1
|
Zhang H, Ma T, Wen X, Jiang J, Chen J, Jiang J, Xie J, Mo T, Li R, Xie H, Liang G, Wang L, Zheng Z, Huang X, Liu C, Baihetiyaer Y, Abulimiti A, He X, Chen Z, Hu T, Pan W. SIK1 promotes ferroptosis resistance in pancreatic cancer via HDAC5-STAT6-SLC7A11 axis. Cancer Lett 2025; 623:217726. [PMID: 40250791 DOI: 10.1016/j.canlet.2025.217726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The activation of protein kinases is ubiquitous in pancreatic ductal adenocarcinoma (PDAC), yet its impact on ferroptosis remains unclear. SIK1 was identified as a key regulator of ferroptosis resistance in PDAC by kinase database screening. Targeting SIK1 could significantly reverse ferroptosis resistance and enhance cytotoxic effects of gemcitabine via increasing ferroptosis sensitivity in PDAC cells. Mechanistically, SIK1 phosphorylated HDAC5 at Ser498 residue and promoted its interaction with 14-3-3 protein, which further protected HDAC5 from TRIM28-mediated ubiquitylation and degradation. SIK1-stabilized HDAC5 deacetylated STAT6 and enhanced its transcriptional activity to upregulate SLC7A11 expression, ultimately rendering PDAC cells resistance to ferroptosis. SIK1 inhibitor (YKL-05-099) could synergistically enhance the antitumor effects of gemcitabine in organoid and patient-derived xenograft (PDX) models by inducing ferroptosis, suggesting a novel therapeutic target for PDAC. Clinically, SIK1 was positively correlated with SLC7A11 expression in PDAC specimens, which was associated with poor prognosis. These findings unveil a crucial mechanism through which PDAC counters ferroptosis via SIK1-mediated HDAC5 stabilization and subsequent SLC7A11 upregulation. This study underscores the promising potential of targeting SIK1-HDAC5 axis as a therapeutic strategy to overcome drug resistance in PDAC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Ma
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Wen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianlong Jiang
- Department of Gastrointestinal Surgery, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Jing Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junfeng Jiang
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiancong Xie
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Taiwei Mo
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruibing Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanlin Xie
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanzhan Liang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Wang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zheyu Zheng
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Huang
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuanyuan Liu
- Department of General Surgery, The Ganzhou People's Hospital, Ganzhou, China
| | - Yimamu Baihetiyaer
- Department of General Surgery, The First People's Hospital of Kashgar Region, Kashgar, China
| | - Abuduhalike Abulimiti
- Department of General Surgery, The Second People's Hospital of Kashgar Region, Kashgar, China
| | - Xiaosheng He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zexian Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Tuo Hu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Weidong Pan
- Department of General Surgery (Pancreatic Hepatobiliary Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Li C, Syed MU, Nimbalkar A, Shen Y, Vieira MD, Fraser C, Inde Z, Qin X, Ouyang J, Kreuzer J, Clark SE, Kelley G, Hensley EM, Morris R, Lazaro R, Belmonte B, Oh A, Walcott M, Nabel CS, Caenepeel S, Saiki AY, Rex K, Lipford JR, Heist RS, Lin JJ, Haas W, Sarosiek K, Hughes PE, Hata AN. LKB1 regulates JNK-dependent stress signaling and apoptotic dependency of KRAS-mutant lung cancers. Nat Commun 2025; 16:4112. [PMID: 40316540 PMCID: PMC12048556 DOI: 10.1038/s41467-025-58753-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025] Open
Abstract
The efficacy of molecularly targeted therapies may be limited by co-occurring mutations within a tumor. Conversely, these alterations may confer collateral vulnerabilities that can be therapeutically leveraged. KRAS-mutant lung cancers are distinguished by recurrent loss of the tumor suppressor STK11/LKB1. Whether LKB1 modulates cellular responses to therapeutic stress seems unknown. Here we show that in LKB1-deficient KRAS-mutant lung cancer cells, inhibition of KRAS or its downstream effector MEK leads to hyperactivation of JNK due to loss of NUAK-mediated PP1B phosphatase activity. JNK-mediated inhibitory phosphorylation of BCL-XL rewires apoptotic dependencies, rendering LKB1-deficient cells vulnerable to MCL-1 inhibition. These results uncover an unknown role for LKB1 in regulating stress signaling and mitochondrial apoptosis independent of its tumor suppressor activity mediated by AMPK and SIK. Additionally, our study reveals a therapy-induced vulnerability in LKB1-deficient KRAS-mutant lung cancers that could be exploited as a genotype-informed strategy to improve the efficacy of KRAS-targeted therapies.
Collapse
Affiliation(s)
- Chendi Li
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Yi Shen
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Cameron Fraser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zintis Inde
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xingping Qin
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jian Ouyang
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Biochemistry & Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Johannes Kreuzer
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah E Clark
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Grace Kelley
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Emily M Hensley
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Robert Morris
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Raul Lazaro
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Audris Oh
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Makeba Walcott
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Christopher S Nabel
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Koch Institute for Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Anne Y Saiki
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | - Karen Rex
- Amgen Research, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Rebecca S Heist
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica J Lin
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Kristopher Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Lab for Systems Pharmacology, Harvard Program in Therapeutics Science, Harvard Medical School, Boston, MA, USA
| | | | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Zhang C, Yin X, Jiang J, Wang P, Wang Y. Downregulation of GFPT2 enhances cisplatin chemotherapy sensitivity in STK11/KRAS mutant non-small cell lung cancer by regulating the hexosamine biosynthesis pathway, resisting tumor growth. Cytokine 2025; 191:156943. [PMID: 40253947 DOI: 10.1016/j.cyto.2025.156943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE To explore the role of GFPT2 in the sensitivity of STK11/KRAS lung cancer cells to cisplatin chemotherapy, and its underlying mechanism. MATERIALS & METHODS A549 and H460 cells were used to analyze the effect of GFPT2 on cisplatin chemotherapy sensitivity, as both carry KRAS mutations and H460 has LKB1 inactivation mutations, meeting the requirements of a KRAS/LKB1 co mutant tumor model. The levels of UDP-GlcNAc, OGT, OGA, and O-GlcNAc in the HBP pathway were also determined. To verify the potential role of HBP, we added OGT inhibitors. In vivo, we constructed a nude mouse model bearing A549 tumor to further validate the results of in vitro cell experiments. RESULTS GFPT2 silencing can significantly inhibit cell proliferation, invasion, and migration, promote cell apoptosis, and enhance the effect of cisplatin (p < 0.05). After OSMI-1 processing, GFPT2 enhances O-GlcNAc modification levels via the OGT-mediated HBP, thereby decreasing the sensitivity of STK11/KRAS mutant cells to cisplatin chemotherapy. In addition, GFPT2 silencing enhances the chemotherapy sensitivity of cisplatin and inhibits tumor growth, while overexpression of GFPT2 weakens this effect (p < 0.05). The above results provide new targets and combination therapy options for the clinical treatment of KRAS/LKB1 mutant lung cancer. CONCLUSION Our study found that inhibiting GFPT2 can enhance the chemotherapy sensitivity of cisplatin to STK11/KRAS/LKB1 mutant NSCLCs cells through the OGT mediated HBP pathway, filling a key gap in the chemotherapy resistance mechanism of KRAS/LKB1 mutant lung cancer.
Collapse
Affiliation(s)
- Cheng Zhang
- Thoracic Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Xuelei Yin
- Yantai Keyou Biotechnology Co., Ltd, Yantai, Shandong, China
| | - Jun Jiang
- Key Laboratory of Genetics Research and Evaluation of the National Drug Administration, Shandong Food and Drug Inspection and Research Institute, Jinan, Shandong, China
| | - Peng Wang
- Ministry of Scientific and Technological Innovation, Yantai Hi-tech Industrial Development Zone, Yantai, Shandong, China
| | - Yirong Wang
- Department of Radiotherapy, Yantaishan Hospital, Yantai, Shandong, China.
| |
Collapse
|
4
|
Huang G, Lin W, Jiang T, Cai Y, Lin C, Sun P. Serine/threonine kinase 11 (STK11) associated adnexal tumors: from biology to therapeutic impact. Hum Genomics 2025; 19:28. [PMID: 40102938 PMCID: PMC11921566 DOI: 10.1186/s40246-025-00741-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Female adnexal malignancies, while relatively uncommon, exhibit high mortality rates due to often-late diagnosis. The serine/threonine kinase 11 (STK11) is a tumor suppressor gene, and its inactivation or mutation often leads to an autosomal dominant genetic disorder known as Peutz-Jeghers syndrome (PJS), which is associated with ovarian and cervical cancers. STK11-associated adnexal tumors mostly originate from the ovary, with a low incidence rate but high metastasis rates worldwide. In addition to surgery and chemotherapy, it is necessary to optimize relevant screening policy and targeted therapy. STK11-associated adnexal tumors are difficult to diagnose by histopathology. Although genetic testing involves higher costs, it can serve as a primary preventive measure for high-risk populations with STK11-associated tumors. A more intensive screening program (MISP) is needed for individuals with significant clinical symptoms and a family history of PJS. These tumors may be adequately treated with fertility-sparing surgery in young women with lower malignant potential tumors. Prophylactic adnexectomy, chemotherapy, and immunotherapy may offer potential clinical benefits but also pose significant challenges. Therefore, surgery should be undertaken with careful and comprehensive consideration of the patient's age, reproductive history, risk of malignancy, genetic mutation lineages, post-operative complications, and other conditions. Further research is essential to develop better screening, diagnostic, and treatment strategies.
Collapse
Affiliation(s)
- Guanxiang Huang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18 Daoshan Road, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Wenyu Lin
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18 Daoshan Road, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Tingting Jiang
- Department of Gynecology and Obstetrics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Yuanjun Cai
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
| | - Chengbin Lin
- Department of Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18 Daoshan Road, Fuzhou, Fujian, 350001, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| |
Collapse
|
5
|
Middleton G, Robbins HL, Fletcher P, Savage J, Mehmi M, Summers Y, Greystoke A, Steele N, Popat S, Jain P, Spicer J, Cave J, Shaw P, Gilligan D, Power D, Fennell D, Bajracharya M, McBride DJ, Maheswari U, Frankell AM, Swanton C, Beggs AD, Billingham L. A phase II trial of mTORC1/2 inhibition in STK11 deficient non small cell lung cancer. NPJ Precis Oncol 2025; 9:67. [PMID: 40069402 PMCID: PMC11897347 DOI: 10.1038/s41698-025-00838-4] [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: 07/30/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025] Open
Abstract
There are no current stratified medicine options for STK11-deficient NSCLC. STK11 loss mediates mTORC activation, GLUT1 up-regulation and increased glycolysis. This metabolic reprogramming might represent a therapeutic vulnerability targetable with mTORC1/2 inhibition. In arm B2 of the National Lung Matrix Trial 54 patients with NSCLC received vistusertib, of which 49 were STK11-deficient (30 with KRAS mutation (B2D), 19 without (B2S)). Objective response (OR) and durable clinical benefit (DCB) rates with 95% credible intervals (CrI) were estimated from posterior probability distributions generated using Bayesian beta-binomial conjugate analysis. In B2D, 2 per-protocol patients obtained OR (estimated true OR rate (95%CrI) 9.8% (2.4-24.3). Estimates of true DCB rate (95%CrI): B2D 24.4% (11.1-42.3), B2S 14.6% (3.6-34.7). Overall, vistusertib cannot be recommended in this context. Longitudinal ctDNA analysis demonstrates enrichment of SMARCA4 mutations post-treatment. In vitro studies show adaptive resistance to mTORC1/2 inhibition via AKT reactivation. (NCT02664935, ISRCTN38344105, EudraCT 2014-000814-73, 10 June 2015).
Collapse
Affiliation(s)
- Gary Middleton
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
- Department of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Helen L Robbins
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Peter Fletcher
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Joshua Savage
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Manita Mehmi
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | | | | | | | | | - Pooja Jain
- St James's University Hospital, Leeds, UK
| | - James Spicer
- King's College London, Guy's Hospital, London, UK
| | - Judith Cave
- Southampton University Hospitals NHS Trust, Southampton, UK
| | | | | | | | | | | | | | | | - Alexander M Frankell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Andrew D Beggs
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Cancer & Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Lucinda Billingham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| |
Collapse
|
6
|
Shu C, Li J, Rui J, Fan D, Niu Q, Bai R, Cicka D, Doyle S, Wahafu A, Zheng X, Du Y, Ivanov AA, Doxie DB, Dhodapkar KM, Carlisle J, Owonikoko T, Sica G, Liu Y, Ramalingam S, Dhodapkar MV, Zhou W, Mo X, Fu H. Uncovering the rewired IAP-JAK regulatory axis as an immune-dependent vulnerability of LKB1-mutant lung cancer. Nat Commun 2025; 16:2324. [PMID: 40057483 PMCID: PMC11890758 DOI: 10.1038/s41467-025-57297-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
Harnessing the power of immune system to treat cancer has become a core clinical approach. However, rewiring of intrinsic circuitry by genomic alterations enables tumor cells to escape immune surveillance, leading to therapeutic failure. Uncovering the molecular basis of how tumor mutations induce therapeutic resistance may guide the development of intervention approaches to advance precision immunotherapy. Here we report the identification of the Liver Kinase B1 (LKB1)-Inhibitor of Apoptosis Protein (IAP)- Janus Kinase 1 (JAK1) dynamic complex as a molecular determinant for immune response of LKB1-mut lung cancer cells. LKB1 alteration exposes a critical dependency of lung cancer cells on IAP for their immune resistance. Indeed, pharmacological inhibition of IAP re-establishes JAK1-regulated Stimulator of interferon genes (STING) expression and DNA sensing signaling, enhances cytotoxic immune cell infiltration, and augmentes immune-dependent anti-tumor activity in an LKB1-mutant immune-competent mouse model. Thus, IAP-JAK1-targeted strategies, like IAP inhibitors, may offer a promising therapeutic approach to restore the responsiveness of immunologically-cold LKB1-mutant tumors to immune checkpoint inhibitors or STING-directed therapies.
Collapse
Affiliation(s)
- Changfa Shu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, P R China
| | - Jianfeng Li
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jin Rui
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Dacheng Fan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Qiankun Niu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ruiyang Bai
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Danielle Cicka
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sean Doyle
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Alafate Wahafu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shannxi, P R China
| | - Xi Zheng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P R China
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Deon B Doxie
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Kavita M Dhodapkar
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | - Jennifer Carlisle
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Taofeek Owonikoko
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Gabriel Sica
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Suresh Ramalingam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Xiulei Mo
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
- Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
7
|
Trelford CB, Shepherd TG. Insights into targeting LKB1 in tumorigenesis. Genes Dis 2025; 12:101402. [PMID: 39735555 PMCID: PMC11681833 DOI: 10.1016/j.gendis.2024.101402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 12/31/2024] Open
Abstract
Genetic alterations to serine-threonine kinase 11 (STK11) have been implicated in Peutz-Jeghers syndrome and tumorigenesis. Further exploration of the context-specific roles of liver kinase B1 (LKB1; encoded by STK11) observed that it regulates AMP-activated protein kinase (AMPK) and AMPK-related kinases. Given that both migration and proliferation are enhanced with the loss of LKB1 activity combined with the prevalence of STK11 genetic alterations in cancer biopsies, LKB1 was marked as a tumor suppressor. However, the role of LKB1 in tumorigenesis is paradoxical as LKB1 activates autophagy and reactive oxygen species scavenging while dampening anoikis, which contribute to cancer cell survival. Due to the pro-tumorigenic properties of LKB1, targeting LKB1 pathways is now relevant for cancer treatment. With the recent successes of targeting LKB1 signaling in research and clinical settings, and enhanced cytotoxicity of chemical compounds in LKB1-deficient tumors, there is now a need for LKB1 inhibitors. However, validating LKB1 inhibitors is challenging as LKB1 adaptor proteins, nucleocytoplasmic shuttling, and splice variants all manipulate LKB1 activity. Furthermore, STE-20-related kinase adaptor protein (STRAD) and mouse protein 25 dictate LKB1 cellular localization and kinase activity. For these reasons, prior to assessing the efficacy and potency of pharmacological candidates, the functional status of LKB1 needs to be defined. Therefore, to improve the understanding of LKB1 in physiology and oncology, this review highlights the role of LKB1 in tumorigenesis and addresses the therapeutic relevancy of LKB1 inhibitors.
Collapse
Affiliation(s)
- Charles B. Trelford
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Trevor G. Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
8
|
Gritti I, Wan J, Weeresekara V, Vaz JM, Tarantino G, Bryde TH, Vijay V, Kammula AV, Kattel P, Zhu S, Vu P, Chan M, Wu MJ, Gordan JD, Patra KC, Silveira VS, Manguso RT, Wein MN, Ott CJ, Qi J, Liu D, Sakamoto K, Gujral TS, Bardeesy N. DNAJB1-PRKACA Fusion Drives Fibrolamellar Liver Cancer through Impaired SIK Signaling and CRTC2/p300-Mediated Transcriptional Reprogramming. Cancer Discov 2025; 15:382-400. [PMID: 39326063 PMCID: PMC11803398 DOI: 10.1158/2159-8290.cd-24-0634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024]
Abstract
SIGNIFICANCE This work combines functional studies in model systems and examination of human tumor specimens to define a central oncogenic pathway driven by DNAJB1-PRKACA fusions in FLC. DNAJB1-PRKACA-mediated inactivation of the SIK stimulates CRTC2-p300-mediated transcription to drive tumor growth. The findings illuminate pathogenic mechanisms and inform therapeutic development.
Collapse
Affiliation(s)
- Ilaria Gritti
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Jinkai Wan
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Vajira Weeresekara
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Joel M. Vaz
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Giuseppe Tarantino
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tenna Holgersen Bryde
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Vindhya Vijay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Ashwin V. Kammula
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Prabhat Kattel
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Songli Zhu
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Phuong Vu
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Marina Chan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Meng-Ju Wu
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - John D. Gordan
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, California
| | | | - Vanessa S. Silveira
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Robert T. Manguso
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Marc N. Wein
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher J. Ott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Jun Qi
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Liu
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Nabeel Bardeesy
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| |
Collapse
|
9
|
Onda DA, Zhu Y, Yuan X, Loh K. Central and Peripheral Roles of Salt-inducible Kinases in Metabolic Regulation. Endocrinology 2025; 166:bqaf024. [PMID: 39919030 DOI: 10.1210/endocr/bqaf024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/09/2025]
Abstract
Salt-inducible kinases (SIKs), a member of the serine/threonine protein kinase family, have recently garnered considerable research interest as one of the emerging key regulators of metabolism. The 3 SIK isoforms-SIK1, SIK2, and SIK3-exhibit diverse roles both in central and peripheral physiological processes. While early studies focused on their role in inflammation, spurring the development of SIK inhibitors for chronic inflammatory diseases currently in clinical trials, emerging evidence highlights their broader functions in metabolism. In this review, we will summarize the current state of research on the central roles of SIKs in the brain, particularly in regulating energy balance and glucose homeostasis, alongside their peripheral functions in critical metabolic tissues such as the liver, adipose tissue, and pancreas. By integrating insights into their central and peripheral roles, this review underscores the importance of SIKs in maintaining metabolic homeostasis and highlights their therapeutic potential as novel targets for metabolic disease.
Collapse
Affiliation(s)
- Danise-Ann Onda
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
| | - Yifei Zhu
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
| | - XiaoZhuo Yuan
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
| | - Kim Loh
- Diabetes and Metabolic Disease, St. Vincent's Institute of Medical Research, Melbourne, VIC 3065, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
10
|
Zhong ZT, Wang XY, Pan Y, Zhou K, Chen JH, Gao YQ, Dai B, Zhou ZL, Wang RQ. AMPK: An energy sensor for non-small cell lung cancer progression and treatment. Pharmacol Res 2025; 212:107592. [PMID: 39805353 DOI: 10.1016/j.phrs.2025.107592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Lung cancer (LC) is the leading cause of cancer-related morbidity and mortality in China, with non-small cell lung cancer (NSCLC) accounting for 85 % of the overall lung cancer cases. AMP-activated protein kinase (AMPK) is a key regulator of energy balance and homeostasis, and its dysregulation is a common feature in various malignancies, particularly in NSCLC with mutations in Liver kinase B1 (LKB1). Studies have shown that the AMPK signalling pathway has a dual role in NSCLC progression, both inhibiting and promoting the progression of malignant tumours. Therefore, drugs targeting the AMPK signalling pathway may hold significant promise for therapeutic application in NSCLC. This review aims to examine the manifestations and mechanisms by which AMPK and its associated signalling molecules influence NSCLC progression and treatment. Firstly, we discuss the critical importance of AMPK within the mutational context of NSCLC. Secondly, we summarise the drugs and related substances that modulate the AMPK signalling pathway in NSCLC and evaluate the evidence from preclinical studies on combination AMPK-targeted therapies to address the issue of drug resistance in NSCLC under current clinical treatments. In summary, this paper highlights the critical importance of developing AMPK-targeted drugs to enhance therapeutic efficacy in NSCLC, as well as the potential for applying these drugs in clinical therapy to overcome drug resistance.
Collapse
Affiliation(s)
- Zhi-Ting Zhong
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China; College of Pharmacy, Jinan University, Guangzhou, China
| | - Xu-Yan Wang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ying Pan
- Department of Oncology, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ke Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Jing-Hui Chen
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Yu-Qi Gao
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Bo Dai
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan City, Guangdong Province 528200, China.
| | - Zhi-Ling Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| | - Rui-Qi Wang
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| |
Collapse
|
11
|
Huang Q, Tang X, Gan C, Deng Q, Zhi S, Huang Q, Zheng X, Li X, Pan Z, Huang M. EFHD1 Activates SIK3 to Limit Colorectal Cancer Initiation and Progression via the Hippo Pathway. J Cancer 2025; 16:1348-1362. [PMID: 39895792 PMCID: PMC11786025 DOI: 10.7150/jca.103229] [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: 09/04/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers, with high rates of metastasis and lethality. EF-hand domain-containing protein D1 (EFHD1) and salt-inducible kinase 3 (SIK3) have been studied in several cancer types. Aberrant expression of EFHD1 and SIK3 has been observed in CRC, but little research has addressed their regulatory abilities and signaling pathways. In this study, we aimed to explore the efficacy of EFHD1 in inhibiting CRC proliferation and metastasis and to elucidate the underlying mechanisms involved in the upregulation of SIK3 expression. Cell viability, colony formation, wound healing, Transwell assay, orthotopic xenograft, and pulmonary metastasis mouse models were used to detect the antiproliferative and anti-metastatic effects of EFHD1 against CRC in vitro and in vivo. The Gene Expression Profiling Interactive Analysis (GEPIA) database was used to determine EFHD1 and SIK3 expression in CRC. The regulatory roles of EFHD1 and SIK3 in mediating anti-metastatic effects in CRC were measured using western blotting, immunohistochemical, and immunofluorescence analyses. The results showed that EFHD1 expression was significantly repressed in the clinical CRC samples. EFHD1 markedly suppressed cell proliferation, migration, and invasion in vitro and inhibited tumor growth and metastasis in vivo. Analysis of the GEPIA database revealed that EFHD1 expression positively correlated with SIK3 expression. SIK3 overexpression inhibited the migration of CRC cells, and SIK3 knockdown partially eliminated the inhibitory effects of EFHD1 on CRC metastasis. EFHD1 exerted anti-metastatic effects against CRC via upregulating SIK3 and inhibiting epithelial-mesenchymal transition (EMT) processing through modulating the Hippo signaling pathway. Collectively, these findings identify EFHD1 as a potent SIK3 agonist and highlight the EFHD1-SIK3 axis as a key modulator of the Hippo signaling pathway in CRC. EFHD1 serves as a novel regulator and is worthy of further development as a novel therapeutic target in CRC.
Collapse
Affiliation(s)
- Qionghui Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Xiaoyan Tang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Caiyan Gan
- Institute of basic medical sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Qiaoting Deng
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Shaobin Zhi
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
- GuangDong Engineering Technological Research Center of Molecular Diagnosis in Cardiovascular Diseases, Meizhou, China
| | - Qingyan Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Xiaoqi Zheng
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Xueqiong Li
- Medical College of Jiaying University, Meizhou, China
| | - Zengfeng Pan
- Institute of basic medical sciences, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Mingfeng Huang
- Institute of Cardiovascular Disease, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| |
Collapse
|
12
|
Yao Y, Qiu X, Chen M, Qin Z, Zhang X, Zhang W. LKB1 Mutations Enhance Radiosensitivity in Non-Small Cell Lung Cancer Cells by Inducing G2/M Cell Cycle Phase Arrest. Curr Mol Med 2025; 25:353-360. [PMID: 38310550 DOI: 10.2174/0115665240280822231221060656] [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: 09/23/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND Radiosensitivity remains an important factor affecting the clinical outcome of radiotherapy for non-small cell lung cancer (NSCLC). Liver kinase B1 (LKB1) as a tumor suppressor, is one of the most commonly mutated genes in NSCLC. However, the role of LKB1 on radiosensitivity and the possible mechanism have not been elucidated in the NSCLC. In this study, we investigated the regulatory function of LKB1 in the radiosensitivity of NSCLC cells and its possible signaling pathways. METHODS After regulating the expression of LKB1, cell proliferation was determined by Cell Counting Kit-8 (CCK-8) assay. The flow cytometry assay was used to analyse cell cycle distribution. Survival fraction and sensitization enhancement ratio (SER) were generated by clonogenic survival assay. Western blot analysis was used to assess expression levels of LKB1, p53, p21, γ-H2AX and p-Chk2. RESULTS Our study found that when the NSCLC cells were exposed to ionizing radiation, LKB1 could inhibit NSCLC cell proliferation by promoting DNA double strand break and inducing DNA repair. In addition, LKB1 could induce NSCLC cells G1 and G2/M phase arrest through up-regulating expression of p53 and p21 proteins. CONCLUSION This current study demonstrates that LKB1 enhances the radiosensitivity of NSCLC cells via inhibiting NSCLC cell proliferation and inducing G2/M phase arrest, and the mechanism of cell cycle arrest associated with signaling pathways of p53 and p21 probably.
Collapse
Affiliation(s)
- Yuanhu Yao
- Department of Radiation Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xiangnan Qiu
- Department of Radiation Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Graduate School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Meng Chen
- Department of Radiation Oncology, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Zhaohui Qin
- Research Center for Medical and Health Emergency Rescue, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xinjun Zhang
- Graduate School of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Radiation Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Zhang
- Graduate School of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Radiation Oncology, The Third People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Manoharan R. Salt-inducible kinases (SIKs) in cancer: mechanisms of action and therapeutic prospects. Drug Discov Today 2025; 30:104279. [PMID: 39710233 DOI: 10.1016/j.drudis.2024.104279] [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: 11/15/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
Salt-inducible kinases (SIKs), a group of serine/threonine kinases in the adenosine monophosphate-activated protein kinase (AMPK) family, exist in three isoforms: SIK1, SIK2 and SIK3. These kinases are crucial in various physiological processes. Emerging evidence indicates that dysregulation of SIK expression and activation significantly contributes to carcinogenesis by promoting cellular proliferation, metabolic dysregulation, metastasis and chemoresistance through the modulation of crucial signaling pathways. The role of SIKs in cancer progression and metastasis involves complex mechanisms that vary among cancer types. Additionally, research on SIK inhibitors suggests that targeting these kinases might offer promising avenues for improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Ravi Manoharan
- Cell Signaling and Cancer Biology Laboratory, Department of Biochemistry, Guindy Campus, University of Madras, Chennai 600025, India.
| |
Collapse
|
14
|
Zhao Z, Zhao Q, Mao Z, Tian Y, Yang L, Ma Y, Gu J, Tan R. High-throughput screening of the natural STK11 agonist dauricine: A biphenylisoquinoline alkaloid exerting anti-NSCLC effects and reversing gefitinib resistance. Eur J Pharmacol 2024; 984:177024. [PMID: 39362388 DOI: 10.1016/j.ejphar.2024.177024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Serine/threonine kinase 11 (STK11) deletion and downregulation caused cancer progression, and were widely associated with drug resistance. Accurate screening of natural small molecules about anti-cancer and anti-drug resistance is the key to the development and utilization of natural product application, which could promote traditional Chinese medicine in the treatment of cancer. Dauricine, which is derived from the rhizome of Menispermum dauricum DC., has certain potential but unexplored mechanism for the treatment of cancer. PURPOSE The aim of this study was to screen and validate the role and mechanism of natural STK11 agonists with anti-drug resistance from plants in the treatment of NSCLC. METHODS A lentiviral STK11 overexpression cell model was employed for the screening of natural STK11 agonists. The efficacy of dauricine in the treatment of NSCLC was validated on PC-9 and HCC827 cells. In vivo validation of dauricine activity was performed using nude mouse models equipped with PC9 xenografts. To investigate the anti-resistant effects of dauricine, gefitinib-resistant PC9 cell models were constructed. RESULTS As a natural agonist of STK11, it causes the activation of the STK11/AMPK pathway and inhibits the growth of PC-9 cells. Dauricine synergises the inhibitory effect with gefitinib on PC9. The up-regulation of STK11 protein expression by dauricine was demonstrated in vitro and in vivo, while restoring the sensitivity of PC9/GR to gefitinib by down-regulating the protein expression of Nrf2 and Pgp. CONCLUSION Dauricine, a natural agonist of STK11, effectively inhibited NSCLC, and its combination treatment with gefitinib reversed drug-resistant NSCLC.
Collapse
Affiliation(s)
- Zhigao Zhao
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhiyuan Mao
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yang Tian
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Li Yang
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yu Ma
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu, 610225, China
| | - Rui Tan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
15
|
Zhang X, Liu J, Zuo C, Peng X, Xie J, Shu Y, Ao D, Zhang Y, Ye Q, Cai J. Role of SIK1 in tumors: Emerging players and therapeutic potentials (Review). Oncol Rep 2024; 52:169. [PMID: 39422046 PMCID: PMC11544583 DOI: 10.3892/or.2024.8828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Salt‑induced kinase 1 (SIK1) is a serine/threonine protein kinase that is a member of the AMP‑activated protein kinase family. SIK is catalytically activated through its phosphorylation by the upstream kinase LKB1. SIK1 has been reported to be associated with numerous types of cancer. The present review summarizes the structure, regulatory factors and inhibitors of SIK1, and also describes how SIK1 is a signal regulatory factor that fulfills connecting roles in various signal regulatory pathways. Furthermore, the anti‑inflammatory effects of SIK1 during the early stage of tumor occurrence and its different regulatory effects following tumor occurrence, are summarized, and through collating the tumor signal regulatory mechanisms in which SIK1 participates, it has been demonstrated that SIK1 acts as a necessary node in cancer signal transduction. In conclusion, SIK1 is discussed independent of the SIKs family, its research results and recent progress in oncology are summarized in detail with a focus on SIK1, and its potential as a therapeutic target is highlighted, underscoring the need for SIK1‑targeted regulatory strategies in future cancer therapy.
Collapse
Affiliation(s)
- Xinran Zhang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Jing Liu
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Chenyang Zuo
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Xiaochun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jinyuan Xie
- Department of Joint Surgery and Sports Medicine, Jingmen Central Hospital, Jingmen, Hubei 448000, P.R. China
| | - Ya Shu
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Dongxu Ao
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Yang Zhang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Qingqing Ye
- Department of Breast Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jun Cai
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
16
|
Skoulidis F, Araujo HA, Do MT, Qian Y, Sun X, Cobo AG, Le JT, Montesion M, Palmer R, Jahchan N, Juan JM, Min C, Yu Y, Pan X, Arbour KC, Vokes N, Schmidt ST, Molkentine D, Owen DH, Memmott R, Patil PD, Marmarelis ME, Awad MM, Murray JC, Hellyer JA, Gainor JF, Dimou A, Bestvina CM, Shu CA, Riess JW, Blakely CM, Pecot CV, Mezquita L, Tabbó F, Scheffler M, Digumarthy S, Mooradian MJ, Sacher AG, Lau SCM, Saltos AN, Rotow J, Johnson RP, Liu C, Stewart T, Goldberg SB, Killam J, Walther Z, Schalper K, Davies KD, Woodcock MG, Anagnostou V, Marrone KA, Forde PM, Ricciuti B, Venkatraman D, Van Allen EM, Cummings AL, Goldman JW, Shaish H, Kier M, Katz S, Aggarwal C, Ni Y, Azok JT, Segal J, Ritterhouse L, Neal JW, Lacroix L, Elamin YY, Negrao MV, Le X, Lam VK, Lewis WE, Kemp HN, Carter B, Roth JA, Swisher S, Lee R, Zhou T, Poteete A, Kong Y, Takehara T, Paula AG, Parra Cuentas ER, Behrens C, Wistuba II, Zhang J, Blumenschein GR, Gay C, Byers LA, Gibbons DL, Tsao A, Lee JJ, Bivona TG, Camidge DR, Gray JE, Leighl NB, Levy B, Brahmer JR, Garassino MC, et alSkoulidis F, Araujo HA, Do MT, Qian Y, Sun X, Cobo AG, Le JT, Montesion M, Palmer R, Jahchan N, Juan JM, Min C, Yu Y, Pan X, Arbour KC, Vokes N, Schmidt ST, Molkentine D, Owen DH, Memmott R, Patil PD, Marmarelis ME, Awad MM, Murray JC, Hellyer JA, Gainor JF, Dimou A, Bestvina CM, Shu CA, Riess JW, Blakely CM, Pecot CV, Mezquita L, Tabbó F, Scheffler M, Digumarthy S, Mooradian MJ, Sacher AG, Lau SCM, Saltos AN, Rotow J, Johnson RP, Liu C, Stewart T, Goldberg SB, Killam J, Walther Z, Schalper K, Davies KD, Woodcock MG, Anagnostou V, Marrone KA, Forde PM, Ricciuti B, Venkatraman D, Van Allen EM, Cummings AL, Goldman JW, Shaish H, Kier M, Katz S, Aggarwal C, Ni Y, Azok JT, Segal J, Ritterhouse L, Neal JW, Lacroix L, Elamin YY, Negrao MV, Le X, Lam VK, Lewis WE, Kemp HN, Carter B, Roth JA, Swisher S, Lee R, Zhou T, Poteete A, Kong Y, Takehara T, Paula AG, Parra Cuentas ER, Behrens C, Wistuba II, Zhang J, Blumenschein GR, Gay C, Byers LA, Gibbons DL, Tsao A, Lee JJ, Bivona TG, Camidge DR, Gray JE, Leighl NB, Levy B, Brahmer JR, Garassino MC, Gandara DR, Garon EB, Rizvi NA, Scagliotti GV, Wolf J, Planchard D, Besse B, Herbst RS, Wakelee HA, Pennell NA, Shaw AT, Jänne PA, Carbone DP, Hellmann MD, Rudin CM, Albacker L, Mann H, Zhu Z, Lai Z, Stewart R, Peters S, Johnson ML, Wong KK, Huang A, Winslow MM, Rosen MJ, Winters IP, Papadimitrakopoulou VA, Cascone T, Jewsbury P, Heymach JV. CTLA4 blockade abrogates KEAP1/STK11-related resistance to PD-(L)1 inhibitors. Nature 2024; 635:462-471. [PMID: 39385035 PMCID: PMC11560846 DOI: 10.1038/s41586-024-07943-7] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 08/13/2024] [Indexed: 10/11/2024]
Abstract
For patients with advanced non-small-cell lung cancer (NSCLC), dual immune checkpoint blockade (ICB) with CTLA4 inhibitors and PD-1 or PD-L1 inhibitors (hereafter, PD-(L)1 inhibitors) is associated with higher rates of anti-tumour activity and immune-related toxicities, when compared with treatment with PD-(L)1 inhibitors alone. However, there are currently no validated biomarkers to identify which patients will benefit from dual ICB1,2. Here we show that patients with NSCLC who have mutations in the STK11 and/or KEAP1 tumour suppressor genes derived clinical benefit from dual ICB with the PD-L1 inhibitor durvalumab and the CTLA4 inhibitor tremelimumab, but not from durvalumab alone, when added to chemotherapy in the randomized phase III POSEIDON trial3. Unbiased genetic screens identified loss of both of these tumour suppressor genes as independent drivers of resistance to PD-(L)1 inhibition, and showed that loss of Keap1 was the strongest genomic predictor of dual ICB efficacy-a finding that was confirmed in several mouse models of Kras-driven NSCLC. In both mouse models and patients, KEAP1 and STK11 alterations were associated with an adverse tumour microenvironment, which was characterized by a preponderance of suppressive myeloid cells and the depletion of CD8+ cytotoxic T cells, but relative sparing of CD4+ effector subsets. Dual ICB potently engaged CD4+ effector cells and reprogrammed the tumour myeloid cell compartment towards inducible nitric oxide synthase (iNOS)-expressing tumoricidal phenotypes that-together with CD4+ and CD8+ T cells-contributed to anti-tumour efficacy. These data support the use of chemo-immunotherapy with dual ICB to mitigate resistance to PD-(L)1 inhibition in patients with NSCLC who have STK11 and/or KEAP1 alterations.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- AMP-Activated Protein Kinase Kinases/genetics
- AMP-Activated Protein Kinase Kinases/metabolism
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- B7-H1 Antigen/metabolism
- B7-H1 Antigen/antagonists & inhibitors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/immunology
- Clinical Trials, Phase III as Topic
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Kelch-Like ECH-Associated Protein 1/genetics
- Kelch-Like ECH-Associated Protein 1/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Mutation
- Nitric Oxide Synthase Type II/metabolism
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Genes, Tumor Suppressor
Collapse
Affiliation(s)
- Ferdinandos Skoulidis
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Haniel A Araujo
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Minh Truong Do
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yu Qian
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Sun
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Galan Cobo
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John T Le
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | - Yi Yu
- Tango Therapeutics, Boston, MA, USA
| | | | - Kathryn C Arbour
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natalie Vokes
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie T Schmidt
- Department of Genomic Medicine and the Institute for Data Science in Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Molkentine
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dwight H Owen
- Division of Medical Oncology, Ohio State University-James Comprehensive Cancer Center, Columbus, OH, USA
| | - Regan Memmott
- Division of Medical Oncology, Ohio State University-James Comprehensive Cancer Center, Columbus, OH, USA
| | - Pradnya D Patil
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Melina E Marmarelis
- Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mark M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joseph C Murray
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Jonathan W Riess
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | | - Chad V Pecot
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Laura Mezquita
- Department of Medical Oncology, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Matthias Scheffler
- Department of Internal Medicine, Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Subba Digumarthy
- Department of Radiology, Massachussetts General Hospital, Boston, MA, USA
| | | | | | - Sally C M Lau
- Department of Medical Oncology, NYU Langone Perlmutter Cancer Center, New York, NY, USA
| | - Andreas N Saltos
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Julia Rotow
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rocio Perez Johnson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Corinne Liu
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tyler Stewart
- Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | | | | | - Zenta Walther
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Kurt Schalper
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Kurtis D Davies
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mark G Woodcock
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Valsamo Anagnostou
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristen A Marrone
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick M Forde
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deepti Venkatraman
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eliezer M Van Allen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy L Cummings
- David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | - Jonathan W Goldman
- David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | | | - Melanie Kier
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sharyn Katz
- Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Charu Aggarwal
- Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Ni
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph T Azok
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeremy Segal
- Department of Pathology, University of Chicago, Chicago, USA
| | | | - Joel W Neal
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Yasir Y Elamin
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcelo V Negrao
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiuning Le
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vincent K Lam
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Whitney E Lewis
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haley N Kemp
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brett Carter
- Department of Thoracic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen Swisher
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Lee
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Teng Zhou
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alissa Poteete
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yifan Kong
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomohiro Takehara
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alvaro Guimaraes Paula
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra Cuentas
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmen Behrens
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George R Blumenschein
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carl Gay
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren A Byers
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anne Tsao
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Trever G Bivona
- University of California San Francisco, San Francisco, CA, USA
| | | | - Jhannelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - Benjamin Levy
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie R Brahmer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - David R Gandara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Edward B Garon
- David Geffen School of Medicine at the University of California, Los Angeles, CA, USA
| | | | | | - Jürgen Wolf
- Department of Internal Medicine, Center for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | | | | | | | | | | | - Alice T Shaw
- Novartis Institute for Biomedical Research, Cambridge, MA, USA
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David P Carbone
- Division of Medical Oncology, Ohio State University-James Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - Melissa L Johnson
- Sarah Cannon Research Institute, Tennessee Oncology, Nashville, TN, USA
| | - Kwok K Wong
- Division of Hematology & Medical Oncology, NYU Langone Perlmutter Cancer Center, New York, NY, USA
| | | | - Monte M Winslow
- D2G Oncology, Mountain View, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | | | | | - Tina Cascone
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Zhang Y, Ren J, Liao Z, Li X, Zhang C, Huang B, Cao Y, Chen J. Downregulating LKB1 in bone marrow mesenchymal stem cells could inhibit CD4 + T cell proliferation via the PD-1/PD-L1 signaling pathway. Immunobiology 2024; 229:152856. [PMID: 39369651 DOI: 10.1016/j.imbio.2024.152856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Our previous research has shown that LKB1 in amniotic mesenchymal stem cells (MSCs) serves as a vital regulator of regulatory T cell differentiation and T cell proliferation, which may have a similar role in bone marrow MSCs (BMMSCs). Therefore, we investigated the role of LKB1 in BMMSCs for regulating CD4+ T cell proliferation in the bone micro-environment of AML. METHODS RT-PCR was used to assessed LKB1 expression in BMMSCs derived from AML patients and healthy controls. Subsequently, LKB1 was knocked down in the BMMSCs line HS-5 (HS-5-LKB1KD). Co-cultures in vitro were established to analyze the effect of HS-5-LKB1KD on CD4+ T cell. Flow cytometry was employed to measure PD-L1 and CD4+ T cell proliferation levels. Western blot was utilized to detect related proteins. RESULTS The expression of LKB1 in BMMSCs derived from AML patients was decreased. Knockdown of LKB1 in HS-5 resulted in upregulation of PD-L1 expression. Co-culture of peripheral blood CD4+ T cell with HS-5-LKB1KD exhibited reduced CD4+ T cell proliferation compared to co-culture with HS-5-LKB1con. Furthermore, blocking PD-L1 in the co-culture conditions could restore the reduced CD4+ T cell proliferation. Additionally, it was found that upregulation of the Wnt signaling pathway-related proteins following LKB1 knockdown in HS-5, indicating that downregulating LKB1 could promote PD-L1 expression through activation of the Wnt signaling pathway. CONCLUSIONS The decreased expression of LKB1 in BMMSCs may activate the Wnt signaling pathway, leading to increased PD-L1 expression. This inhibited CD4+ T cell proliferation, which might lead to impaired anti-tumor immunity in AML patients and promote AML progression.
Collapse
Affiliation(s)
- Yaqin Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Jingyi Ren
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Zhongxian Liao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Xiaoyu Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Chunying Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Bihan Huang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China.
| | - Jiadi Chen
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China.
| |
Collapse
|
18
|
Kang J, Gallucci S, Pan J, Oakhill JS, Sanij E. The role of STK11/LKB1 in cancer biology: implications for ovarian tumorigenesis and progression. Front Cell Dev Biol 2024; 12:1449543. [PMID: 39544365 PMCID: PMC11560430 DOI: 10.3389/fcell.2024.1449543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
STK11 (serine-threonine kinase 11), also known as LKB1 (liver kinase B1) is a highly conserved master kinase that regulates cellular metabolism and polarity through a complex signaling network involving AMPK and 12 other AMPK-related kinases. Germline mutations in LKB1 have been causatively linked to Peutz-Jeghers Syndrome (PJS), an autosomal dominant hereditary disease with high cancer susceptibility. The identification of inactivating somatic mutations in LKB1 in different types of cancer further supports its tumor suppressive role. Deleterious mutations in LKB1 are frequently observed in patients with epithelial ovarian cancer. However, its inconsistent effects on tumorigenesis and cancer progression suggest that its functional impact is genetic context-dependent, requiring cooperation with other oncogenic lesions. In this review, we summarize the pleiotropic functions of LKB1 and how its altered activity in cancer cells is linked to oncogenic proliferation and growth, metastasis, metabolic reprogramming, genomic instability, and immune modulation. We also review the current mechanistic understandings of this master kinase as well as therapeutic implications with particular focus on the effects of LKB1 deficiency in ovarian cancer pathogenesis. Lastly, we discuss whether LKB1 deficiency can be exploited as an Achilles heel in ovarian cancer.
Collapse
Affiliation(s)
- Jian Kang
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Stefano Gallucci
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Junqi Pan
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Jonathan S. Oakhill
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Elaine Sanij
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine-St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
19
|
McGuire CK, Meehan AS, Couser E, Bull L, Minor AC, Kuhlmann-Hogan A, Kaech SM, Shaw RJ, Eichner LJ. Transcriptional repression by HDAC3 mediates T cell exclusion from Kras mutant lung tumors. Proc Natl Acad Sci U S A 2024; 121:e2317694121. [PMID: 39388266 PMCID: PMC11494357 DOI: 10.1073/pnas.2317694121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/29/2024] [Indexed: 10/12/2024] Open
Abstract
Histone Deacetylase 3 (HDAC3) function in vivo is nuanced and directed in a tissue-specific fashion. The importance of HDAC3 in Kras mutant lung tumors has recently been identified, but HDAC3 function in this context remains to be fully elucidated. Here, we identified HDAC3 as a lung tumor cell-intrinsic transcriptional regulator of the tumor immune microenvironment. In Kras mutant lung cancer cells, we found that HDAC3 is a direct transcriptional repressor of a cassette of secreted chemokines, including Cxcl10. Genetic and pharmacological inhibition of HDAC3 robustly up-regulated this gene set in human and mouse Kras, LKB1 (KL) and Kras, p53 (KP) mutant lung cancer cells through an NF-κB/p65-dependent mechanism. Using genetically engineered mouse models, we found that HDAC3 inactivation in vivo induced expression of this gene set selectively in lung tumors and resulted in enhanced T cell recruitment at least in part via Cxcl10. Furthermore, we found that inhibition of HDAC3 in the presence of Kras pathway inhibitors dissociated Cxcl10 expression from that of immunosuppressive chemokines and that combination treatment of entinostat with trametinib enhanced T cell recruitment into lung tumors in vivo. Finally, we showed that T cells contribute to in vivo tumor growth control in the presence of entinostat and trametinib combination treatment. Together, our findings reveal that HDAC3 is a druggable endogenous repressor of T cell recruitment into Kras mutant lung tumors.
Collapse
Affiliation(s)
- Caroline K. McGuire
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL60611
| | - Ambryn S. Meehan
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL60611
| | - Evan Couser
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL60611
| | - Lois Bull
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL60611
| | - Allegra C. Minor
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL60611
| | - Alexandra Kuhlmann-Hogan
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Reuben J. Shaw
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La JollaCA92037
| | - Lillian J. Eichner
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL60611
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La JollaCA92037
| |
Collapse
|
20
|
Ghazi PC, O'Toole KT, Srinivas Boggaram S, Scherzer MT, Silvis MR, Zhang Y, Bogdan M, Smith BD, Lozano G, Flynn DL, Snyder EL, Kinsey CG, McMahon M. Inhibition of ULK1/2 and KRAS G12C controls tumor growth in preclinical models of lung cancer. eLife 2024; 13:RP96992. [PMID: 39213022 PMCID: PMC11364435 DOI: 10.7554/elife.96992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent U.S. Food and Drug Administration approval of covalent inhibitors of KRASG12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRASG12C-driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients who do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRASG12C, efforts are underway to develop effective combination therapies. Here, we report that the inhibition of KRASG12C signaling increases autophagy in KRASG12C-expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRASG12C-driven lung cancer cell proliferation in vitro and superior tumor control in vivo. Additionally, in genetically engineered mouse models of KRASG12C-driven NSCLC, inhibition of either KRASG12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRASG12C in lung cancer.
Collapse
Affiliation(s)
- Phaedra C Ghazi
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Kayla T O'Toole
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Sanjana Srinivas Boggaram
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Michael T Scherzer
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Mark R Silvis
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Yun Zhang
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | | | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | - Eric L Snyder
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Pathology, University of UtahSalt Lake CityUnited States
| | - Conan G Kinsey
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Internal Medicine, Division of Medical Oncology, University of UtahSalt Lake CityUnited States
| | - Martin McMahon
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Dermatology, University of UtahSalt Lake CityUnited States
| |
Collapse
|
21
|
Magrath JW, Espinosa-Cotton M, Flinchum DA, Sampath SS, Cheung NK, Lee SB. Desmoplastic small round cell tumor: from genomics to targets, potential paths to future therapeutics. Front Cell Dev Biol 2024; 12:1442488. [PMID: 39139449 PMCID: PMC11319132 DOI: 10.3389/fcell.2024.1442488] [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: 06/02/2024] [Accepted: 06/26/2024] [Indexed: 08/15/2024] Open
Abstract
Desmoplastic Small Round Cell Tumor (DSRCT) is a highly aggressive pediatric cancer caused by a reciprocal translocation between chromosomes 11 and 22, leading to the formation of the EWSR1::WT1 oncoprotein. DSRCT presents most commonly in the abdominal and pelvic peritoneum and remains refractory to current treatment regimens which include chemotherapy, radiotherapy, and surgery. As a rare cancer, sample and model availability have been a limiting factor to DSRCT research. However, the establishment of rare tumor banks and novel cell lines have recently propelled critical advances in the understanding of DSRCT biology and the identification of potentially promising targeted therapeutics. Here we review model and dataset availability, current understanding of the EWSR1::WT1 oncogenic mechanism, and promising preclinical therapeutics, some of which are now advancing to clinical trials. We discuss efforts to inhibit critical dependencies including NTRK3, EGFR, and CDK4/6 as well as novel immunotherapy strategies targeting surface markers highly expressed in DSRCT such as B7-H3 or neopeptides either derived from or driven by the fusion oncoprotein. Finally, we discuss the prospect of combination therapies and strategies for prioritizing clinical translation.
Collapse
Affiliation(s)
- Justin W. Magrath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Madelyn Espinosa-Cotton
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dane A. Flinchum
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Shruthi Sanjitha Sampath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Nai Kong Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sean B. Lee
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
22
|
Chen G, Li MY, Yang JY, Zhou ZH. Will AMPK be a potential therapeutic target for hepatocellular carcinoma? Am J Cancer Res 2024; 14:3241-3258. [PMID: 39113872 PMCID: PMC11301289 DOI: 10.62347/yavk1315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer is the disease that poses the greatest threat to human health today. Among them, hepatocellular carcinoma (HCC) is particularly prominent due to its high recurrence rate and extremely low five-year postoperative survival rate. In addition to surgical treatment, radiotherapy, chemotherapy, and immunotherapy are the main methods for treating HCC. Due to the natural drug resistance of chemoradiotherapy and targeted drugs, satisfactory results have not been achieved in terms of therapeutic efficacy and cost. AMP-Activated Protein Kinase (AMPK) is a serine/threonine protein kinase. It mainly coordinates the metabolism and transformation of energy between cells, which maintaining a balance between energy supply and demand. The processes of cell growth, proliferation, autophagy, and survival all involve various reaction of cells to energy changes. The regulatory role of AMPK in cellular energy metabolism plays an important role in the occurrence, development, treatment, and prognosis of HCC. Here, we reviewed the latest progress on the regulatory role of AMPK in the occurrence and development of HCC. Firstly, the molecular structure and activation mechanism of AMPK were introduced. Secondly, the emerging regulator related to AMPK and tumors were elaborated. Next, the multitasking roles of AMPK in the occurrence and development mechanism of HCC were discussed separately. Finally, the translational implications and the challenges of AMPK-targeted therapies for HCC treatment were elaborated. In summary, these pieces of information suggest that AMPK can serve as a promising specific therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Guo Chen
- Department of Oncology, Anhui Hospital, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineHefei, Anhui, China
| | - Ming-Yuan Li
- Department of Oncology, Anhui Hospital, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineHefei, Anhui, China
| | - Jing-Yi Yang
- Department of Oncology, Feixi Hospital of Traditional Chinese MedicineFeixi, Hefei, Anhui, China
| | - Zhen-Hua Zhou
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese MedicineShanghai, China
| |
Collapse
|
23
|
Lee HM, Muhammad N, Lieu EL, Cai F, Mu J, Ha YS, Cao G, Suchors C, Joves K, Chronis C, Li K, Ducker GS, Olszewski K, Cai L, Allison DB, Bachert SE, Ewing WR, Wong H, Seo H, Kim IY, Faubert B, Kim J, Kim J. Concurrent loss of LKB1 and KEAP1 enhances SHMT-mediated antioxidant defence in KRAS-mutant lung cancer. Nat Metab 2024; 6:1310-1328. [PMID: 38877143 PMCID: PMC11809267 DOI: 10.1038/s42255-024-01066-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 05/16/2024] [Indexed: 06/16/2024]
Abstract
Non-small-cell lung cancer (NSCLC) with concurrent mutations in KRAS and the tumour suppressor LKB1 (KL NSCLC) is refractory to most therapies and has one of the worst predicted outcomes. Here we describe a KL-induced metabolic vulnerability associated with serine-glycine-one-carbon (SGOC) metabolism. Using RNA-seq and metabolomics data from human NSCLC, we uncovered that LKB1 loss enhanced SGOC metabolism via serine hydroxymethyltransferase (SHMT). LKB1 loss, in collaboration with KEAP1 loss, activated SHMT through inactivation of the salt-induced kinase (SIK)-NRF2 axis and satisfied the increased demand for one-carbon units necessary for antioxidant defence. Chemical and genetic SHMT suppression increased cellular sensitivity to oxidative stress and cell death. Further, the SHMT inhibitor enhanced the in vivo therapeutic efficacy of paclitaxel (first-line NSCLC therapy inducing oxidative stress) in KEAP1-mutant KL tumours. The data reveal how this highly aggressive molecular subtype of NSCLC fulfills their metabolic requirements and provides insight into therapeutic strategies.
Collapse
Affiliation(s)
- Hyun Min Lee
- Department of Urology, Yale School of Medicine, New Haven, CT, USA
| | - Nefertiti Muhammad
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Elizabeth L Lieu
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Feng Cai
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jiawei Mu
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Guoshen Cao
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Chamey Suchors
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kenneth Joves
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Constantinos Chronis
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Kailong Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Gregory S Ducker
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | | | - Ling Cai
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, TX, USA
| | - Derek B Allison
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Sara E Bachert
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | - Harvey Wong
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hyosun Seo
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Isaac Y Kim
- Department of Urology, Yale School of Medicine, New Haven, CT, USA
| | - Brandon Faubert
- Department of Medicine-Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - James Kim
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jiyeon Kim
- Department of Urology, Yale School of Medicine, New Haven, CT, USA.
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
24
|
Ghazi PC, O'Toole KT, Srinivas Boggaram S, Scherzer MT, Silvis MR, Zhang Y, Bogdan M, Smith BD, Lozano G, Flynn DL, Snyder EL, Kinsey CG, McMahon M. Inhibition of ULK1/2 and KRAS G12C controls tumor growth in preclinical models of lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579200. [PMID: 38370808 PMCID: PMC10871191 DOI: 10.1101/2024.02.06.579200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent FDA approval of covalent inhibitors of KRAS G12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRAS G12C -driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients that do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRAS G12C , efforts are underway to develop effective combination therapies. Here we report that inhibition of KRAS G12C signaling increases autophagy in KRAS G12C expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRAS G12C -driven lung cancer cell proliferation in vitro and superior tumor control in vivo . Additionally, in genetically engineered mouse models of KRAS G12C -driven NSCLC, inhibition of either KRAS G12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRAS G12C in lung cancer.
Collapse
|
25
|
Yao G, Yu S, Hou F, Xiao Z, Li G, Ji X, Wang J. Rab3B enhances the stabilization of DDX6 to promote lung adenocarcinoma aggressiveness. Mol Med 2024; 30:75. [PMID: 38834947 PMCID: PMC11151598 DOI: 10.1186/s10020-024-00848-1] [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: 07/31/2023] [Accepted: 05/28/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Liver kinase B1 (LKB1) is frequently mutated in lung adenocarcinoma, and its loss contributes to tumor progression. METHODS To identify LKB1 downstream genes that promote lung adenocarcinoma aggressiveness, we performed bioinformatical analysis using publicly available datasets. RESULTS Rab3B was upregulated in LKB1-depleted lung adenocarcinoma cells and suppressed by LKB1 overexpression. CREB protein was enriched at the promoter of Rab3B in lung cancer cells. Silencing of CREB abrogated the upregulation of Rab3B upon LKB1 loss. Immunohistochemistry revealed the elevated expression of Rab3B in lung adenocarcinomas relative to adjacent normal tissues. Upregulation of Rab3B was significantly associated with lymph node metastasis, advanced tumor stage, and reduced overall survival in lung adenocarcinoma patients. Knockdown of Rab3B suppressed and overexpression of Rab3B promoted the proliferation, colony formation, and migration of lung adenocarcinoma cells in vitro. In a mouse xenograft model, Rab3B depletion restrained and Rab3B overexpression augmented the growth of lung adenocarcinoma tumors. Mechanistically, Rab3B interacted with DDX6 and enhanced its protein stability. Ectopic expression of DDX6 significantly promoted the proliferation, colony formation, and migration of lung adenocarcinoma cells. DDX6 knockdown phenocopied the effects of Rab3B depletion on lung adenocarcinoma cells. Additionally, DDX6 overexpression partially rescued the aggressive phenotype of Rab3B-depleted lung adenocarcinoma cells. CONCLUSION LKB1 deficiency promotes Rab3B upregulation via a CREB-dependent manner. Rab3B interacts with and stabilizes DDX6 protein to accelerate lung adenocarcinoma progression. The Rab3B-DDX6 axis may be potential therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Guodong Yao
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Shan Yu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feng Hou
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Zunyu Xiao
- Department of Imaging, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guangqi Li
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Xiaobin Ji
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Jigang Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong Province, China.
| |
Collapse
|
26
|
Ferrarone JR, Thomas J, Unni AM, Zheng Y, Nagiec MJ, Gardner EE, Mashadova O, Li K, Koundouros N, Montalbano A, Mustafa M, Cantley LC, Blenis J, Sanjana NE, Varmus H. Genome-wide CRISPR screens in spheroid culture reveal that the tumor suppressor LKB1 inhibits growth via the PIKFYVE lipid kinase. Proc Natl Acad Sci U S A 2024; 121:e2403685121. [PMID: 38743625 PMCID: PMC11127050 DOI: 10.1073/pnas.2403685121] [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: 02/22/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor-suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1-mediated growth suppression, we developed a spheroid-based cell culture assay to study LKB1-dependent growth. We then performed genome-wide CRISPR screens in spheroidal culture and found that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase. Finally, we used chemical inhibitors and a pH-sensitive reporter to determine that LKB1 impairs growth by promoting the internalization of wild-type EGFR in a PIKFYVE-dependent manner.
Collapse
Affiliation(s)
- John R. Ferrarone
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY10021
| | - Jerin Thomas
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Arun M. Unni
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Yuxiang Zheng
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Michal J. Nagiec
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10021
| | - Eric E. Gardner
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | | | - Kate Li
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Nikos Koundouros
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10021
| | - Antonino Montalbano
- New York Genome Center, New York, NY10013
- Department of Biology, New York University, New York, NY10003
| | - Meer Mustafa
- New York Genome Center, New York, NY10013
- Department of Biology, New York University, New York, NY10003
| | - Lewis C. Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Medicine, Weill Cornell Medicine, New York, NY10021
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10021
| | - Neville E. Sanjana
- New York Genome Center, New York, NY10013
- Department of Biology, New York University, New York, NY10003
| | - Harold Varmus
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Medicine, Weill Cornell Medicine, New York, NY10021
| |
Collapse
|
27
|
Zheng J, Deng Y, Huang B, Chen X. Prognostic implications of STK11 with different mutation status and its relationship with tumor-infiltrating immune cells in non-small cell lung cancer. Front Immunol 2024; 15:1387896. [PMID: 38736875 PMCID: PMC11082287 DOI: 10.3389/fimmu.2024.1387896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Background Mutations in STK11 (STK11Mut) gene may present a negative impact on survival in Non-small Cell Lung Cancer (NSCLC) patients, however, its relationship with immune related genes remains unclear. This study is to unveil whether overexpressed- and mutated-STK11 impact survival in NSCLC and to explore whether immune related genes (IRGs) are involved in STK11 mutations. Methods 188 NSCLC patients with intact formalin-fixed paraffin-embedded (FFPE) tissue available for detecting STK11 protein expression were included in the analysis. After immunohistochemical detection of STK11 protein, patients were divided into high STK11 expression group (STK11High) and low STK11 expression group (STK11Low), and then Kaplan-Meier survival analysis and COX proportional hazards model were used to compare the overall survival (OS) and progression-free survival (PFS) of the two groups of patients. In addition, the mutation data from the TCGA database was used to categorize the NSCLC population, namely STK11 Mutated (STK11Mut) and wild-type (STK11Wt) subgroups. The difference in OS between STK11Mut and STK11Wt was compared. Finally, bioinformatics analysis was used to compare the differences in IRGs expression between STK11Mut and STK11Wt populations. Results The median follow-up time was 51.0 months (range 3.0 - 120.0 months) for real-life cohort. At the end of follow-up, 64.36% (121/188) of patients experienced recurrence or metastasis. 64.89% (122/188) of patients ended up in cancer-related death. High expression of STK11 was a significant protective factor for NSCLC patients, both in terms of PFS [HR=0.42, 95% CI= (0.29-0.61), P<0.001] and OS [HR=0.36, 95% CI= (0.25, 0.53), P<0.001], which was consistent with the finding in TCGA cohorts [HR=0.76, 95%CI= (0.65, 0.88), P<0.001 HR=0.76, 95%CI= (0.65, 0.88), P<0.001]. In TCGA cohort, STK11 mutation was a significant risk factor for NSCLC in both lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) histology in terms of OS [HR=6.81, 95%CI= (2.16, 21.53), P<0.001; HR=1.50, 95%CI= (1.00, 2.26), P=0.051, respectively]. Furthermore, 7 IRGs, namely CALCA, BMP6, S100P, THPO, CGA, PCSK1 and MUC5AC, were found significantly overexpressed in STK11-mutated NSCLC in both LUSC and LUAD histology. Conclusions Low STK11 expression at protein level and presence of STK11 mutation were associated with poor prognosis in NSCLC, and mutated STK11 might probably alter the expression IRGs profiling.
Collapse
Affiliation(s)
- Jianqing Zheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yujie Deng
- Department of Medical Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Bifen Huang
- Department of Obstetrics and Gynecology, Quanzhou Medical College People’s Hospital Affiliated, Quanzhou, Fujian, China
| | - Xiaohui Chen
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Interdisciplinary Institute of Medical Engineering of Fuzhou University, Fuzhou, Fujian, China
| |
Collapse
|
28
|
Abdolvand M, Chermahini ZM, Bahaloo S, Emami MH, Fahim A, Rahimi H, Amjadi E, Maghool F, Rohani F, Dadkhah M, Farhadian N, Vatandoust N, Abdolvand S, Darehsari MR, Chehelgerdi M, Beni FA, Khodadoostan M, Hemati S, Salehi M. New long noncoding RNA biomarkers and ceRNA networks on miR-616-3p in colorectal cancer: Bioinformatics-based study. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:10. [PMID: 38524750 PMCID: PMC10956565 DOI: 10.4103/jrms.jrms_786_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 10/18/2023] [Accepted: 11/01/2023] [Indexed: 03/26/2024]
Abstract
Background Cancer development is aided by the role of long noncoding RNAs (lncRNAs) that act as competing endogenous RNAs (ceRNAs) absorbing microRNAs (miRNAs). We aimed to discover a novel regulatory axis in colorectal cancer (CRC) and potential biomarkers based on miR-616-3p. Materials and Methods The gene expression omnibus database was mined for differentially expressed lncRNAs (DELs) and mRNAs. LncRNAs and mRNAs were predicted using the RegRNA and TargetScan databases. A combination of the ciBioPortal and Ensemble databases was used to locate the mRNAs. Cytoscape 3.7.1-built CeRNA networks. A quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to confirm the expression levels of these RNA molecules. Statistical analyses were implemented by GraphPad Prism 9. Results qRT-PCR showed (Linc01282, lnc-MYADM-1:1, and Zinc Finger Protein 347 [ZNF347]) were overexpressed whereas, (salt-inducible kinases 1 [SIK1], and miR-616-3p) were down regulated. Conclusion These results identify unique, unreported lncRNAs as CRC prognostic biomarkers, as well as prospective mRNAs as new treatment targets and predictive biomarkers for CRC. In addition, our study uncovered unexplored ceRNA networks that should be studied further in CRC.
Collapse
Affiliation(s)
- Mohammad Abdolvand
- Cellular, Molecular and Genetics Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Medical Genetics Research Center of Genome, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Mohammadi Chermahini
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sahar Bahaloo
- Department of Biology, Faculty of Sciences, Yazd University, Yazd, Iran
| | - Mohammad Hassan Emami
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Fahim
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hojjatolah Rahimi
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Amjadi
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Maghool
- Poursina Hakim Digestive Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fattah Rohani
- Faculty of Veterinary Medicine of Shahrekord, Shahrekord, Iran
| | - Mina Dadkhah
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nooshin Farhadian
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasimeh Vatandoust
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Abdolvand
- Department of Genetics, Faculty of Sciences, Islamic Azad University, Shahrekord, Iran
| | | | - Mohammad Chehelgerdi
- Department of Genetics, Faculty of Sciences, Islamic Azad University, Shahrekord, Iran
| | - Faeze Ahmadi Beni
- Cellular, Molecular and Genetics Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Medical Genetics Research Center of Genome, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Khodadoostan
- Department of Gastroenterology and Hepatology, AlZahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Simin Hemati
- Department of Radiooncology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansoor Salehi
- Cellular, Molecular and Genetics Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Medical Genetics Research Center of Genome, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
29
|
Gong X, Du J, Peng RW, Chen C, Yang Z. CRISPRing KRAS: A Winding Road with a Bright Future in Basic and Translational Cancer Research. Cancers (Basel) 2024; 16:460. [PMID: 38275900 PMCID: PMC10814442 DOI: 10.3390/cancers16020460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Once considered "undruggable" due to the strong affinity of RAS proteins for GTP and the structural lack of a hydrophobic "pocket" for drug binding, the development of proprietary therapies for KRAS-mutant tumors has long been a challenging area of research. CRISPR technology, the most successful gene-editing tool to date, is increasingly being utilized in cancer research. Here, we provide a comprehensive review of the application of the CRISPR system in basic and translational research in KRAS-mutant cancer, summarizing recent advances in the mechanistic understanding of KRAS biology and the underlying principles of drug resistance, anti-tumor immunity, epigenetic regulatory networks, and synthetic lethality co-opted by mutant KRAS.
Collapse
Affiliation(s)
- Xian Gong
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; (X.G.); (J.D.)
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou 350001, China
| | - Jianting Du
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; (X.G.); (J.D.)
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou 350001, China
| | - Ren-Wang Peng
- Division of General Thoracic Surgery, Department of BioMedical Research (DBMR), Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008 Bern, Switzerland;
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; (X.G.); (J.D.)
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou 350001, China
| | - Zhang Yang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; (X.G.); (J.D.)
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
30
|
Ross FA, Hawley SA, Russell FM, Goodman N, Hardie DG. Frequent loss-of-function mutations in the AMPK-α2 catalytic subunit suggest a tumour suppressor role in human skin cancers. Biochem J 2023; 480:1951-1968. [PMID: 37962491 PMCID: PMC10754287 DOI: 10.1042/bcj20230380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/15/2023]
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status activated by increases in AMP or ADP relative to ATP. Once activated, it phosphorylates targets that promote ATP-generating catabolic pathways or inhibit ATP-consuming anabolic pathways, helping to restore cellular energy balance. Analysis of human cancer genome studies reveals that the PRKAA2 gene (encoding the α2 isoform of the catalytic subunit) is often subject to mis-sense mutations in cancer, particularly in melanoma and non-melanoma skin cancers, where up to 70 mis-sense mutations have been documented, often accompanied by loss of the tumour suppressor NF1. Recently it has been reported that knockout of PRKAA2 in NF1-deficient melanoma cells promoted anchorage-independent growth in vitro, as well as growth as xenografts in immunodeficient mice in vivo, suggesting that AMPK-α2 can act as a tumour suppressor in that context. However, very few of the mis-sense mutations in PRKAA2 that occur in human skin cancer and melanoma have been tested to see whether they cause loss-of-function. We have addressed this by making most of the reported mutations and testing their activity when expressed in AMPK knockout cells. Of 55 different mis-sense mutations (representing 75 cases), 9 (12%) appeared to cause a total loss of activity, 18 (24%) a partial loss, 11 (15%) an increase in phenformin-stimulated kinase activity, while just 37 (49%) had no clear effect on kinase activity. This supports the idea that AMPK-α2 acts as a tumour suppressor in the context of human skin cancer.
Collapse
Affiliation(s)
- Fiona A. Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - Simon A. Hawley
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - Fiona M. Russell
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - Nicola Goodman
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - D. Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| |
Collapse
|
31
|
Lei P, Ju Y, Peng F, Luo J. Applications and advancements of CRISPR-Cas in the treatment of lung cancer. Front Cell Dev Biol 2023; 11:1295084. [PMID: 38188023 PMCID: PMC10768725 DOI: 10.3389/fcell.2023.1295084] [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: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 01/09/2024] Open
Abstract
Lung cancer is one of the most malignant diseases and a major contributor to cancer-related deaths worldwide due to the deficiency of early diagnosis and effective therapy that are of great importance for patient prognosis and quality of life. Over the past decade, the advent of clustered regularly interspaced short palindromic repeats/CRISPR associated protein (CRISPR/Cas) system has significantly propelled the progress of both fundamental research and clinical trials of lung cancer. In this review, we review the current applications of the CRISPR/Cas system in diagnosis, target identification, and treatment resistance of lung cancer. Furthermore, we summarize the development of lung cancer animal models and delivery methods based on CRISPR system, providing novel insights into clinical diagnosis and treatment strategies of lung cancer.
Collapse
Affiliation(s)
- Pan Lei
- Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Yixin Ju
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Fenfen Peng
- Department of Pharmacy, Jianyang City Hospital of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Jianyang, Sichuan, China
| | - Jie Luo
- Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
32
|
Terry AR, Nogueira V, Rho H, Ramakrishnan G, Li J, Kang S, Pathmasiri KC, Bhat SA, Jiang L, Kuchay S, Cologna SM, Hay N. CD36 maintains lipid homeostasis via selective uptake of monounsaturated fatty acids during matrix detachment and tumor progression. Cell Metab 2023; 35:2060-2076.e9. [PMID: 37852255 PMCID: PMC11748917 DOI: 10.1016/j.cmet.2023.09.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 04/11/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
A high-fat diet (HFD) promotes metastasis through increased uptake of saturated fatty acids (SFAs). The fatty acid transporter CD36 has been implicated in this process, but a detailed understanding of CD36 function is lacking. During matrix detachment, endoplasmic reticulum (ER) stress reduces SCD1 protein, resulting in increased lipid saturation. Subsequently, CD36 is induced in a p38- and AMPK-dependent manner to promote preferential uptake of monounsaturated fatty acids (MUFAs), thereby maintaining a balance between SFAs and MUFAs. In attached cells, CD36 palmitoylation is required for MUFA uptake and protection from palmitate-induced lipotoxicity. In breast cancer mouse models, CD36-deficiency induced ER stress while diminishing the pro-metastatic effect of HFD, and only a palmitoylation-proficient CD36 rescued this effect. Finally, AMPK-deficient tumors have reduced CD36 expression and are metastatically impaired, but ectopic CD36 expression restores their metastatic potential. Our results suggest that, rather than facilitating HFD-driven tumorigenesis, CD36 plays a supportive role by preventing SFA-induced lipotoxicity.
Collapse
Affiliation(s)
- Alexander R Terry
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Veronique Nogueira
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Hyunsoo Rho
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Gopalakrishnan Ramakrishnan
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jing Li
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Soeun Kang
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Koralege C Pathmasiri
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sameer Ahmed Bhat
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Liping Jiang
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Shafi Kuchay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Stephanie M Cologna
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA; Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
33
|
Feng S, Wei F, Shi H, Chen S, Wang B, Huang D, Luo L. Roles of salt‑inducible kinases in cancer (Review). Int J Oncol 2023; 63:118. [PMID: 37654200 PMCID: PMC10546379 DOI: 10.3892/ijo.2023.5566] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
Salt inducible kinases (SIKs) with three subtypes SIK1, SIK2 and SIK3, belong to the AMP‑activated protein kinase family. They are expressed ubiquitously in humans. Under normal circumstances, SIK1 regulates adrenocortical function in response to high salt or adrenocorticotropic hormone stimulation, SIK2 is involved in cell metabolism, controlling insulin signaling and gluconeogenesis and SIK3 coordinates with the mTOR complex, promoting cancer. The dysregulation of SIKs has been widely detected in various types of cancers. Based on most of the existing studies, SIK1 is mostly considered a tumor inhibitor, SIK2 and SIK3 are usually associated with tumor promotion. However, the functions of SIKs have shown contradictory in certain tumors, suggesting that SIKs cannot be simply classified as oncogenes or tumor suppressor genes. The present review provided a comprehensive summary of the roles of SIKs in the initiation and progression of different cancers, aiming to elucidate their clinical value and discuss potential strategies for targeting SIKs in cancer therapy.
Collapse
Affiliation(s)
- Shenghui Feng
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fangyi Wei
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haoran Shi
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shen Chen
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bangqi Wang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Deqiang Huang
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lingyu Luo
- Department of Gastroenterology, Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
34
|
Ferrarone JR, Thomas J, Unni AM, Zheng Y, Nagiec MJ, Gardner EE, Mashadova O, Li K, Koundouros N, Montalbano A, Mustafa M, Cantley LC, Blenis J, Sanjana NE, Varmus H. LKB1 suppresses growth and promotes the internalization of EGFR through the PIKFYVE lipid kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563158. [PMID: 37904985 PMCID: PMC10614957 DOI: 10.1101/2023.10.19.563158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1- mediated growth suppression we developed a spheroid-based cell culture assay to study LKB1- dependent growth. Using this assay, along with genome-wide CRISPR screens and validation with orthogonal methods, we discovered that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase, which promotes the internalization of wild-type EGFR. Our findings reveal a new mechanism of regulation of EGFR, which may have implications for the treatment of LKB1 -mutant LUAD.
Collapse
|
35
|
Li Y, Li C, Liu Y, Yu J, Yang J, Cui Y, Wang TV, Li C, Jiang L, Song M, Rao Y. Sleep need, the key regulator of sleep homeostasis, is indicated and controlled by phosphorylation of threonine 221 in salt-inducible kinase 3. Genetics 2023; 225:iyad136. [PMID: 37477881 DOI: 10.1093/genetics/iyad136] [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: 06/11/2023] [Revised: 06/11/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
Sleep need drives sleep and plays a key role in homeostatic regulation of sleep. So far sleep need can only be inferred by animal behaviors and indicated by electroencephalography (EEG). Here we report that phosphorylation of threonine (T) 221 of the salt-inducible kinase 3 (SIK3) increased the catalytic activity and stability of SIK3. T221 phosphorylation in the mouse brain indicates sleep need: more sleep resulting in less phosphorylation and less sleep more phosphorylation during daily sleep/wake cycle and after sleep deprivation (SD). Sleep need was reduced in SIK3 loss of function (LOF) mutants and by T221 mutation to alanine (T221A). Rebound after SD was also decreased in SIK3 LOF and T221A mutant mice. By contrast, SIK1 and SIK2 do not satisfy criteria to be both an indicator and a controller of sleep need. Our results reveal SIK3-T221 phosphorylation as a chemical modification which indicates and controls sleep need.
Collapse
Affiliation(s)
- Yang Li
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Chengang Li
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Jianjun Yu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jingqun Yang
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Yunfeng Cui
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chaoyi Li
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Lifen Jiang
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Meilin Song
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Institute of Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China
- Capital Medical University, Beijing 10069, China
- Chinese Institute for Brain Research, Changping Laboratory, Yard 28, Science Park Road, ZGC Life Science Park, Changping District, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
36
|
Shi F, de Fatima Silva F, Liu D, Patel HU, Xu J, Zhang W, Türk C, Krüger M, Collins S. Salt-inducible kinase inhibition promotes the adipocyte thermogenic program and adipose tissue browning. Mol Metab 2023; 74:101753. [PMID: 37321371 PMCID: PMC10319839 DOI: 10.1016/j.molmet.2023.101753] [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: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
OBJECTIVE Norepinephrine stimulates the adipose tissue thermogenic program through a β-adrenergic receptor (βAR)-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling cascade. We discovered that a noncanonical activation of the mechanistic target of rapamycin complex 1 (mTORC1) by PKA is required for the βAR-stimulation of adipose tissue browning. However, the downstream events triggered by PKA-phosphorylated mTORC1 activation that drive this thermogenic response are not well understood. METHODS We used a proteomic approach of Stable Isotope Labeling by/with Amino acids in Cell culture (SILAC) to characterize the global protein phosphorylation profile in brown adipocytes treated with the βAR agonist. We identified salt-inducible kinase 3 (SIK3) as a candidate mTORC1 substrate and further tested the effect of SIK3 deficiency or SIK inhibition on the thermogenic gene expression program in brown adipocytes and in mouse adipose tissue. RESULTS SIK3 interacts with RAPTOR, the defining component of the mTORC1 complex, and is phosphorylated at Ser884 in a rapamycin-sensitive manner. Pharmacological SIK inhibition by a pan-SIK inhibitor (HG-9-91-01) in brown adipocytes increases basal Ucp1 gene expression and restores its expression upon blockade of either mTORC1 or PKA. Short-hairpin RNA (shRNA) knockdown of Sik3 augments, while overexpression of SIK3 suppresses, Ucp1 gene expression in brown adipocytes. The regulatory PKA phosphorylation domain of SIK3 is essential for its inhibition. CRISPR-mediated Sik3 deletion in brown adipocytes increases type IIa histone deacetylase (HDAC) activity and enhances the expression of genes involved in thermogenesis such as Ucp1, Pgc1α, and mitochondrial OXPHOS complex protein. We further show that HDAC4 interacts with PGC1α after βAR stimulation and reduces lysine acetylation in PGC1α. Finally, a SIK inhibitor well-tolerated in vivo (YKL-05-099) can stimulate the expression of thermogenesis-related genes and browning of mouse subcutaneous adipose tissue. CONCLUSIONS Taken together, our data reveal that SIK3, with the possible contribution of other SIKs, functions as a phosphorylation switch for β-adrenergic activation to drive the adipose tissue thermogenic program and indicates that more work to understand the role of the SIKs is warranted. Our findings also suggest that maneuvers targeting SIKs could be beneficial for obesity and related cardiometabolic disease.
Collapse
Affiliation(s)
- Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Flaviane de Fatima Silva
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Dianxin Liu
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hari U Patel
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan Xu
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Wei Zhang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Clara Türk
- CECAD Research Center, Institute for Genetics, University of Cologne, Cologne 50931, Germany
| | - Marcus Krüger
- CECAD Research Center, Institute for Genetics, University of Cologne, Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
37
|
Rashidfarrokhi A, Pillai R, Hao Y, Wu WL, Karadal-Ferrena B, Dimitriadoy SG, Cross M, Yeaton AH, Huang SM, Bhutkar AJ, Herrera A, Rajalingam S, Hayashi M, Huang KL, Bartnicki E, Zavitsanou AM, Wohlhieter CA, Leboeuf SE, Chen T, Loomis C, Mezzano V, Kulicke R, Davis FP, Stransky N, Smolen GA, Rudin CM, Moreira AL, Khanna KM, Pass HI, Wong KK, Koide S, Tsirigos A, Koralov SB, Papagiannakopoulos T. Tumor-intrinsic LKB1-LIF signaling axis establishes a myeloid niche to promote immune evasion and tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549147. [PMID: 37502974 PMCID: PMC10370066 DOI: 10.1101/2023.07.15.549147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Tumor mutations can influence the surrounding microenvironment leading to suppression of anti-tumor immune responses and thereby contributing to tumor progression and failure of cancer therapies. Here we use genetically engineered lung cancer mouse models and patient samples to dissect how LKB1 mutations accelerate tumor growth by reshaping the immune microenvironment. Comprehensive immune profiling of LKB1 -mutant vs wildtype tumors revealed dramatic changes in myeloid cells, specifically enrichment of Arg1 + interstitial macrophages and SiglecF Hi neutrophils. We discovered a novel mechanism whereby autocrine LIF signaling in Lkb1 -mutant tumors drives tumorigenesis by reprogramming myeloid cells in the immune microenvironment. Inhibiting LIF signaling in Lkb1 -mutant tumors, via gene targeting or with a neutralizing antibody, resulted in a striking reduction in Arg1 + interstitial macrophages and SiglecF Hi neutrophils, expansion of antigen specific T cells, and inhibition of tumor progression. Thus, targeting LIF signaling provides a new therapeutic approach to reverse the immunosuppressive microenvironment of LKB1 -mutant tumors.
Collapse
|
38
|
Qian Y, Galan-Cobo A, Guijarro I, Dang M, Molkentine D, Poteete A, Zhang F, Wang Q, Wang J, Parra E, Panda A, Fang J, Skoulidis F, Wistuba II, Verma S, Merghoub T, Wolchok JD, Wong KK, DeBerardinis RJ, Minna JD, Vokes NI, Meador CB, Gainor JF, Wang L, Reuben A, Heymach JV. MCT4-dependent lactate secretion suppresses antitumor immunity in LKB1-deficient lung adenocarcinoma. Cancer Cell 2023; 41:1363-1380.e7. [PMID: 37327788 PMCID: PMC11161201 DOI: 10.1016/j.ccell.2023.05.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/18/2023]
Abstract
Inactivating STK11/LKB1 mutations are genomic drivers of primary resistance to immunotherapy in KRAS-mutated lung adenocarcinoma (LUAD), although the underlying mechanisms remain unelucidated. We find that LKB1 loss results in enhanced lactate production and secretion via the MCT4 transporter. Single-cell RNA profiling of murine models indicates that LKB1-deficient tumors have increased M2 macrophage polarization and hypofunctional T cells, effects that could be recapitulated by the addition of exogenous lactate and abrogated by MCT4 knockdown or therapeutic blockade of the lactate receptor GPR81 expressed on immune cells. Furthermore, MCT4 knockout reverses the resistance to PD-1 blockade induced by LKB1 loss in syngeneic murine models. Finally, tumors from STK11/LKB1 mutant LUAD patients demonstrate a similar phenotype of enhanced M2-macrophages polarization and hypofunctional T cells. These data provide evidence that lactate suppresses antitumor immunity and therapeutic targeting of this pathway is a promising strategy to reversing immunotherapy resistance in STK11/LKB1 mutant LUAD.
Collapse
Affiliation(s)
- Yu Qian
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Ana Galan-Cobo
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Irene Guijarro
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Minghao Dang
- Department of Genomic Medicine, Houston, TX, USA
| | - David Molkentine
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Alissa Poteete
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Fahao Zhang
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, Houston, TX, USA
| | - Edwin Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jacy Fang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Svena Verma
- Ludwig Collaborative and Swim Across America Laboratory, MSK, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, MSK, New York, NY, USA
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, MSK, New York, NY, USA
| | - Kwok-Kin Wong
- Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Natalie I Vokes
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - Catherine B Meador
- Department of Medicine, Division of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA; Center for Thoracic Cancers, Massachusetts General Hospital, Boston, MA, USA
| | - Justin F Gainor
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Center for Thoracic Cancers, Massachusetts General Hospital, Boston, MA, USA
| | - Linghua Wang
- Department of Genomic Medicine, Houston, TX, USA
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, Houston, TX, USA.
| |
Collapse
|
39
|
Compton SE, Kitchen-Goosen SM, DeCamp LM, Lau KH, Mabvakure B, Vos M, Williams KS, Wong KK, Shi X, Rothbart SB, Krawczyk CM, Jones RG. LKB1 controls inflammatory potential through CRTC2-dependent histone acetylation. Mol Cell 2023:S1097-2765(23)00288-5. [PMID: 37172591 DOI: 10.1016/j.molcel.2023.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/17/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
Deregulated inflammation is a critical feature driving the progression of tumors harboring mutations in the liver kinase B1 (LKB1), yet the mechanisms linking LKB1 mutations to deregulated inflammation remain undefined. Here, we identify deregulated signaling by CREB-regulated transcription coactivator 2 (CRTC2) as an epigenetic driver of inflammatory potential downstream of LKB1 loss. We demonstrate that LKB1 mutations sensitize both transformed and non-transformed cells to diverse inflammatory stimuli, promoting heightened cytokine and chemokine production. LKB1 loss triggers elevated CRTC2-CREB signaling downstream of the salt-inducible kinases (SIKs), increasing inflammatory gene expression in LKB1-deficient cells. Mechanistically, CRTC2 cooperates with the histone acetyltransferases CBP/p300 to deposit histone acetylation marks associated with active transcription (i.e., H3K27ac) at inflammatory gene loci, promoting cytokine expression. Together, our data reveal a previously undefined anti-inflammatory program, regulated by LKB1 and reinforced through CRTC2-dependent histone modification signaling, that links metabolic and epigenetic states to cell-intrinsic inflammatory potential.
Collapse
Affiliation(s)
- Shelby E Compton
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Susan M Kitchen-Goosen
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kin H Lau
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Batsirai Mabvakure
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Matthew Vos
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kelsey S Williams
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Scott B Rothbart
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Connie M Krawczyk
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA; Metabolism and Nutrition (MeNu) Program, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
40
|
Iyengar P, Gandhi AY, Granados J, Guo T, Gupta A, Yu J, Llano EM, Zhang F, Gao A, Kandathil A, Williams D, Gao B, Girard L, Malladi VS, Shelton JM, Evers BM, Hannan R, Ahn C, Minna JD, Infante RE. Tumor loss-of-function mutations in STK11/LKB1 induce cachexia. JCI Insight 2023; 8:e165419. [PMID: 37092555 PMCID: PMC10243820 DOI: 10.1172/jci.insight.165419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 02/23/2023] [Indexed: 04/25/2023] Open
Abstract
Cancer cachexia (CC), a wasting syndrome of muscle and adipose tissue resulting in weight loss, is observed in 50% of patients with solid tumors. Management of CC is limited by the absence of biomarkers and knowledge of molecules that drive its phenotype. To identify such molecules, we injected 54 human non-small cell lung cancer (NSCLC) lines into immunodeficient mice, 17 of which produced an unambiguous phenotype of cachexia or non-cachexia. Whole-exome sequencing revealed that 8 of 10 cachexia lines, but none of the non-cachexia lines, possessed mutations in serine/threonine kinase 11 (STK11/LKB1), a regulator of nutrient sensor AMPK. Silencing of STK11/LKB1 in human NSCLC and murine colorectal carcinoma lines conferred a cachexia phenotype after cell transplantation into immunodeficient (human NSCLC) and immunocompetent (murine colorectal carcinoma) models. This host wasting was associated with an alteration in the immune cell repertoire of the tumor microenvironments that led to increases in local mRNA expression and serum levels of CC-associated cytokines. Mutational analysis of circulating tumor DNA from patients with NSCLC identified 89% concordance between STK11/LKB1 mutations and weight loss at cancer diagnosis. The current data provide evidence that tumor STK11/LKB1 loss of function is a driver of CC, simultaneously serving as a genetic biomarker for this wasting syndrome.
Collapse
Affiliation(s)
- Puneeth Iyengar
- Center for Human Nutrition
- Department of Radiation Oncology
- Harold C. Simmons Comprehensive Cancer Center
| | - Aakash Y. Gandhi
- Center for Human Nutrition
- Harold C. Simmons Comprehensive Cancer Center
| | | | | | - Arun Gupta
- Center for Human Nutrition
- Department of Radiation Oncology
| | - Jinhai Yu
- Center for Human Nutrition
- Harold C. Simmons Comprehensive Cancer Center
- Department of Internal Medicine
| | | | - Faya Zhang
- Department of Radiation Oncology
- Harold C. Simmons Comprehensive Cancer Center
| | - Ang Gao
- Harold C. Simmons Comprehensive Cancer Center
- Department of Population and Data Sciences
| | | | | | - Boning Gao
- Harold C. Simmons Comprehensive Cancer Center
- Department of Pharmacology
- Hamon Center for Therapeutic Oncology Research
| | - Luc Girard
- Harold C. Simmons Comprehensive Cancer Center
- Department of Pharmacology
- Hamon Center for Therapeutic Oncology Research
| | | | | | | | - Raquibul Hannan
- Department of Radiation Oncology
- Harold C. Simmons Comprehensive Cancer Center
| | - Chul Ahn
- Harold C. Simmons Comprehensive Cancer Center
- Department of Population and Data Sciences
| | - John D. Minna
- Harold C. Simmons Comprehensive Cancer Center
- Department of Internal Medicine
- Department of Pharmacology
- Hamon Center for Therapeutic Oncology Research
| | - Rodney E. Infante
- Center for Human Nutrition
- Harold C. Simmons Comprehensive Cancer Center
- Department of Internal Medicine
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Deng J, Peng DH, Fenyo D, Yuan H, Lopez A, Levin DS, Meynardie M, Quinteros M, Ranieri M, Sahu S, Lau SCM, Shum E, Velcheti V, Punekar SR, Rekhtman N, Dowling CM, Weerasekara V, Xue Y, Ji H, Siu Y, Jones D, Hata AN, Shimamura T, Poirier JT, Rudin CM, Hattori T, Koide S, Papagiannakopoulos T, Neel BG, Bardeesy N, Wong KK. In vivo metabolomics identifies CD38 as an emergent vulnerability in LKB1 -mutant lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537350. [PMID: 37131623 PMCID: PMC10153147 DOI: 10.1101/2023.04.18.537350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
LKB1/STK11 is a serine/threonine kinase that plays a major role in controlling cell metabolism, resulting in potential therapeutic vulnerabilities in LKB1-mutant cancers. Here, we identify the NAD + degrading ectoenzyme, CD38, as a new target in LKB1-mutant NSCLC. Metabolic profiling of genetically engineered mouse models (GEMMs) revealed that LKB1 mutant lung cancers have a striking increase in ADP-ribose, a breakdown product of the critical redox co-factor, NAD + . Surprisingly, compared with other genetic subsets, murine and human LKB1-mutant NSCLC show marked overexpression of the NAD+-catabolizing ectoenzyme, CD38 on the surface of tumor cells. Loss of LKB1 or inactivation of Salt-Inducible Kinases (SIKs)-key downstream effectors of LKB1- induces CD38 transcription induction via a CREB binding site in the CD38 promoter. Treatment with the FDA-approved anti-CD38 antibody, daratumumab, inhibited growth of LKB1-mutant NSCLC xenografts. Together, these results reveal CD38 as a promising therapeutic target in patients with LKB1 mutant lung cancer. SIGNIFICANCE Loss-of-function mutations in the LKB1 tumor suppressor of lung adenocarcinoma patients and are associated with resistance to current treatments. Our study identified CD38 as a potential therapeutic target that is highly overexpressed in this specific subtype of cancer, associated with a shift in NAD homeostasis.
Collapse
|
42
|
Nguyen K, Hebert K, McConnell E, Cullen N, Cheng T, Awoyode S, Martin E, Chen W, Wu T, Alahari SK, Izadpanah R, Collins-Burow BM, Lee SB, Drewry DH, Burow ME. LKB1 Signaling and Patient Survival Outcomes in Hepatocellular Carcinoma. Pharmacol Res 2023; 192:106757. [PMID: 37023992 DOI: 10.1016/j.phrs.2023.106757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023]
Abstract
The liver is a major organ that is involved in essential biological functions such as digestion, nutrient storage, and detoxification. Furthermore, it is one of the most metabolically active organs with active roles in regulating carbohydrate, protein, and lipid metabolism. Hepatocellular carcinoma is a cancer of the liver that is associated in settings of chronic inflammation such as viral hepatitis, repeated toxin exposure, and fatty liver disease. Furthermore, liver cancer is the most common cause of death associated with cirrhosis and is the 3rd leading cause of global cancer deaths. LKB1 signaling has been demonstrated to play a role in regulating cellular metabolism under normal and nutrient deficient conditions. Furthermore, LKB1 signaling has been found to be involved in many cancers with most reports identifying LKB1 to have a tumor suppressive role. In this review, we use the KMPlotter database to correlate RNA levels of LKB1 signaling genes and hepatocellular carcinoma patient survival outcomes with the hopes of identifying potential biomarkers clinical usage. Based on our results STRADß, CAB39L, AMPKα, MARK2, SIK1, SIK2, BRSK1, BRSK2, and SNRK expression has a statistically significant impact on patient survival.
Collapse
Affiliation(s)
- Khoa Nguyen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Katherine Hebert
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Emily McConnell
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicole Cullen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Thomas Cheng
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Susanna Awoyode
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth Martin
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA, USA
| | - Reza Izadpanah
- Applied Stem Cell Laboratory, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Sean B Lee
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - David H Drewry
- UNC Eshelman School of Pharmacy and UNC Lineberger Comprehensive Cancer Center, Chemical Biology and Medicinal Chemistry Division, SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew E Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
43
|
Stein BD, Ferrarone JR, Gardner EE, Chang JW, Wu D, Hollstein PE, Liang RJ, Yuan M, Chen Q, Coukos JS, Sindelar M, Ngo B, Gross SS, Shaw RJ, Zhang C, Asara JM, Moellering RE, Varmus H, Cantley LC. LKB1-Dependent Regulation of TPI1 Creates a Divergent Metabolic Liability between Human and Mouse Lung Adenocarcinoma. Cancer Discov 2023; 13:1002-1025. [PMID: 36715544 PMCID: PMC10068449 DOI: 10.1158/2159-8290.cd-22-0805] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/14/2022] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
KRAS is the most frequently mutated oncogene in human lung adenocarcinomas (hLUAD), and activating mutations frequently co-occur with loss-of-function mutations in TP53 or STK11/LKB1. However, mutation of all three genes is rarely observed in hLUAD, even though engineered comutation is highly aggressive in mouse lung adenocarcinoma (mLUAD). Here, we provide a mechanistic explanation for this difference by uncovering an evolutionary divergence in the regulation of triosephosphate isomerase (TPI1). In hLUAD, TPI1 activity is regulated via phosphorylation at Ser21 by the salt inducible kinases (SIK) in an LKB1-dependent manner, modulating flux between the completion of glycolysis and production of glycerol lipids. In mice, Ser21 of TPI1 is a Cys residue that can be oxidized to alter TPI1 activity without a need for SIKs or LKB1. Our findings suggest this metabolic flexibility is critical in rapidly growing cells with KRAS and TP53 mutations, explaining why the loss of LKB1 creates a liability in these tumors. SIGNIFICANCE Utilizing phosphoproteomics and metabolomics in genetically engineered human cell lines and genetically engineered mouse models (GEMM), we uncover an evolutionary divergence in metabolic regulation within a clinically relevant genotype of human LUAD with therapeutic implications. Our data provide a cautionary example of the limits of GEMMs as tools to study human diseases such as cancers. This article is highlighted in the In This Issue feature, p. 799.
Collapse
Affiliation(s)
- Benjamin D. Stein
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - John R. Ferrarone
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Eric E. Gardner
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Jae Won Chang
- Department of Chemistry, University of Chicago, Chicago, Illinois
| | - David Wu
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Pablo E. Hollstein
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Roger J. Liang
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Min Yuan
- Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - John S. Coukos
- Department of Chemistry, University of Chicago, Chicago, Illinois
| | - Miriam Sindelar
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Bryan Ngo
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, New York
| | - Reuben J. Shaw
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California
| | - Chen Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - John M. Asara
- Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | | | - Harold Varmus
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Lewis C. Cantley
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, New York
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Eichner LJ, Curtis SD, Brun SN, McGuire CK, Gushterova I, Baumgart JT, Trefts E, Ross DS, Rymoff TJ, Shaw RJ. HDAC3 is critical in tumor development and therapeutic resistance in Kras-mutant non-small cell lung cancer. SCIENCE ADVANCES 2023; 9:eadd3243. [PMID: 36930718 PMCID: PMC10022903 DOI: 10.1126/sciadv.add3243] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
HDAC3 is one of the main targets of histone deacetylase (HDAC) inhibitors in clinical development as cancer therapies, yet the in vivo role of HDAC3 in solid tumors is unknown. We identified a critical role for HDAC3 in Kras-mutant lung cancer. Using genetically engineered mouse models (GEMMs), we found that HDAC3 is required for lung tumor growth in vivo. HDAC3 was found to direct and enhance the transcription effects of the lung cancer lineage transcription factor NKX2-1 to mediate expression of a common set of target genes. We identified FGFR1 as a critical previously unidentified target of HDAC3. Leveraging this, we identified that an HDAC3-dependent transcriptional cassette becomes hyperactivated as Kras/LKB1-mutant cells develop resistance to the MEK inhibitor trametinib, and this can be reversed by treatment with the HDAC1/HDAC3 inhibitor entinostat. We found that the combination of entinostat plus trametinib treatment elicits therapeutic benefit in the Kras/LKB1 GEMM.
Collapse
Affiliation(s)
- Lillian J. Eichner
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, 303 E. Superior Street, Chicago, IL USA
| | - Stephanie D. Curtis
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| | - Sonja N. Brun
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| | - Caroline K. McGuire
- Department of Biochemistry and Molecular Genetics, Northwestern University, 303 E. Superior Street, Chicago, IL USA
| | - Irena Gushterova
- Department of Biochemistry and Molecular Genetics, Northwestern University, 303 E. Superior Street, Chicago, IL USA
| | - Joshua T. Baumgart
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| | - Elijah Trefts
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| | - Debbie S. Ross
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| | - Tammy J. Rymoff
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| | - Reuben J. Shaw
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA USA
| |
Collapse
|
45
|
CAI X, CAO Z, PAN J, ZHENG H. Transcription factor NFIC activates STK11 transcription to repress the proliferation, migration, and invasion of lung adenocarcinoma cells. MINERVA BIOTECHNOLOGY AND BIOMOLECULAR RESEARCH 2023. [DOI: 10.23736/s2724-542x.23.02918-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
46
|
Tzavlaki K, Ohata Y, Morén A, Watanabe Y, Eriksson J, Tsuchiya M, Kubo Y, Yamamoto K, Sellin ME, Kato M, Caja L, Heldin CH, Moustakas A. The liver kinase B1 supports mammary epithelial morphogenesis by inhibiting critical factors that mediate epithelial-mesenchymal transition. J Cell Physiol 2023; 238:790-812. [PMID: 36791282 DOI: 10.1002/jcp.30975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
The liver kinase B1 (LKB1) controls cellular metabolism and cell polarity across species. We previously established a mechanism for negative regulation of transforming growth factor β (TGFβ) signaling by LKB1. The impact of this mechanism in the context of epithelial polarity and morphogenesis remains unknown. After demonstrating that human mammary tissue expresses robust LKB1 protein levels, whereas invasive breast cancer exhibits significantly reduced LKB1 levels, we focused on mammary morphogenesis studies in three dimensional (3D) acinar organoids. CRISPR/Cas9-introduced loss-of-function mutations of STK11 (LKB1) led to profound defects in the formation of 3D organoids, resulting in amorphous outgrowth and loss of rotation of young organoids embedded in matrigel. This defect was associated with an enhanced signaling by TGFβ, including TGFβ auto-induction and induction of transcription factors that mediate epithelial-mesenchymal transition (EMT). Protein marker analysis confirmed a more efficient EMT response to TGFβ signaling in LKB1 knockout cells. Accordingly, chemical inhibition of the TGFβ type I receptor kinase largely restored the morphogenetic defect of LKB1 knockout cells. Similarly, chemical inhibition of the bone morphogenetic protein pathway or the TANK-binding kinase 1, or genetic silencing of the EMT factor SNAI1, partially restored the LKB1 knockout defect. Thus, LKB1 sustains mammary epithelial morphogenesis by limiting pathways that promote EMT. The observed downregulation of LKB1 expression in breast cancer is therefore predicted to associate with enhanced EMT induced by SNAI1 and TGFβ family members.
Collapse
Affiliation(s)
- Kalliopi Tzavlaki
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Yae Ohata
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Anita Morén
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Yukihide Watanabe
- Department of Experimental Pathology and Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Jens Eriksson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Maiko Tsuchiya
- Department of Oral Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Pathology, Teikyo University School of Medicine, Tokyo, Japan
| | - Yuki Kubo
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mikael E Sellin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Mitsuyasu Kato
- Department of Experimental Pathology and Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Carl-Henrik Heldin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
47
|
Cuttini E, Goi C, Pellarin E, Vida R, Brancolini C. HDAC4 in cancer: A multitasking platform to drive not only epigenetic modifications. Front Mol Biosci 2023; 10:1116660. [PMID: 36762207 PMCID: PMC9902726 DOI: 10.3389/fmolb.2023.1116660] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Controlling access to genomic information and maintaining its stability are key aspects of cell life. Histone acetylation is a reversible epigenetic modification that allows access to DNA and the assembly of protein complexes that regulate mainly transcription but also other activities. Enzymes known as histone deacetylases (HDACs) are involved in the removal of the acetyl-group or in some cases of small hydrophobic moieties from histones but also from the non-histone substrate. The main achievement of HDACs on histones is to repress transcription and promote the formation of more compact chromatin. There are 18 different HDACs encoded in the human genome. Here we will discuss HDAC4, a member of the class IIa family, and its possible contribution to cancer development.
Collapse
Affiliation(s)
- Emma Cuttini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Camilla Goi
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Ester Pellarin
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Riccardo Vida
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Claudio Brancolini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy,Laboratory of Epigenomics, Department of Medicine, Università degli Studi di Udine, Udine, Italy,*Correspondence: Claudio Brancolini,
| |
Collapse
|
48
|
Pan B, Zhu X, Han B, Weng J, Wang Y, Liu Y. The SIK1/CRTC2/CREB1 and TWIST1/PI3K/Akt/GSK3β signaling pathways mediated by microRNA-25-3p are altered in the schizophrenic rat brain. Front Cell Neurosci 2023; 17:1087335. [PMID: 36744005 PMCID: PMC9896578 DOI: 10.3389/fncel.2023.1087335] [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: 11/02/2022] [Accepted: 01/09/2023] [Indexed: 01/21/2023] Open
Abstract
Schizophrenia is a group of severe mental disorders. MiR-25-3p was shown to be involved in various neuropsychiatric diseases and can regulate SIK1 and TWIST1. The CRTC2/CREB1 and PI3K/Akt/GSK3β signaling pathways are downstream pathways of SIK1 and TWIST1, respectively. This study investigated whether miR-25-3p-mediated SIK1/CRTC2/CREB1 and TWIST1/PI3K/Akt/GSK3β signaling pathways are present in an animal model relevant to schizophrenia. A schizophrenic rat model was established by using sub-chronic MK-801 administration. An RNA-seq test was performed to examine the differentially expressed genes (DEGs) in the rat prefrontal cortex (PFC). The mRNA levels of miR-25-3p, SIK1, and TWIST in the PFC and caudate putamen (CPu) were assessed by qRT-PCR. Phosphorylation of the SIK1/CRTC2/CREB1 and TWIST1/PI3K/Akt/GSK3β pathways in the two brain regions was examined by Western blots. The RNA-seq data revealed down-regulated miR-25-3p expression and up-regulated SIK1 and TWIST1 mRNA expression induced by MK-801. Additionally, SIK1 and TWIST1 were shown to be possible downstream responders of miR-25-3p in previous studies. qRT-PCR confirmed the changes of miR-25-3p, SIK1, and TWIST1 induced by MK-801 in both brain regions, which, however, was reversed by risperidone. Furthermore, the phosphorylation of the SIK1/CRTC2/CREB1 pathway was repressed by MK-801, whereas the phosphorylation of the TWIST1/PI3K/Akt/GSK3β pathway was increased by MK-801 in either of the two brain regions. Moreover, the altered phosphorylation of these two signaling pathways induced by MK-801 can be restored by risperidone. In conclusion, this study suggests that altered SIK1/CRTC2/CREB1 and TWIST1/PI3K/Akt/GSK3β signaling pathways mediated by miR-25-3p is very likely to be associated with schizophrenia, revealing potential targets for the treatment and clinical diagnosis of schizophrenia.
Collapse
Affiliation(s)
- Bo Pan
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University Medical College, Yangzhou, China,Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China,Bo Pan,
| | - Xiaoli Zhu
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University Medical College, Yangzhou, China,Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
| | - Bing Han
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University Medical College, Yangzhou, China,Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
| | - Jianjun Weng
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University Medical College, Yangzhou, China,Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
| | - Yuting Wang
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University Medical College, Yangzhou, China,Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
| | - Yanqing Liu
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou University Medical College, Yangzhou, China,Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China,*Correspondence: Yanqing Liu,
| |
Collapse
|
49
|
Baumgartner C, Yadav AK, Chefetz I. AMPK-like proteins and their function in female reproduction and gynecologic cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 134:245-270. [PMID: 36858738 DOI: 10.1016/bs.apcsb.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Serine-threonine kinase (STK11), also known as liver kinase B1 (LKB1), is a regulator of cellular homeostasis through regulating the cellular ATP-to-ADP ratio. LKB1 is classified as a tumor suppressor and functions as the key activator of AMP-activated protein kinase (AMPK) and a family of serine-threonine kinases called AMPK-like proteins. These proteins include novel (nua) kinase family 1 (NUAK1 and 2), salt inducible kinase (SIK1), QIK (known as SIK2), QSK (known as SIK3 kinase), and maternal embryonic leuzine zipper kinase (MELK) on tightly controlled and specific residual sites. LKB1 also regulates brain selective kinases 1 and 2 (BRSK1 and 2), additional members of AMPK-like protein family, which functions are probably less studied. AMPK-like proteins play a role in variety of reproductive physiology functions such as follicular maturation, menopause, embryogenesis, oocyte maturation, and preimplantation development. In addition, dysfunctional activity of AMPK-like proteins contributes to apoptosis blockade in cancer cells and induction of the epithelial-mesenchymal transition required for metastasis. Dysregulation of these proteins occurs in ovarian, endometrial, and cervical cancers. AMPK-like proteins are still undergoing further classification and may represent novel targets for targeted gynecologic cancer therapies. In this chapter, we describe the AMPK-like family of proteins and their roles in reproductive physiology and gynecologic cancers.
Collapse
Affiliation(s)
| | - Anil Kumar Yadav
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Ilana Chefetz
- The Hormel Institute, University of Minnesota, Austin, MN, United States; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States; Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States; Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
50
|
Avilés-Salas A, Díaz-García DA, Lara-Mejía L, Cardona AF, Orozco-Morales M, Catalán R, Hernández-Pedro NY, Rios-Garcia E, Ramos-Ramírez M, Arrieta O. LKB1 Loss Assessed by Immunohistochemistry as a Prognostic Marker to First-Line Therapy in Advanced Non-Small-Cell Lung Cancer. Curr Oncol 2022; 30:333-343. [PMID: 36661676 PMCID: PMC9857995 DOI: 10.3390/curroncol30010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
(1) Background: Liver kinase B1 (LKB1) is a tumor suppressor gene involved in cell growth and metabolism. However, its alterations are not routinely assessed for guiding therapy in clinical practice. We assessed LKB1 expression by immunohistochemistry as a potential biomarker. (2) Methods: This bicentric retrospective cohort study analyzed data from patients with advanced NSCLC who initiated platinum-based chemotherapy or epidermal growth factor receptor- tyrosine kinase inhibitor (EGFR-TKI) between January 2016 and December 2020. Kaplan-Meier and Cox regression models were used for survival curves and multivariate analysis. (3) Results: 110 patients were evaluated, and the clinical stage IV predominated the lung adenocarcinoma histology. LKB1 loss was observed in 66.3% of cases. LKB1 loss was associated with non-smokers, the absence of wood smoke exposure and an EGFR wild-type status. The median progression-free survival (PFS) and overall survival (OS) in the population were 11.1 and 26.8 months, respectively, in the loss group, compared with cases exhibiting a positive expression. After an adjustment by age, smoking status, Eastern Cooperative Oncology Group Performance Score (ECOG-PS), EGFR status and type of administered therapy, LKB1 loss was significantly associated with worse PFS and OS. (4) Conclusion: Patients with an LKB1 loss had worse clinical outcomes. This study warrants prospective assessments to confirm the prognostic role of the LKB1 expression in advanced NSCLC.
Collapse
Affiliation(s)
- Alejandro Avilés-Salas
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Diego A. Díaz-García
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Luis Lara-Mejía
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Andrés F. Cardona
- Direction of Research and Education, Luis Carlos Sarmeinto Angulo Cancer Treatment and Research Center—CTIC, Bogotá 110131, Colombia
| | - Mario Orozco-Morales
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Neuroimmunology Laboratory, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico
| | - Rodrigo Catalán
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Norma Y. Hernández-Pedro
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Eduardo Rios-Garcia
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Maritza Ramos-Ramírez
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City 14080, Mexico
| |
Collapse
|