative expansion of liver cells blocked. We will mainly review upstream mechanisms activating the Hippo signaling pathway following partial hepatectomy in mouse model and patients, its roles during different steps of liver regeneration, metabolism, and cancer. We will also discuss how targeting the Hippo signaling cascade might improve liver regeneration and suppress liver tumorigenesis.
Collapse
Affiliation(s)
- Anh Thu Nguyen-Lefebvre
- Department of Medicine, Multi-Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Nazia Selzner
- Department of Medicine, Multi-Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | | | - Mamatha Bhat
- Department of Medicine, Multi-Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
48
|
Islam MS, Afrin S, Singh B, Jayes FL, Brennan JT, Borahay MA, Leppert PC, Segars JH. Extracellular matrix and Hippo signaling as therapeutic targets of antifibrotic compounds for uterine fibroids. Clin Transl Med 2021; 11:e475. [PMID: 34323413 PMCID: PMC8255059 DOI: 10.1002/ctm2.475] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Uterine fibroids are highly prevalent, collagen-rich, mechanically stiff, fibrotic tumors for which new therapeutic options are needed. Increased extracellular matrix (ECM) stiffness activates mechanical signaling and Hippo/YAP promoting fibroid growth, but no prior studies have tested either as a therapeutic target. We tested the hypothesis that injection of a purified form of collagenase Clostridium histolyticum (CCH) that selectively digests type I and type III collagens would alter ECM stiffness, Hippo signaling, and selectively reduce fibroid cell growth. We also used two FDA-approved drugs, verteporfin and nintedanib, to elucidate the role of Hippo/YAP signaling in uterine fibroid and myometrial cells. METHODS The clinical trial was registered (NCT02889848). Stiffness of samples was measured by rheometry. Protein expression in surgical samples was analyzed via immunofluorescence. Protein and gene expression in uterine fibroid or myometrial cell lines were measured by real time PCR and western blot, and immunofluorescence. RESULTS Injection of CCH at high doses (0.1-0.2 mg/cm3 ) into fibroids resulted in a 46% reduction in stiffness in injected fibroids compared to controls after 60 days. Levels of the cell proliferation marker proliferative cell nuclear antigen (PCNA) were decreased in fibroids 60 days after injection at high doses of CCH. Key Hippo signaling factors, specifically the transcriptionally inactive phosphorylated YAP (p-YAP), was increased at high CCH doses, supporting the role of YAP in fibroid growth. Furthermore, inhibition of YAP via verteporfin (YAP inhibitor) decreased cell proliferation, gene and protein expression of key factors promoting fibrosis and mechanotransduction in fibroid cells. Additionally, the anti-fibrotic drug, nintedanib, inhibited YAP and showed anti-fibrotic effects. CONCLUSIONS This is the first report that in vivo injection of collagenase into uterine fibroids led to a reduction in Hippo/YAP signaling and crucial genes and pathways involved in fibroid growth. These results indicate that targeting ECM stiffness and Hippo signaling might be an effective strategy for uterine fibroids.
Collapse
Affiliation(s)
- Md Soriful Islam
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Sadia Afrin
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Bhuchitra Singh
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Friederike L. Jayes
- Department of Obstetrics and GynecologyDuke UniversityDurhamNorth CarolinaUSA
| | - Joshua T. Brennan
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Mostafa A. Borahay
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| | - Phyllis C. Leppert
- Department of Obstetrics and GynecologyDuke UniversityDurhamNorth CarolinaUSA
| | - James H. Segars
- Department of Gynecology and Obstetrics, Division of Reproductive Sciences & Women's Health ResearchJohns Hopkins MedicineBaltimoreMarylandUSA
| |
Collapse
|
49
|
Eibl G, Rozengurt E. Metformin: review of epidemiology and mechanisms of action in pancreatic cancer. Cancer Metastasis Rev 2021; 40:865-878. [PMID: 34142285 DOI: 10.1007/s10555-021-09977-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma continues to be a lethal disease, for which efficient treatment options are very limited. Increasing efforts have been taken to understand how to prevent or intercept this disease at an early stage. There is convincing evidence from epidemiologic and preclinical studies that the antidiabetic drug metformin possesses beneficial effects in pancreatic cancer, including reducing the risk of developing the disease and improving survival in patients with early-stage disease. This review will summarize the current literature about the epidemiological data on metformin and pancreatic cancer as well as describe the preclinical evidence illustrating the anticancer effects of metformin in pancreatic cancer. Underlying mechanisms and targets of metformin will also be discussed. These include direct effects on transformed pancreatic epithelial cells and indirect, systemic effects on extra-pancreatic tissues.
Collapse
Affiliation(s)
- Guido Eibl
- Department of Surgery, David Geffen School of Medicine At UCLA, Los Angeles, CA, USA.
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine At UCLA, Los Angeles, CA, USA
| |
Collapse
|
50
|
Singhatanadgit W, Hankamolsiri W, Janvikul W. Geranylgeraniol prevents zoledronic acid-mediated reduction of viable mesenchymal stem cells via induction of Rho-dependent YAP activation. ROYAL SOCIETY OPEN SCIENCE 2021; 8:202066. [PMID: 34113452 PMCID: PMC8187992 DOI: 10.1098/rsos.202066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/23/2021] [Indexed: 05/03/2023]
Abstract
Long-term use of zoledronic acid (ZA) increases the risk of medication-related osteonecrosis of the jaw (MRONJ). This may be attributed to ZA-mediated reduction of viable mesenchymal stem cells (MSCs). ZA inhibits protein geranylgeranylation, thus suppressing cell viability and proliferation. Geranylgeraniol (GGOH), which is a naturally found intermediate compound in the mevalonate pathway, has positive effects against ZA. However, precise mechanisms by which GGOH may help preserve stem cell viability against ZA are not fully understood. The objective of this study was to investigate the cytoprotective mechanisms of GGOH against ZA. The results showed that while ZA dramatically decreased the number of viable MSCs, GGOH prevented this negative effect. GGOH-rescued ZA-exposed MSCs formed mineralization comparable to that produced by normal MSCs. Mechanistically, GGOH preserved the number of viable MSCs by its reversal of ZA-mediated Ki67+ MSC number reduction, cell cycle arrest and apoptosis. Moreover, GGOH prevented ZA-suppressed RhoA activity and YAP activation. The results also established the involvement of Rho-dependent YAP and YAP-mediated CDK6 in the cytoprotective ability of GGOH against ZA. In conclusion, GGOH preserves a pool of viable MSCs with osteogenic potency against ZA by rescuing the activity of Rho-dependent YAP activation, suggesting GGOH as a promising agent and YAP as a potential therapeutic target for MRONJ.
Collapse
Affiliation(s)
- Weerachai Singhatanadgit
- Faculty of Dentistry, Thammasat University, Pathumthani, 12121, Thailand
- Research Unit in Mineralized Tissue Reconstruction, Thammasat University, Pathumthani, 12121, Thailand
| | - Weerawan Hankamolsiri
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, Pathumthani 12120, Thailand
| | - Wanida Janvikul
- Biofunctional Materials and Devices Research Group, National Metal and Materials Technology Center, Pathumthani 12120, Thailand
| |
Collapse
|