1
|
Fonseca-Pereira D, Bae S, Clay SL, Michaud M, MacDonald MH, Glickman JN, Garrett WS. The metabolic sensor LKB1 regulates ILC3 homeostasis and mitochondrial function. Cell Rep 2025; 44:115456. [PMID: 40120107 DOI: 10.1016/j.celrep.2025.115456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are tissue-resident cells that sense environmental cues, control infections, and promote tissue homeostasis at mucosal surfaces. The metabolic sensor liver kinase B1 (LKB1) integrates intracellular stress, metabolism, and mitochondrial function to promote the development and effector functions of a variety of immune cells; however, the role of LKB1 in ILC3 function was unknown. Here, we show that LKB1 is crucial for adult ILC3 homeostasis, cytokine production, and mitochondrial function. ILC3-specific LKB1 deletion resulted in a reduced number of ILC3s and interleukin-22 (IL-22) production. LKB1-deficient ILC3s had decreased survival, mitochondrial dysfunction, cytoplasmic lipid accumulation, and altered bioenergetics. Using LKB1 downstream kinase modulators, we found that LKB1 regulation of ILC3 survival and IL-22 production requires signaling through microtubule affinity-regulating kinases (MARKs). Mechanistically, LKB1 deficiency resulted in increased reactive oxygen species (ROS) production and NFAT2 and PD-1 expression. Our work reveals that metabolic regulation of enteric ILC3 function by an LKB1-dependent signaling network is crucial for intestinal immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Diogo Fonseca-Pereira
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Sena Bae
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Slater L Clay
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Monia Michaud
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Meghan H MacDonald
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Jonathan N Glickman
- Gastrointestinal Pathology, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA; Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
2
|
Wang C, Ye Y, Zhao M, Chen Q, Liu B, Ren W. Asparagine transporter supports macrophage inflammation via histone phosphorylation. SCIENCE ADVANCES 2025; 11:eads3506. [PMID: 40203093 PMCID: PMC11980831 DOI: 10.1126/sciadv.ads3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Solute carrier (SLC) family is essential for immune responses; nevertheless, whether and how SLCs regulate macrophage inflammation remains unclear. Here, we demonstrate that K636 acetylation mediates high abundance of SLC6A14 in inflammatory macrophages. Notably, the pharmacological inhibition or genetic modulation of SLC6A14 reduces macrophage interleukin-1β (IL-1β) secretion dependently of lower asparagine uptake and subsequently enhanced nuclear LKB1. Mechanistically, nuclear LKB1 lessens MAPK pathway-mediated NLRP3 inflammasome activation by increased histone 3 S10/28 phosphorylation-dependent cyclin O transcription. Moreover, myeloid Slc6a14 deficiency alleviates pulmonary inflammation via suppressing inflammatory macrophage responses. Overall, these results uncover a network by which SLC6A14-mediated asparagine uptake orchestrates macrophage inflammation through histone phosphorylation, providing a crucial target for modulation of inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Muyang Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingyi Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Bingnan Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
3
|
Baskin A, Soudah N, Gilad N, Halevy N, Darlyuk-Saadon I, Schoffman H, Engelberg D. All intrinsically active Erk1/2 mutants autophosphorylate Threonine207/188, a plausible regulator of the TEY motif phosphorylation. J Biol Chem 2025:108509. [PMID: 40222547 DOI: 10.1016/j.jbc.2025.108509] [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: 10/23/2024] [Revised: 03/19/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025] Open
Abstract
The extracellular-activated kinases 1 & 2 (Erk1/2) are catalytically active when dually phosphorylated on a TEY motif located at the activation loop. In human patients with cardiac hypertrophy Erk1/2 are phosphorylated on yet another activation loop's residue, T207/188. Intrinsically active variants of Erk1/2, mutated at R84/65, are also (auto)phosphorylated on T207/188. It is not known whether T207/188 phosphorylation is restricted to these cases nor how it affects Erks' activity. We report that T207/188 phosphorylation is not rare, as we found that: 1) All known auto-activated Erk1/2 variants are phosphorylated on T207/188. 2) It occurs in various cell lines and mouse tissues. 3) It is extremely high in patients with skeletal muscle atrophies or myopathies. We propose that T207/188 controls the permissiveness of the TEY motif for phosphorylation because T207/188-mutated Erk1/2 and the yeast Erk/Mpk1 were efficiently dually phosphorylated when expressed in HEK293 or yeast cells, respectively. The T207/188-mutated Mpk1 was not TEY-phosphorylated in cells knocked out for MEKs, suggesting that its enhanced phosphorylation in wildtype cells is MEK-dependent. Thus, as T207/188-mutated Erk1/2 and Mpk1 recruit MEKs the role of T207/188 is to impede MEKs' ability to phosphorylate Erks. T207/188 also impedes autophosphorylation as recombinant Erk2 mutated at T188 is spontaneously autophosphorylated, although exclusively on Y185. The role of T207/188 in regulating activation loop phosphorylation may be common to most Ser/Thr kinases as 86% of them (in the human kinome) possess T207/188 orthologs, and 160 of them were already reported to be phosphorylated on this residue.
Collapse
Affiliation(s)
- Alexey Baskin
- Dept. of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Nadine Soudah
- Dept. of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Nechama Gilad
- Dept. of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel; Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore, 138602
| | - Neriah Halevy
- Dept. of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Ilona Darlyuk-Saadon
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore, 138602
| | - Hanan Schoffman
- Stein Family mass spectrometry unit, the Research infrastructure center, the Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - David Engelberg
- Dept. of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel; Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore, 138602; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456.
| |
Collapse
|
4
|
Bendzunas GN, Byrne DP, Shrestha S, Daly LA, Oswald SO, Katiyar S, Venkat A, Yeung W, Eyers CE, Eyers PA, Kannan N. Redox regulation and dynamic control of brain-selective kinases BRSK1/2 in the AMPK family through cysteine-based mechanisms. eLife 2025; 13:RP92536. [PMID: 40172959 PMCID: PMC11964447 DOI: 10.7554/elife.92536] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
In eukaryotes, protein kinase signaling is regulated by a diverse array of post-translational modifications, including phosphorylation of Ser/Thr residues and oxidation of cysteine (Cys) residues. While regulation by activation segment phosphorylation of Ser/Thr residues is well understood, relatively little is known about how oxidation of cysteine residues modulate catalysis. In this study, we investigate redox regulation of the AMPK-related brain-selective kinases (BRSK) 1 and 2, and detail how broad catalytic activity is directly regulated through reversible oxidation and reduction of evolutionarily conserved Cys residues within the catalytic domain. We show that redox-dependent control of BRSKs is a dynamic and multilayered process involving oxidative modifications of several Cys residues, including the formation of intramolecular disulfide bonds involving a pair of Cys residues near the catalytic HRD motif and a highly conserved T-loop Cys with a BRSK-specific Cys within an unusual CPE motif at the end of the activation segment. Consistently, mutation of the CPE-Cys increases catalytic activity in vitro and drives phosphorylation of the BRSK substrate Tau in cells. Molecular modeling and molecular dynamics simulations indicate that oxidation of the CPE-Cys destabilizes a conserved salt bridge network critical for allosteric activation. The occurrence of spatially proximal Cys amino acids in diverse Ser/Thr protein kinase families suggests that disulfide-mediated control of catalytic activity may be a prevalent mechanism for regulation within the broader AMPK family.
Collapse
Affiliation(s)
- George N Bendzunas
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
| | - Dominic P Byrne
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Safal Shrestha
- Institute of Bioinformatics, University of GeorgiaAthensUnited States
| | - Leonard A Daly
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Sally O Oswald
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Samiksha Katiyar
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
| | - Aarya Venkat
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
| | - Wayland Yeung
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
- Institute of Bioinformatics, University of GeorgiaAthensUnited States
| | - Claire E Eyers
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Patrick A Eyers
- Institute of Bioinformatics, University of GeorgiaAthensUnited States
| | - Natarajan Kannan
- Department of Biochemistry and Molecular Biology, University of GeorgiaAthensUnited States
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| |
Collapse
|
5
|
Liu Z, Wu B, Shi X, Zhou J, Huang H, Li Z, Yang M. Immune profiling of premalignant lesions in patients with Peutz-Jeghers syndrome. United European Gastroenterol J 2025; 13:338-348. [PMID: 39174496 PMCID: PMC11999043 DOI: 10.1002/ueg2.12650] [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: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Peutz-Jeghers syndrome (PJS), is a rare autosomal dominant hereditary disease characterized by an elevated risk of various cancers. Serine/Threonine Kinase 11 (STK11) gene is a major tumor suppressor crucial for immune evasion with and beyond tumorigenic cells. It has garnered increasing attention in the realm of oncology treatment, particularly in the context of immunotherapy development. OBJECTIVE This study aimed to assess the suitability of polyps obtained from individuals with PJS, resulting from germline STK11 deficiency, for immunotherapy. Additionally, we seek to identify potential shared mechanisms related to immune evasion between PJS polyps and cancers. To achieve this, we examined PJS polyps alongside familial adenomatous polyposis (FAP) and sporadic polyps. METHODS Polyps were compared among themselves and with either the paracancerous tissues or colon cancers. Pathological and gene expression profiling approaches were employed to characterize infiltrating immune cells and assess the expression of immune checkpoint genes. RESULTS Our findings revealed that PJS polyps exhibited a closer resemblance to cancer tissues than other polyps in terms of their immune microenvironment. Notably, PJS polyps displayed heightened expression of the immune checkpoint gene CD80 and an accumulation of myeloid cells, particularly myeloid-derived suppressor cells (MDSCs). CONCLUSION The findings suggest an immunobiological foundation for the increased cancer susceptibility in PJS patients, paving the way for potential immune therapy applications in this population. Furthermore, utilizing PJS as a model may facilitate the exploration of immune evasion mechanisms, benefiting both PJS and cancer patients.
Collapse
Affiliation(s)
- Zhongyue Liu
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Boda Wu
- Department of GastroenterologyPeking University Shenzhen HospitalShenzhenGuangdongChina
| | - Xiaoliu Shi
- Department of Medical GeneticsThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of GastroenterologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Junfeng Zhou
- Endoscopic Medical CenterThe Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Hui Huang
- Department of Medical GeneticsThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zhihong Li
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Mei Yang
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of OrthopedicsThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
6
|
Rong D, Gao L, Chen Y, Gao XZ, Tang M, Tang H, Gao Y, Lu G, Ling ZQ, Shen HM. Suppression of the LKB1-AMPK-SLC7A11-GSH signaling pathway sensitizes NSCLC to albumin-bound paclitaxel via oxidative stress. Redox Biol 2025; 81:103567. [PMID: 40023979 PMCID: PMC11915006 DOI: 10.1016/j.redox.2025.103567] [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: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Albumin-bound paclitaxel (nab-PTX) is an important chemotherapeutic drug used for the treatment of advanced and metastatic non-small cell lung cancer (NSCLC). One critical issue in its clinical application is the development of resistance; thus, a deeper understanding of the mechanisms underlying the primary resistance to nab-PTX is expected to help to develop effective therapeutic strategies to overcome resistance. In this study, we made an unexpected discovery that NSCLC with wild-type (WT) Liver kinase B1 (LKB1), an important tumor suppressor and upstream kinase of AMP-activated protein kinase (AMPK), is more resistant to nab-PTX than NSCLC with mutant LKB1. Mechanistically, LKB1 status does not alter the intracellular concentration of nab-PTX or affect its canonical pharmacological action in promoting microtubule polymerization. Instead, we found that LKB1 mediates AMPK activation, leading to increased expression of SLC7A11, a key amino acid transporter and intracellular level of glutathione (GSH), which then attenuates the production of reactive oxygen species (ROS) and apoptotic cell death induced by nab-PTX. On the other hand, genetic or pharmacological inhibition of AMPK in LKB1-WT NSCLC reduces the expression of SLC7A11 and intracellular GSH, increases ROS level, and eventually promotes the apoptotic cell death induced by nab-PTX in vitro. Consistently, the combination of nab-PTX with an AMPK inhibitor exhibits a greater therapeutic efficacy in LKB1-WT NSCLC using xenograft models in vivo. Taken together, our data reveal a novel role of LKB1-AMPK-SLC7A11-GSH signaling pathway in the primary resistance to nab-PTX, and provide a therapeutic strategy for the treatment of LKB1-WT NSCLC by targeting the LKB1-AMPK-SLC7A11-GSH pathway.
Collapse
Affiliation(s)
- Dade Rong
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Yiguan Chen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Xiang-Zheng Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Mingzhu Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Haimei Tang
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China; Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Gao
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Qiang Ling
- Experimental Research Centre, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Han-Ming Shen
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
7
|
Mohanty SS, Warrier S, Rangarajan A. Rethinking AMPK: A Reversible Switch Fortifying Cancer Cell Stress-Resilience. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2025; 98:33-52. [PMID: 40165808 PMCID: PMC11952127 DOI: 10.59249/jkbb6336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Stress adaptation is an evolutionarily conserved mechanism that promotes survival in the face of adverse conditions. AMP-activated protein kinase (AMPK) is a highly conserved energy-sensing kinase found in nearly all eukaryotic cells. It maintains energy homeostasis by promoting catabolism and inhibiting anabolism. In the context of cancer, the role of AMPK is controversial. It was initially touted as a tumor suppressor due to its association with Liver Kinase B1 (LKB1) (an upstream regulator and a known tumor suppressor) and ensuing growth-suppressive actions. However, emerging studies across a variety of cancer types unambiguously reveal AMPK's pro-survival and, thus, tumor-promoting activity, especially under cancer-associated stresses such as hypoxia, nutrient deprivation, oxidative stress, matrix detachment, and chemotherapy. In cancer cells, AMPK is activated in response to stress-induced increases in the levels of adenosine monophosphate (AMP), Ca2+, or reactive oxygen species (ROS). Upon activation, AMPK engages in metabolic rewiring and crosstalk with signaling molecules to mobilize resources toward survival while compromising proliferation. Here, we posit that AMPK is a non-genetic "reversible switch," allowing cancer cells' phenotype to switch to dormant, stem-like, and drug-resistant states, thereby enabling tumor cell survival, pathological progression, and therapy resistance. This review underscores the critical role of AMPK in driving cancer cell stress resilience and survival, advocating for the strategic use of AMPK inhibitors to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Shraddha S. Mohanty
- Department of Developmental Biology and Genetics, Indian Institute of Science,
Bengaluru, India
| | - Shweta Warrier
- Department of Developmental Biology and Genetics, Indian Institute of Science,
Bengaluru, India
| | - Annapoorni Rangarajan
- Department of Developmental Biology and Genetics, Indian Institute of Science,
Bengaluru, India
| |
Collapse
|
8
|
Chen Y, Lee K, Woo J, Kim DW, Keum C, Babbi G, Casadio R, Martelli PL, Savojardo C, Manfredi M, Shen Y, Sun Y, Katsonis P, Lichtarge O, Pejaver V, Seward DJ, Kamandula A, Bakolitsa C, Brenner SE, Radivojac P, O'Donnell-Luria A, Mooney SD, Jain S. Evaluating predictors of kinase activity of STK11 variants identified in primary human non-small cell lung cancers. Hum Genet 2025; 144:127-142. [PMID: 39934475 PMCID: PMC11976797 DOI: 10.1007/s00439-025-02726-0] [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: 06/15/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025]
Abstract
Critical evaluation of computational tools for predicting variant effects is important considering their increased use in disease diagnosis and driving molecular discoveries. In the sixth edition of the Critical Assessment of Genome Interpretation (CAGI) challenge, a dataset of 28 STK11 rare variants (27 missense, 1 single amino acid deletion), identified in primary non-small cell lung cancer biopsies, was experimentally assayed to characterize computational methods from four participating teams and five publicly available tools. Predictors demonstrated a high level of performance on key evaluation metrics, measuring correlation with the assay outputs and separating loss-of-function (LoF) variants from wildtype-like (WT-like) variants. The best participant model, 3Cnet, performed competitively with well-known tools. Unique to this challenge was that the functional data was generated with both biological and technical replicates, thus allowing the assessors to realistically establish maximum predictive performance based on experimental variability. Three out of the five publicly available tools and 3Cnet approached the performance of the assay replicates in separating LoF variants from WT-like variants. Surprisingly, REVEL, an often-used model, achieved a comparable correlation with the real-valued assay output as that seen for the experimental replicates. Performing variant interpretation by combining the new functional evidence with computational and population data evidence led to 16 new variants receiving a clinically actionable classification of likely pathogenic (LP) or likely benign (LB). Overall, the STK11 challenge highlights the utility of variant effect predictors in biomedical sciences and provides encouraging results for driving research in the field of computational genome interpretation.
Collapse
Affiliation(s)
- Yile Chen
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, 98105, USA
| | - Kyoungyeul Lee
- 3billion, 3billion Biotechnology Company, Seoul, South Korea
| | - Junwoo Woo
- 3billion, 3billion Biotechnology Company, Seoul, South Korea
| | - Dong-Wook Kim
- 3billion, 3billion Biotechnology Company, Seoul, South Korea
| | - Changwon Keum
- 3billion, 3billion Biotechnology Company, Seoul, South Korea
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Matteo Manfredi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Panagiotis Katsonis
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Olivier Lichtarge
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Vikas Pejaver
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David J Seward
- Department of Pathology, University of Vermont, Burlington, VT, 5445, USA
| | - Akash Kamandula
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | | | | | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Anne O'Donnell-Luria
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - Sean D Mooney
- Center for Information Technology, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Shantanu Jain
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA.
- The Institute for Experiential AI, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Su P, Lu Q, Wang Y, Mou Y, Jin W. Targeting MELK in tumor cells and tumor microenvironment: from function and mechanism to therapeutic application. Clin Transl Oncol 2025; 27:887-900. [PMID: 39187643 DOI: 10.1007/s12094-024-03664-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
Maternal embryonic leucine zipper kinase (MELK), a member of the adenosine monophosphate-activated protein kinase (AMPK) protein family, has been reported to be involved in the regulation of many cellular events. The aberrant expression of MELK is associated with tumorigenesis and malignant progression of various tumors. Moreover, MELK plays an essential role in the regulation of tumor microenvironment (TME), which affects the function of immune cells and the responsiveness to immunotherapy. Currently, small molecule inhibitors targeting MELK have been developed and evaluated in clinical trials. A comprehensive understanding of MELK may provide clues and confidence for subsequent basic research and scientific transformation. In this review, we provide a comprehensive overview of the structural features, molecular biological functions, and critical roles of MELK in tumors and TME, as well as the targeted agents under development for the treatment of tumors and discuss the perspective for MELK-targeted therapies for tumors.
Collapse
Affiliation(s)
- Pengfei Su
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China
| | - Qiliang Lu
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China
| | - Yuanyu Wang
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China
| | - Yiping Mou
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China
| | - Weiwei Jin
- Department of General Surgery, Cancer Center, Division of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China.
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310000, People's Republic of China.
| |
Collapse
|
10
|
Krauss RS, Kyba M. When everything is a master regulator, nothing is. Mol Biol Cell 2025; 36:pe3. [PMID: 39913302 PMCID: PMC11974949 DOI: 10.1091/mbc.e24-11-0494] [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: 11/13/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 04/09/2025] Open
Abstract
The term "master regulator" emerged in the 1960s and 1970s and referred to autoregulatory transcription factors that sat atop a developmental lineage. Since that time, usage of the term has increased and broadened to the point where it has lost clear meaning. Here we discuss the term "master regulator" with the goals of developing a consensus view of its definition and stimulating discussion on use of similar terms. We propose that the designation "master regulator" be reserved for transcription factors that are: 1) positioned at the top of a regulatory hierarchy specifying a cell lineage (and potentially specific cell states, such as hypoxia); and 2) sufficient to drive the transcriptional program characterizing that lineage or state. It is hoped that this piece will provide a precedent for use of additional terms applied to incompletely understood biological processes, resulting in experimentation that sheds light on such processes.
Collapse
Affiliation(s)
- Robert S. Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
11
|
Yin D, Lu X, Liang X, Lu Y, Xiong L, Wu P, Wang T, Chen J. STK11 genetic alterations in metastatic EGFR mutant lung cancer. Sci Rep 2025; 15:5729. [PMID: 39962098 PMCID: PMC11832735 DOI: 10.1038/s41598-024-74779-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/30/2024] [Indexed: 02/20/2025] Open
Abstract
This study was conducted to investigate the relationship between STK11 genetic alterations and the outcomes of patients with metastatic EGFR mutant lung cancer. Clinical characteristics and genomic data were downloaded from the cBioPortal database. The information of the case with STK11 mutation was collected from Jiangyin People's Hospital. Univariate and multivariate analyses were performed to distinguish the prognostic differences. Outcomes were analyzed before and after propensity score matching (PSM). A patient with STK11 mutation was insensitive to osimertinib and had an extremely poor prognosis. Further analysis showed that STK11 mutations had a strong mutual exclusion with EGFR mutations. A total of 960 patients with metastatic EGFR mutant lung adenocarcinoma were enrolled in the prognostic analysis. STK11 alternation was a significant predictor of worse outcomes in univariate or multivariate analyses. After PSM, patients with STK11 alternations still exhibited poor prognoses. Cell culture experiments also showed that the loss of STK11 could contribute to the resistance of osimertinib. Functionally, STK11 mutation was positively associated with metabolic signaling pathways and immune infiltrates negatively. Through drug screening, trametinib was identified to sensitize osimertinib in the STK11-deficient cell. This study found that STK11 genetic alterations portend a worse prognosis for patients with metastatic EGFR mutant lung cancer and led to osimertinib resistance potentially. MEK inhibitors could sensitize osimertinib in the STK11-deficient cell.
Collapse
Affiliation(s)
- Dandan Yin
- The Second Hospital of Nanjing, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, People's Republic of China
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, Jiangsu, People's Republic of China
| | - Xiyi Lu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xiao Liang
- Department of Oncology, The Affiliated Jiangyin Hospital of Nantong University, Wuxi, Jiangsu, People's Republic of China
| | - Yiting Lu
- Department of Radiology, The Affiliated Jiangyin Hospital of Nantong University, Wuxi, Jiangsu, People's Republic of China
| | - Lei Xiong
- Department of Cardiothoracic Surgery, Jinling Hospital, Jinling Clinical Medical School, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| | - Pingping Wu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institue of Cancer Research, Nanjing, Jiangsu, People's Republic of China.
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, People's Republic of China.
- Medical School, Nanjing University, Nanjing, People's Republic of China.
| | - Jinfei Chen
- Medical School, Nanjing University, Nanjing, People's Republic of China.
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
12
|
Nakata S, Fujiyama T, Asano F, Komiya H, Hotta-Hirashima N, Juichi M, Komine D, Kakizaki M, Ikkyu A, Mizuno S, Takahashi S, Miyoshi C, Funato H, Yanagisawa M. Partial activation of salt-inducible kinase 3 delays the onset of wakefulness and alleviates hypersomnia due to the lack of protein kinase A-phosphorylation site. Sleep 2025; 48:zsae279. [PMID: 39656684 PMCID: PMC11807893 DOI: 10.1093/sleep/zsae279] [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: 01/12/2024] [Revised: 10/23/2024] [Indexed: 12/17/2024] Open
Abstract
STUDY OBJECTIVES Sleep/wakefulness is regulated by intracellular signaling pathways composed of protein kinases such as salt-inducible kinase 3 (Sik3). Sik3-deficiency in neurons decreases nonrapid eye movement (NREM) sleep time and electroencephalogram (EEG) delta power during NREM sleep, while Sik3Slp mice lacking a protein kinase A (PKA)-phosphorylation site, S551, show hypersomnia phenotype. In this study, we examined how a phosphomimetic mutation of the 221st threonine residue (T221E), which provides a partial (weak) constitutive activity of the kinase, affects sleep/wakefulness and circadian behavior. We also examined the effect of T221E substitution on the hypersomnia phenotype of Sik3Slp mice. METHODS We examined the sleep/wake behavior of heterozygous and homozygous Sik3T221E mice and Sik3T221E;Slp mice using EEG and electromyogram recording. We also examined the circadian behavior of Sik3T221E mice using a running wheel under the light-dark cycle and constant darkness. RESULTS Heterozygous and homozygous Sik3T221E mice showed normal sleep time and sleep homeostatic responses. Homozygous Sik3T221E mice exhibited a delayed onset of wakefulness at the early dark phase and longer circadian periods. Sik3T221E;Slp mice showed decreased NREM sleep time and homeostatic responses compared to Sik3Slp mice. CONCLUSIONS Our results suggest that the peak onset of wakefulness is sensitive to disturbed kinase activity of SIK3, and the relationship between phosphorylation at T221 and S551 is critical for regulating sleep need.
Collapse
Affiliation(s)
- Shinya Nakata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tomoyuki Fujiyama
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Fuyuki Asano
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruna Komiya
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Noriko Hotta-Hirashima
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Motoki Juichi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Daiki Komine
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Miyo Kakizaki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aya Ikkyu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center and Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chika Miyoshi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
13
|
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
|
14
|
Qin W, Ding Y, Zhang W, Sun L, Weng J, Zheng X, Luo S. Small molecule-driven LKB1 deacetylation is responsible for the inhibition of hepatic lipid response in NAFLD. J Lipid Res 2025; 66:100740. [PMID: 39755206 PMCID: PMC11808498 DOI: 10.1016/j.jlr.2024.100740] [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: 02/29/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive condition characterized by ectopic fat accumulation in the liver, for which no FAD-approved drugs currently exist. Emerging evidence highlights the role of liver kinase B1 (LKB1), a key metabolic regulator, has been proposed in NAFLD, particularly in response to excessive nutrient levels. However, few agents have been identified that can prevent the progression of nonalcoholic steatohepatitis (NASH) by targeting LKB1 deacetylation. Through comprehensive screening of our in-house chemical library, we identified tranilast, a small molecule with remarkable inhibitory efficacy against lipid deposition induced by palmitic acid/oleic acid (PO). In this study, we investigated the novel biological function and mechanism of tranilast in regulating hepatic lipid response in NAFLD, focusing on its role in LKB1 deacetylation within hepatocytes. Our findings demonstrate that tranilast effectively reduced hepatic steatosis, inflammation, and fibrosis in NASH models induced by high-fat and high-cholesterol (HFHC) and methionine choline-deficient (MCD) diets. Mechanistic analysis using RNA sequencing revealed that tranilast mitigated hepatic lipid response by promoting LKB1 deacetylation and activating AMPK. Notably, in vivo experiments showed that the beneficial effects of tranilast in MCD diet-induced NASH model were reversed by the compound C (C-C), a known AMPK inhibitor, confirming that tranilast's effects on hepatic lipid response are mediated through the AMPK pathway. In summary, tranilast inhibits hepatic lipid response in NAFLD through LKB1 deacetylation, providing robust experimental evidence for the role of LKB1 in NAFLD. These findings position tranilast as a promising therapeutic candidate for the pharmacological management of metabolic diseases.
Collapse
Affiliation(s)
- Weiwei Qin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China; Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Ding
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenhao Zhang
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lu Sun
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xueying Zheng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Sihui Luo
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
15
|
Liu Z, Guo Z, Xu J, Zhou R, Shi B, Chen L, Wu C, Wang H, Wang X, Wang F, Li Q, Liu Q. Regulation of Sleep Amount by CRTC1 via Transcription of Crh in Mice. J Neurosci 2025; 45:e0786242024. [PMID: 39622645 PMCID: PMC11780352 DOI: 10.1523/jneurosci.0786-24.2024] [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: 04/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 01/31/2025] Open
Abstract
The cAMP response element binding protein (CREB) is required for regulation of daily sleep amount, whereas gain of function of CREB-regulated transcription coactivator 1 (CRTC1) causes severe insomnia in mice. However, the physiological functions of CRTCs and their downstream target genes in the regulation of sleep amount remain unclear. Here, we use an adult brain chimeric (ABC)-expression/knock-out platform for somatic genetic analysis of sleep in adult male mice. ABC expression of constitutively active mutant CRTC1/2CA in the mouse brain neurons significantly reduces the amount of non-rapid eye movement sleep (NREMS) and/or rapid eye movement sleep (REMS). Consistent with the fact that SIK3 phosphorylates and inhibits CRTCs, ABC expression of CRTC1/2/3CA rescues the hypersomnia phenotype of Sleepy (Sik3Slp ) mice. While ABC-Crtc2KO or Crtc3KO causes no sleep phenotype, ABC-Crtc1KO or ABC expression of dominant-negative CRTC (dnCRTC) results in a modest reduction of NREMS amount accompanied with elevated NREMS delta power. Moreover, ABC expression of CRTC1CA or dnCRTC in the excitatory neurons causes bidirectional changes of NREMS/REMS amount and/or NREMS delta power. The ability of CRTC1CA to regulate sleep requires its transactivation domain and CREB-binding domain and is dependent on CREB. Furthermore, we showed that inducible ABC expression of corticotropin-releasing hormone (Crh) and brain-derived neurotrophic factor (Bdnf)-two target genes of CRTCs-significantly reduces daily sleep amount. Notably, ABC-CrhKO , but not BdnfKO , rescues the insomnia phenotype of ABC-CRTC1CA mice. Taken together, these results indicate that the CREB-CRTC1 complex regulates daily sleep amount by modulating the transcription of Crh in the mouse brain neurons.
Collapse
Affiliation(s)
- Zhihao Liu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Zhiyong Guo
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junjie Xu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Rui Zhou
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Bihan Shi
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Lin Chen
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Chongyang Wu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Haiyan Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Xia Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing (NIBS), Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| |
Collapse
|
16
|
Yu J, Liu H, Gao R, Wang TV, Li C, Liu Y, Yang L, Xu Y, Cui Y, Jia C, Huang J, Chen PR, Rao Y. Calcineurin: An essential regulator of sleep revealed by biochemical, chemical biological, and genetic approaches. Cell Chem Biol 2025; 32:157-173.e7. [PMID: 39740665 DOI: 10.1016/j.chembiol.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025]
Abstract
Research into mechanisms underlying sleep traditionally relies on electrophysiology and genetics. Because sleep can only be measured on whole animals by behavioral observations and physical means, no sleep research was initiated by biochemical and chemical biological approaches. We used phosphorylation sites of kinases important for sleep as targets for biochemical and chemical biological approaches. Sleep was increased in mice carrying a threonine-to-alanine substitution at residue T469 of salt-inducible kinase 3 (SIK3). Our biochemical purification and photo-crosslinking revealed calcineurin (CaN) dephosphorylation, both in vitro and in vivo, of SIK3 at T469 and S551, but not T221. Knocking down CaN regulatory subunit reduced daily sleep by more than 5 h, exceeding all known mouse mutants. Our work uncovered a critical physiological role for CaN in sleep and pioneered biochemical purification and chemical biology as effective approaches to study sleep.
Collapse
Affiliation(s)
- Jianjun Yu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Huijie Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Rui Gao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenggang Li
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Lu Yang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Ying Xu
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Yunfeng Cui
- Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenxi Jia
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Juan Huang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Peng R Chen
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Liu X, Wang S, Lv H, Chen E, Yan L, Yu J. Advances in the relationship of immune checkpoint inhibitors and DNA damage repair. Curr Res Transl Med 2025; 73:103494. [PMID: 39824061 DOI: 10.1016/j.retram.2025.103494] [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: 01/22/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Cancer immunotherapy, alongside surgery, radiation therapy, and chemotherapy, has emerged as a key treatment modality. Immune checkpoint inhibitors (ICIs) represent a promising immunotherapy that plays a critical role in the management of various solid tumors. However, the limited efficacy of ICI monotherapy and the development of primary or secondary resistance to combination therapy remain a challenge. Consequently, identifying molecular markers for predicting ICI efficacy has become an area of active clinical research. Notably, the correlation between DNA damage repair (DDR) mechanisms and the effectiveness of ICI treatment has been established. This review outlines the two primary pathways of DDR, namely, the homologous recombination repair pathway and the mismatch repair pathway. The relationship between these key genes and ICIs has been discussed and the potential of these genes as molecular markers for predicting ICI efficacy summarized.
Collapse
Affiliation(s)
- Xiaolin Liu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Hongwei Lv
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Enli Chen
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Li Yan
- School of Humanities, Beijing University of Chinese Medicine, Beijing, PR China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
18
|
Ahn EH, Park JB. Molecular Mechanisms of Alzheimer's Disease Induced by Amyloid-β and Tau Phosphorylation Along with RhoA Activity: Perspective of RhoA/Rho-Associated Protein Kinase Inhibitors for Neuronal Therapy. Cells 2025; 14:89. [PMID: 39851517 PMCID: PMC11764136 DOI: 10.3390/cells14020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Amyloid-β peptide (Aβ) is a critical cause of Alzheimer's disease (AD). It is generated from amyloid precursor protein (APP) through cleavages by β-secretase and γ-secretase. γ-Secretase, which includes presenilin, is regulated by several stimuli. Tau protein has also been identified as a significant factor in AD. In particular, Tau phosphorylation is crucial for neuronal impairment, as phosphorylated Tau detaches from microtubules, leading to the formation of neurofibrillary tangles and the destabilization of the microtubule structure. This instability in microtubules damages axons and dendrites, resulting in neuronal impairment. Notably, Aβ is linked to Tau phosphorylation. Another crucial factor in AD is neuroinflammation, primarily occurring in the microglia. Microglia possess several receptors that bind with Aβ, triggering the expression and release of an inflammatory factor, although their main physiological function is to phagocytose debris and pathogens in the brain. NF-κB activation plays a major role in neuroinflammation. Additionally, the production of reactive oxygen species (ROS) in the microglia contributes to this neuroinflammation. In microglia, superoxide is produced through NADPH oxidase, specifically NOX2. Rho GTPases play an essential role in regulating various cellular processes, including cytoskeletal rearrangement, morphology changes, migration, and transcription. The typical function of Rho GTPases involves regulating actin filament formation. Neurons, with their complex processes and synapse connections, rely on cytoskeletal dynamics for structural support. Other brain cells, such as astrocytes, microglia, and oligodendrocytes, also depend on specific cytoskeletal structures to maintain their unique cellular architectures. Thus, the aberrant regulation of Rho GTPases activity can disrupt actin filaments, leading to altered cell morphology, including changes in neuronal processes and synapses, and potentially contributing to brain diseases such as AD.
Collapse
Affiliation(s)
- Eun Hee Ahn
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea;
- Department of Neurology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- ELMED Co., Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| |
Collapse
|
19
|
Cohen P, Snelling T. Diseases caused by altered specificity of a protein kinase for its allosteric activators. Trends Biochem Sci 2025; 50:61-70. [PMID: 39580356 DOI: 10.1016/j.tibs.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/11/2024] [Accepted: 10/25/2024] [Indexed: 11/25/2024]
Abstract
Protein kinases regulate many intracellular processes, and their dysregulation causes cancers and other diseases. This review focuses on the atypical alpha-kinase 1 (ALPK1), which is activated in mammalian cells by nucleoside diphosphate heptoses (ADP-heptose, UDP-heptose, and CDP-heptose) produced by microbial pathogens but not by mammalian cells. Mutations in human ALPK1 cause ROSAH syndrome and spiradenoma, which result from an alteration in its specificity for nucleoside diphosphate heptoses, causing aberrant activation by mammalian nucleoside diphosphate sugars without microbial infection. These may be the first diseases caused by altered specificity of an enzyme for its allosteric activators and has suggested ways in which selective drugs could be developed to treat them without compromising the innate immune system.
Collapse
Affiliation(s)
- Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Scotland, UK.
| | - Tom Snelling
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Scotland, UK.
| |
Collapse
|
20
|
Poullennec KG, Jnoff E, Abendroth J, Bhuma N, Calmiano M, Calmus L, Cardenas A, Courade JP, Delatour C, Hall A, de Haro T, Delker SL, Demaude T, Gaikwad N, Ghavate D, Gholap AR, Kierkowicz M, Le Mestre R, Van Hijfte N, Verheijden S, Vernerova K, De Wever V, Waghmode N. Discovery of UCB9386: A Potent, Selective, and Brain-Penetrant Nuak1 Inhibitor Suitable for In Vivo Pharmacological Studies. J Med Chem 2024; 67:20879-20910. [PMID: 39588908 DOI: 10.1021/acs.jmedchem.4c01237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Nuak1 (NUAK family SnF1-like kinase-1) is a serine-threonine kinase and a member of the AMPK family. Interest in Nuak1 has increased over the years due to the role it plays in several biological processes, from tumor cell invasion and proliferation to Tau stabilization. Nuak1 is expressed in many cancer cell lines and many reports describe this target as an oncogene, the inhibition of which is hypothesized to be valuable for treating various cancer types including glioma. We report here the discovery of Nuak1 inhibitors originating from HTS hit 9 with excellent selectivity and the subsequent medicinal chemistry optimization program, supported by structural information from the first crystal structures of a Nuak1 chimeric protein which provided insights into the binding modes of our compounds. These efforts yielded a nanomolar cell potent, highly selective and brain penetrant Nuak1 inhibitor UCB9386 (56) suitable for in vivo pharmacological studies for central nervous system (CNS) disorders.
Collapse
Affiliation(s)
| | - Eric Jnoff
- Chemin du Foriest, UCB, 1420 Braine l'Alleud, Belgium
| | - Jan Abendroth
- UCB Seattle, Bainbridge Island, Washington 98110, United States
| | - Naresh Bhuma
- Illumina Centre, 19 Granta Park, Great Abingdon CB21 6DF, United Kingdom
| | | | - Laurent Calmus
- NovAliX, 240 Parc d'Affaires des Portes, 27100 Val-de-Reuil, France
| | | | | | | | - Adrian Hall
- Chemin du Foriest, UCB, 1420 Braine l'Alleud, Belgium
| | | | | | | | - Nilesh Gaikwad
- Sai Life Sciences Ltd, IKP Knowledge Park, Genome Valley, Turkapally, Hyderabad 500078, India
| | - Dnyaneshwar Ghavate
- Sai Life Sciences Ltd, IKP Knowledge Park, Genome Valley, Turkapally, Hyderabad 500078, India
| | - Atul R Gholap
- Sai Life Sciences Ltd, IKP Knowledge Park, Genome Valley, Turkapally, Hyderabad 500078, India
| | | | - Régis Le Mestre
- Minoryx Therapeutics, Avenue Jean Mermoz 32, 6041 Charleroi, Belgium
| | | | - Simon Verheijden
- Janssen Research and Development, Antwerpseweg 15, 2340 Beerse, Belgium
| | | | | | | |
Collapse
|
21
|
Lambies G, Lee SW, Duong-Polk K, Aza-Blanc P, Maganti S, Galapate CM, Deshpande A, Deshpande AJ, Scott DA, Dawson DW, Commisso C. Cell polarity proteins promote macropinocytosis in response to metabolic stress. Nat Commun 2024; 15:10541. [PMID: 39627191 PMCID: PMC11614886 DOI: 10.1038/s41467-024-54788-9] [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: 09/29/2023] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
Macropinocytosis has emerged as a scavenging pathway that cancer cells exploit to survive in a nutrient-deprived microenvironment. Tumor cells are especially reliant on glutamine for their survival, and in pancreatic ductal adenocarcinoma (PDAC) cells, glutamine deficiency can enhance the stimulation of macropinocytosis. Here, we identify the atypical protein kinase C (aPKC) enzymes, PKCζ and PKCι, as regulators of macropinocytosis. In normal epithelial cells, aPKCs associate with the scaffold proteins Par3 and Par6 to regulate cell polarity, affecting several targets, including the Par1 kinases and we find that each of these proteins is required for macropinocytosis. Mechanistically, aPKCs are regulated by EGFR signaling or by the transcription factor CREM to promote the Par3 relocation to microtubules, facilitating macropinocytosis in a dynein-dependent manner. Importantly, cell fitness impairment caused by aPKC depletion is rescued by the restoration of macropinocytosis and aPKCs support PDAC growth in vivo. Our findings enhance our understanding of the mechanistic underpinnings that control macropinocytic uptake in the context of metabolic stress.
Collapse
Affiliation(s)
- Guillem Lambies
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Szu-Wei Lee
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Karen Duong-Polk
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Pedro Aza-Blanc
- Functional Genomics Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Swetha Maganti
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Cheska M Galapate
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Anagha Deshpande
- Cancer Genome and Epigenetics Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Aniruddha J Deshpande
- Cancer Genome and Epigenetics Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - David A Scott
- Cancer Metabolism Core Resource, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - David W Dawson
- Department of Pathology and Laboratory Medicine and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Cosimo Commisso
- Cancer Metabolism and Microenvironment Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
22
|
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
|
23
|
Smiles WJ, Ovens AJ, Oakhill JS, Kofler B. The metabolic sensor AMPK: Twelve enzymes in one. Mol Metab 2024; 90:102042. [PMID: 39362600 PMCID: PMC11752127 DOI: 10.1016/j.molmet.2024.102042] [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: 08/02/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) is an evolutionarily conserved regulator of energy metabolism. AMPK is sensitive to acute perturbations to cellular energy status and leverages fundamental bioenergetic pathways to maintain cellular homeostasis. AMPK is a heterotrimer comprised of αβγ-subunits that in humans are encoded by seven individual genes (isoforms α1, α2, β1, β2, γ1, γ2 and γ3), permitting formation of at least 12 different complexes with personalised biochemical fingerprints and tissue expression patterns. While the canonical activation mechanisms of AMPK are well-defined, delineation of subtle, as well as substantial, differences in the regulation of heterogenous AMPK complexes remain poorly defined. SCOPE OF REVIEW Here, taking advantage of multidisciplinary findings, we dissect the many aspects of isoform-specific AMPK function and links to health and disease. These include, but are not limited to, allosteric activation by adenine nucleotides and small molecules, co-translational myristoylation and post-translational modifications (particularly phosphorylation), governance of subcellular localisation, and control of transcriptional networks. Finally, we delve into current debate over whether AMPK can form novel protein complexes (e.g., dimers lacking the α-subunit), altogether highlighting opportunities for future and impactful research. MAJOR CONCLUSIONS Baseline activity of α1-AMPK is higher than its α2 counterpart and is more sensitive to synergistic allosteric activation by metabolites and small molecules. α2 complexes however, show a greater response to energy stress (i.e., AMP production) and appear to be better substrates for LKB1 and mTORC1 upstream. These differences may explain to some extent why in certain cancers α1 is a tumour promoter and α2 a suppressor. β1-AMPK activity is toggled by a 'myristoyl-switch' mechanism that likely precedes a series of signalling events culminating in phosphorylation by ULK1 and sensitisation to small molecules or endogenous ligands like fatty acids. β2-AMPK, not entirely beholden to this myristoyl-switch, has a greater propensity to infiltrate the nucleus, which we suspect contributes to its oncogenicity in some cancers. Last, the unique N-terminal extensions of the γ2 and γ3 isoforms are major regulatory domains of AMPK. mTORC1 may directly phosphorylate this region in γ2, although whether this is inhibitory, especially in disease states, is unclear. Conversely, γ3 complexes might be preferentially regulated by mTORC1 in response to physical exercise.
Collapse
Affiliation(s)
- William J Smiles
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria; Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia.
| | - Ashley J Ovens
- Protein Engineering in Immunity & Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia; Department of Medicine, University of Melbourne, Parkville, Australia
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
24
|
Diaz FM, Godinez DS, Solano F, Jasani A, Alcaide M, Kellogg DR. Mechanisms of growth-dependent regulation of the Gin4 kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624605. [PMID: 39605684 PMCID: PMC11601526 DOI: 10.1101/2024.11.20.624605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cell cycle progression is dependent upon cell growth. Cells must therefore translate growth into a proportional signal that can be used to determine when there has been sufficient growth for cell cycle progression. In budding yeast, the protein kinase Gin4 is required for normal control of cell growth and undergoes gradual hyperphosphorylation and activation that are dependent upon growth and proportional to the extent of growth, which suggests that Gin4 could function in mechanisms that measure cell growth. However, the molecular mechanisms that drive hyperphosphorylation of Gin4 are poorly understood. Here, we used biochemical reconstitution and genetic analysis to test hypotheses for the mechanisms that drive phosphorylation of Gin4. We ruled out a previous model in which phosphatidylserine delivered to sites of plasma membrane growth binds Gin4 to initiate autophosphorylation. Instead, we show that Elm1, a homolog of the mammalian Lkb1 tumor suppressor kinase, is sufficient to promote hyperphosphorylation of Gin4 in vitro, likely via initiation of Gin4 autophosphorylation. Furthermore, we show that casein kinase I is required for growth-dependent hyperphosphorylation of Gin4 and also for normal regulation of Elm1. Together, these discoveries lead to new insight into mechanisms that link cell cycle progression to cell growth.
Collapse
Affiliation(s)
- Francisco Mendez Diaz
- Department of Molecular, Cell and Developmental Biology University of California, Santa Cruz
| | - David Sanchez Godinez
- Department of Molecular, Cell and Developmental Biology University of California, Santa Cruz
| | - Francisco Solano
- Department of Molecular, Cell and Developmental Biology University of California, Santa Cruz
| | - Akshi Jasani
- Department of Molecular, Cell and Developmental Biology University of California, Santa Cruz
| | - Maria Alcaide
- Department of Molecular, Cell and Developmental Biology University of California, Santa Cruz
| | - Douglas R Kellogg
- Department of Molecular, Cell and Developmental Biology University of California, Santa Cruz
| |
Collapse
|
25
|
Salt IP, Carling D. A special issue of Essays in Biochemistry on AMPK and AMPK-related kinases. Essays Biochem 2024; 68:269-271. [PMID: 39552567 DOI: 10.1042/ebc20240038] [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: 10/17/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024]
Abstract
In eukaryotic cells, AMP-activated protein kinase (AMPK) plays a central role in responding to nutrient limitation by switching-off ATP-consuming (anabolic) pathways and switching-on ATP generating (catabolic) pathways. Over the last 30 years or so, a considerable body of research has been carried out that has provided us with a wealth of knowledge regarding the regulation and role of AMPK. Despite this, there is still much to learn about AMPK and the field remains highly active, with many groups around the world continuing to explore new roles for AMPK, providing insight into its biological function. This review series was inspired by recent AMPK-focused meetings in Scotland (2022) and Australia (2023) and draws on some of the research presented at those meetings.
Collapse
Affiliation(s)
- Ian P Salt
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - David Carling
- MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Imperial College London, London W12 0HS, U.K
- Institute of Clinical Sciences, Imperial College London, London W12 0NN, U.K
| |
Collapse
|
26
|
Skalka GL, Whyte D, Lubawska D, Murphy DJ. NUAK: never underestimate a kinase. Essays Biochem 2024; 68:295-307. [PMID: 38939918 DOI: 10.1042/ebc20240005] [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: 04/04/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
NUAK1 and NUAK2 belong to a family of kinases related to the catalytic α-subunits of the AMP-activated protein kinase (AMPK) complexes. Despite canonical activation by the tumour suppressor kinase LKB1, both NUAKs exhibit a spectrum of activities that favour tumour development and progression. Here, we review similarities in structure and function of the NUAKs, their regulation at gene, transcript and protein level, and discuss their phosphorylation of specific downstream targets in the context of the signal transduction pathways and biological activities regulated by each or both NUAKs.
Collapse
Affiliation(s)
- George L Skalka
- School of Cancer Sciences, University of Glasgow, Glasgow, U.K
- CRUK Scotland Institute, Garscube Estate, Glasgow G61 1BD, U.K
| | - Declan Whyte
- CRUK Scotland Institute, Garscube Estate, Glasgow G61 1BD, U.K
| | | | - Daniel J Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, U.K
- CRUK Scotland Institute, Garscube Estate, Glasgow G61 1BD, U.K
| |
Collapse
|
27
|
Trelford CB, Buensuceso A, Tomas E, Valdes YR, Hovey O, Li SSC, Shepherd TG. LKB1 and STRADα Promote Epithelial Ovarian Cancer Spheroid Cell Invasion. Cancers (Basel) 2024; 16:3726. [PMID: 39594681 PMCID: PMC11591840 DOI: 10.3390/cancers16223726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Late-stage epithelial ovarian cancer (EOC) involves the widespread dissemination of malignant disease throughout the peritoneal cavity, often accompanied by ascites. EOC metastasis relies on the formation of multicellular aggregates, called spheroids. Given that Liver Kinase B1 (LKB1) is required for EOC spheroid viability and LKB1 loss in EOC cells decreases tumor burden in mice, we investigated whether the LKB1 complex controls the invasive properties of human EOC spheroids. LKB1 signalling was antagonized through the CRISPR/Cas9 genetic knockout of LKB1 and/or the RNAi-dependent targeting of STE20-related kinase adaptor protein (STRAD, an LKB1 activator). EOC spheroids expressing nuclear GFP (green) or mKate2 (red) constructs were embedded in Matrigel for real-time live-cell invasion monitoring. Migration and invasion were also assessed in spheroid culture using Transwell chambers, spheroid reattachment, and mesothelial clearance assays. The loss of LKB1 and STRAD signalling decreased cell invasion through Matrigel and Transwell membranes, as well as mesothelial cell clearance. In the absence of LKB1, zymographic assays identified a loss of matrix metalloproteinase (MMP) activity, whereas spheroid reattachment assays found that coating plates with fibronectin restored their invasive potential. A three-dimensional EOC organoid model demonstrated that organoid area was greatly reduced by LKB1 loss. Overall, our data indicated that LKB1 and STRAD facilitated EOC metastasis by promoting MMP activity and fibronectin expression. Given that LKB1 and STRAD are crucial for EOC metastasis, targeting LKB1 and/or STRAD could disrupt the dissemination of EOC, making inhibitors of the LKB1 pathway an alternative therapeutic strategy for EOC patients.
Collapse
Affiliation(s)
- Charles B. Trelford
- The Mary & John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 4L6, Canada; (C.B.T.)
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
| | - Adrian Buensuceso
- The Mary & John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 4L6, Canada; (C.B.T.)
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
| | - Emily Tomas
- The Mary & John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 4L6, Canada; (C.B.T.)
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
| | - Yudith Ramos Valdes
- The Mary & John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 4L6, Canada; (C.B.T.)
| | - Owen Hovey
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
| | - Shawn Shun-Cheng Li
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
- Children’s Health Research Institute, London, ON N6A 4L6, Canada
| | - Trevor G. Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 4L6, Canada; (C.B.T.)
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 4L6, Canada
- Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 4L6, Canada
| |
Collapse
|
28
|
Kuser-Abali G, Ugurlu-Bayarslan A, Yilmaz Y, Ozcan F, Karaer F, Bugra K. SIK2: A Novel Negative Feedback Regulator of FGF2 Signaling. Adv Biol (Weinh) 2024; 8:e2400032. [PMID: 39267218 DOI: 10.1002/adbi.202400032] [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: 01/18/2024] [Revised: 08/20/2024] [Indexed: 09/17/2024]
Abstract
A wide range of cells respond to fibroblast growth factor 2 (FGF2) by proliferation via activation of the Ras/ERK1/2 pathway. In this study, the potential involvement of salt inducible kinase SIK2) in this cascade within retinal Müller glia is explored. It is found that SIK2 phosphorylation status and activity are modulated in an FGF2-dependent manner, possibly via ERK1/2. With SIK2 downregulation, enhanced ERK1/2 activation with delayed attenuation and increased cell proliferation is observed, while SIK2 overexpression hampers FGF2-dependent ERK1/2 activation. In vitro kinase and site-directed mutagenesis studies indicate that SIK2 targets the pathway element GRB2-associated-binding protein 1 (Gab1) on Ser266. This phosphorylation event weakens Gab1 interactions with its partners growth factor receptor-bound protein 2 (Grb2) and Src homology region 2 domain containing phosphatase 2 (Shp2). Collectively, these results suggest that during FGF2-dependent proliferation process ERK1/2-mediated activation of SIK2 targets Gab1, resulting in downregulation of the Ras/ERK1/2 cascade in a feedback loop.
Collapse
Affiliation(s)
- Gamze Kuser-Abali
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, 34342, Turkey
- Faculty of Medicine Nursing & Health Sciences, The Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Asli Ugurlu-Bayarslan
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, 34342, Turkey
- Department of Biology, Kastamonu University, Kastamonu, 37150, Turkey
| | - Yeliz Yilmaz
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, 34342, Turkey
- Izmir Biomedicine and Genome Center, Izmir, 35340, Turkey
| | - Ferruh Ozcan
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, 34342, Turkey
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Kocaeli, 41400, Turkey
| | - Funda Karaer
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, 34342, Turkey
- Ministry of Education, Turkey
| | - Kuyas Bugra
- Department of Molecular Biology and Genetics, Bogazici University, Bebek, Istanbul, 34342, Turkey
- Life Sciences Center, Bogazici University, Bebek, Istanbul, 34342, Turkey
| |
Collapse
|
29
|
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
|
30
|
Homma D, Limlingan SJM, Saito T, Ando K. SARS-CoV-2-derived protein Orf9b enhances MARK2 activity via interaction with the autoinhibitory KA1 domain. FEBS Lett 2024; 598:2385-2393. [PMID: 38969617 DOI: 10.1002/1873-3468.14975] [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: 03/15/2024] [Revised: 06/01/2024] [Accepted: 06/12/2024] [Indexed: 07/07/2024]
Abstract
Microtubule affinity-regulating kinase 2 (MARK2) is a Ser/Thr protein kinase that regulates cell polarity and immune responses. Here, we report that Orf9b, one of the accessory proteins encoded in the SARS-CoV-2 genome, increases MARK2 activity via interaction with the autoinhibitory KAI domain. We found that co-expression of Orf9b enhances the kinase activity of MARK2 in HEK293 cells. Orf9b does not bind to or enhance the activity of the mutant form of MARK2 lacking the KA1 domain. Orf9b lowers inhibitory phosphorylation of MARK2 at T595 while mutation experiments indicate that this site is dispensable for Orf9b-mediated enhancement of MARK2 activity. Our results suggest that Orf9b enhances MARK2 activity by binding the autoinhibitory KA1 domain, which closely interacts with the kinase domain.
Collapse
Affiliation(s)
- Daiki Homma
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
| | | | - Taro Saito
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Japan
| |
Collapse
|
31
|
Conte E, Boccanegra B, Dinoi G, Pusch M, De Luca A, Liantonio A, Imbrici P. Therapeutic Approaches to Tuberous Sclerosis Complex: From Available Therapies to Promising Drug Targets. Biomolecules 2024; 14:1190. [PMID: 39334956 PMCID: PMC11429992 DOI: 10.3390/biom14091190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/29/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare multisystem disorder caused by heterozygous loss-of-function pathogenic variants in the tumour suppressor genes TSC1 and TSC2 encoding the tuberin and hamartin proteins, respectively. Both TSC1 and TSC2 inhibit the mammalian target of rapamycin (mTOR) complexes pathway, which is crucial for cell proliferation, growth, and differentiation, and is stimulated by various energy sources and hormonal signaling pathways. Pathogenic variants in TSC1 and TSC2 lead to mTORC1 hyperactivation, producing benign tumours in multiple organs, including the brain and kidneys, and drug-resistant epilepsy, a typical sign of TSC. Brain tumours, sudden unexpected death from epilepsy, and respiratory conditions are the three leading causes of morbidity and mortality. Even though several therapeutic options are available for the treatment of TSC, there is further need for a better understanding of the pathophysiological basis of the neurologic and other manifestations seen in TSC, and for novel therapeutic approaches. This review provides an overview of the main current therapies for TSC and discusses recent studies highlighting the repurposing of approved drugs and the emerging role of novel targets for future drug design.
Collapse
Affiliation(s)
- Elena Conte
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (B.B.); (G.D.); (A.D.L.); (A.L.)
| | - Brigida Boccanegra
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (B.B.); (G.D.); (A.D.L.); (A.L.)
| | - Giorgia Dinoi
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (B.B.); (G.D.); (A.D.L.); (A.L.)
| | - Michael Pusch
- Institute of Biophysics, National Research Council, 16149 Genova, Italy;
| | - Annamaria De Luca
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (B.B.); (G.D.); (A.D.L.); (A.L.)
| | - Antonella Liantonio
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (B.B.); (G.D.); (A.D.L.); (A.L.)
| | - Paola Imbrici
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (B.B.); (G.D.); (A.D.L.); (A.L.)
| |
Collapse
|
32
|
Sophronea T, Agrawal S, Kumari N, Mishra J, Walecha V, Luthra PM. A 2AR antagonists triggered the AMPK/m-TOR autophagic pathway to reverse the calcium-dependent cell damage in 6-OHDA induced model of PD. Neurochem Int 2024; 178:105793. [PMID: 38880232 DOI: 10.1016/j.neuint.2024.105793] [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: 02/14/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Calcium dyshomeostasis, oxidative stress, autophagy and apoptosis are the pathogenesis of selective dopaminergic neuronal loss in Parkinson's disease (PD). Earlier, we reported that A2A R modulates IP3-dependent intracellular Ca2+ signalling via PKA. Moreover, A2A R antagonist has been reported to reduce oxidative stress and apoptosis in PD models, however intracellular Ca2+ ([Ca2+]i) dependent autophagy regulation in the 6-OHDA model of PD has not been explored. In the present study, we investigated the A2A R antagonists mediated neuroprotective effects in 6-OHDA-induced primary midbrain neuronal (PMN) cells and unilateral lesioned rat model of PD. 6-OHDA-induced oxidative stress (ROS and superoxide) and [Ca2+]i was measured using Fluo4AM, DCFDA and DHE dye respectively. Furthermore, autophagy was assessed by Western blot of p-m-TOR/mTOR, p-AMPK/AMPK, LC3I/II, Beclin and β-actin. Apoptosis was measured by Annexin V-APC-PI detection and Western blot of Bcl2, Bax, caspase3 and β-actin. Dopamine levels were measured by Dopamine ELISA kit and Western blot of tyrosine hydroxylase. Our results suggest that 6-OHDA-induced PMN cell death occurred due to the interruption of [Ca2+]i homeostasis, accompanied by activation of autophagy and apoptosis. A2A R antagonists prevented 6-OHDA-induced neuronal cell death by decreasing [Ca2+]i overload and oxidative stress. In addition, we found that A2A R antagonists upregulated mTOR phosphorylation and downregulated AMPK phosphorylation thereby reducing autophagy and apoptosis both in 6-OHDA induced PMN cells and 6-OHDA unilateral lesioned rat model. In conclusion, A2A R antagonists alleviated 6-OHDA toxicity by modulating [Ca2+]i signalling to inhibit autophagy mediated by the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Tuithung Sophronea
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Saurabh Agrawal
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Namrata Kumari
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Jyoti Mishra
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Vaishali Walecha
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Pratibha Mehta Luthra
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
33
|
Hou J, Mei K, Wang D, Ke S, Chen X, Shang J, Li G, Gao Y, Xiong H, Zhang H, Chen L, Zhang W, Deng Y, Hong X, Liu DA, Hu T, Guo W, Zhan YY. TGM1/3-mediated transamidation of Exo70 promotes tumor metastasis upon LKB1 inactivation. Cell Rep 2024; 43:114604. [PMID: 39146185 DOI: 10.1016/j.celrep.2024.114604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 05/10/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024] Open
Abstract
Exo70, a key exocyst complex component, is crucial for cell motility and extracellular matrix (ECM) remodeling in cancer metastasis. Despite its potential as a drug target, Exo70's post-translational modifications (PTMs) are poorly characterized. Here, we report that Exo70 is transamidated on Gln5 with Lys56 of cystatin A by transglutaminases TGM1 and TGM3, promoting tumor metastasis. This modification enhances Exo70's association with other exocyst subunits, essential for secreting matrix metalloproteinases, forming invadopodia, and delivering integrins to the leading edge. Tumor suppressor liver kinase B1 (LKB1), whose inactivation accelerates metastasis, phosphorylates TGM1 and TGM3 at Thr386 and Thr282, respectively, to inhibit their interaction with Exo70 and the following transamidation. Cantharidin, a US Food and Drug Administration (FDA)-approved drug, inhibits Exo70 transamidation to restrain tumor cell migration and invasion. Together, our findings highlight Exo70 transamidation as a key molecular mechanism and target and propose cantharidin as a therapeutic strategy with direct clinical translational value for metastatic cancers, especially those with LKB1 loss.
Collapse
Affiliation(s)
- Jihuan Hou
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Kunrong Mei
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Daxuan Wang
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Sunkui Ke
- Department of Thoracic Surgery, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361004, China
| | - Xiong Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jin Shang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Guixia Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Huifang Xiong
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Haoran Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Lu Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Wenqing Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yabin Deng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiaoting Hong
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Di-Ao Liu
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tianhui Hu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yan-Yan Zhan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
34
|
Khondker S, Han GS, Carman GM. Protein kinase Hsl1 phosphorylates Pah1 to inhibit phosphatidate phosphatase activity and regulate lipid synthesis in Saccharomyces cerevisiae. J Biol Chem 2024; 300:107572. [PMID: 39009344 PMCID: PMC11342776 DOI: 10.1016/j.jbc.2024.107572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024] Open
Abstract
In Saccharomyces cerevisiae, Pah1 phosphatidate (PA) phosphatase, which catalyzes the Mg2+-dependent dephosphorylation of PA to produce diacylglycerol, plays a key role in utilizing PA for the synthesis of the neutral lipid triacylglycerol and thereby controlling the PA-derived membrane phospholipids. The enzyme function is controlled by its subcellular location as regulated by phosphorylation and dephosphorylation. Pah1 is initially inactivated in the cytosol through phosphorylation by multiple protein kinases and then activated via its recruitment and dephosphorylation by the protein phosphatase Nem1-Spo7 at the nuclear/endoplasmic reticulum membrane where the PA phosphatase reaction occurs. Many of the protein kinases that phosphorylate Pah1 have yet to be characterized with the identification of the target residues. Here, we established Pah1 as a bona fide substrate of septin-associated Hsl1, a protein kinase involved in mitotic morphogenesis checkpoint signaling. The Hsl1 activity on Pah1 was dependent on reaction time and the amounts of protein kinase, Pah1, and ATP. The Hsl1 phosphorylation of Pah1 occurred on Ser-748 and Ser-773, and the phosphorylated protein exhibited a 5-fold reduction in PA phosphatase catalytic efficiency. Analysis of cells expressing the S748A and S773A mutant forms of Pah1 indicated that Hsl1-mediated phosphorylation of Pah1 promotes membrane phospholipid synthesis at the expense of triacylglycerol, and ensures the dependence of Pah1 function on the Nem1-Spo7 protein phosphatase. This work advances the understanding of how Hsl1 facilitates membrane phospholipid synthesis through the phosphorylation-mediated regulation of Pah1.
Collapse
Affiliation(s)
- Shoily Khondker
- Department of Food Science and the Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA
| | - Gil-Soo Han
- Department of Food Science and the Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA
| | - George M Carman
- Department of Food Science and the Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey, USA.
| |
Collapse
|
35
|
Cao L, Lin G, Fan D, Weng K, Chen Y, Wang J, Li P, Zheng C, Huang C, Xie J. NUAK1 activates STAT5/GLI1/SOX2 signaling to enhance cancer cell expansion and drives chemoresistance in gastric cancer. Cell Rep 2024; 43:114446. [PMID: 38996065 DOI: 10.1016/j.celrep.2024.114446] [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: 09/12/2023] [Revised: 06/02/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
The gene encoding the NUAK family kinase 1 (NUAK1) is frequently amplified and its expression is upregulated, activating oncogenic signaling in various cancers. However, little is known about its role in gastric cancer (GC). We investigate the mechanistic links among NUAK1, Hedgehog signaling, and tumorigenesis in GC. NUAK1 overexpression is validated in local and public GC cohorts. Patient-derived xenograft and transgenic mouse models demonstrate that NUAK1 depletion or inhibition dramatically ameliorates gastric tumorigenesis. NUAK1 upregulates GLI1 expression by activating STAT5-mediated transcription and stabilizing GLI1 protein. NUAK1 depletion or inhibition impairs cancer cell expansion, tumor formation, and chemotherapy resistance in in vitro and in vivo models. Clinicopathological analysis confirms that upregulated NUAK1 expression correlates with poor prognosis and chemotherapy resistance in human GC. Our findings demonstrate that the signaling axis NUAK1/STAT5/GLI1 promotes cancer cell expansion and tumorigenesis and indicate that NUAK1 is an attractive therapeutic target and prognostic factor in GC.
Collapse
Affiliation(s)
- Longlong Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| | - Guangtan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Denghui Fan
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Yujing Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiabin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China
| | - Changming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
36
|
Zhang W, Ni Y, Li J, Hua R, Wang Y, Yang H, Li X, Gan M, Chu G. NUAK2 mediated regulation of Schwann Cell proliferation and migration in peripheral nerve injury via YAP. Heliyon 2024; 10:e34127. [PMID: 39071701 PMCID: PMC11282989 DOI: 10.1016/j.heliyon.2024.e34127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/13/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
NUAK2 is a member of the AMP-activated protein kinase (AMPK) family, which plays an essential role in cellular processes such as apoptosis, proliferation, and cell fate. Recent studies have already shown that silencing of NUAK2 blocks proliferation and promotes apoptosis of human melanoma cells and liver cancer cells. In addition, NUAK2 is involved in the development of glioblastoma via regulating the expression of cancer stem cell-related genes, and it promotes the cell cycle entry in the glioblastoma cells. However, the expression and the role of NUAK2 in the progress of peripheral nerve regeneration after injury are yet to be elucidated. We observed that NUAK2 was upregulated following distal sciatic nerve crush (SNC). Interestingly, we discovered that NUAK2 showed co-localization with S100 (Schwann cell marker). Furthermore, we found that the NUAK2 had a spatiotemporal protein expression, which was consistent with proliferating cell nuclear-antigen (PCNA). The protein level of NUAK2 and YAP was upregulated in the model of TNF-α-induced Schwann cell (SC) proliferation. Furthermore, flow cytometry analysis, CCK-8, transwell assays, and wound healing assays were all performed with the purpose of exploring the role of NUAK2 in the regulation of SC proliferation and migration. More importantly, we found that NUAK2-deficient SCs showed significantly reduced expression of Yes-associated protein (YAP). Bioinformatic analysis identified upstream regulators of NUAK2 and NUAK2-associated genes (e.g., YAP1). Finally, we investigated the recovery changes during regeneration progress through the walking track analysis. Thus, we speculated that NUAK2 was involved in biochemical and physiological responses of SCs after SNC via YAP-driven proliferation and migration, and this study determined the importance of NUAK2 as a potential target in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yingchen Ni
- Department of Orthopaedic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jianxin Li
- Department of Orthopaedic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Runjia Hua
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yudong Wang
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xuefeng Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Minfeng Gan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Genglei Chu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
37
|
Chen Y, Lee K, Woo J, Kim DW, Keum C, Babbi G, Casadio R, Martelli PL, Savojardo C, Manfredi M, Shen Y, Sun Y, Katsonis P, Lichtarge O, Pejaver V, Seward DJ, Kamandula A, Bakolitsa C, Brenner SE, Radivojac P, O’Donnell-Luria A, Mooney SD, Jain S. Evaluating predictors of kinase activity of STK11 variants identified in primary human non-small cell lung cancers. RESEARCH SQUARE 2024:rs.3.rs-4587317. [PMID: 39011112 PMCID: PMC11247923 DOI: 10.21203/rs.3.rs-4587317/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Critical evaluation of computational tools for predicting variant effects is important considering their increased use in disease diagnosis and driving molecular discoveries. In the sixth edition of the Critical Assessment of Genome Interpretation (CAGI) challenge, a dataset of 28 STK11 rare variants (27 missense, 1 single amino acid deletion), identified in primary non-small cell lung cancer biopsies, was experimentally assayed to characterize computational methods from four participating teams and five publicly available tools. Predictors demonstrated a high level of performance on key evaluation metrics, measuring correlation with the assay outputs and separating loss-of-function (LoF) variants from wildtype-like (WT-like) variants. The best participant model, 3Cnet, performed competitively with well-known tools. Unique to this challenge was that the functional data was generated with both biological and technical replicates, thus allowing the assessors to realistically establish maximum predictive performance based on experimental variability. Three out of the five publicly available tools and 3Cnet approached the performance of the assay replicates in separating LoF variants from WT-like variants. Surprisingly, REVEL, an often-used model, achieved a comparable correlation with the real-valued assay output as that seen for the experimental replicates. Performing variant interpretation by combining the new functional evidence with computational and population data evidence led to 16 new variants receiving a clinically actionable classification of likely pathogenic (LP) or likely benign (LB). Overall, the STK11 challenge highlights the utility of variant effect predictors in biomedical sciences and provides encouraging results for driving research in the field of computational genome interpretation.
Collapse
Affiliation(s)
- Yile Chen
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, 98105, WA, USA
| | - Kyoungyeul Lee
- 3billion, 3billion Biotechnology company, Seoul, South Korea
| | - Junwoo Woo
- 3billion, 3billion Biotechnology company, Seoul, South Korea
| | - Dong-wook Kim
- 3billion, 3billion Biotechnology company, Seoul, South Korea
| | - Changwon Keum
- 3billion, 3billion Biotechnology company, Seoul, South Korea
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Matteo Manfredi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, 77843, TX, USA
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, 77843, TX, USA
| | - Panagiotis Katsonis
- Molecular and Human Genetics, Baylor College of Medicine, Houston, 77030, TX, USA
| | - Olivier Lichtarge
- Molecular and Human Genetics, Baylor College of Medicine, Houston, 77030, TX, USA
| | - Vikas Pejaver
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - David J. Seward
- Department of Pathology, University of Vermont, Burlington, 5445, VT, USA
| | - Akash Kamandula
- Khoury College of Computer Sciences, Northeastern University, Boston, 02115, MA, USA
| | | | | | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, 02115, MA, USA
| | - Anne O’Donnell-Luria
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, 02115, MA, USA
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, 02142, MA, USA
| | - Sean D. Mooney
- Center for Information Technology, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Shantanu Jain
- Khoury College of Computer Sciences, Northeastern University, Boston, 02115, MA, USA
- The Institute for Experiential AI, Northeastern University, Boston, 02115, MA, USA
| |
Collapse
|
38
|
Debets DO, de Graaf EL, Liefaard MC, Sonke GS, Lips EH, Ressa A, Altelaar M. Predicting treatment outcome using kinome activity profiling in HER2+ breast cancer biopsies. iScience 2024; 27:109858. [PMID: 38784015 PMCID: PMC11112361 DOI: 10.1016/j.isci.2024.109858] [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: 03/31/2023] [Revised: 08/29/2023] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
In this study, we measured the kinase activity profiles of 32 pre-treatment tumor biopsies of HER2-positive breast cancer patients. The aim of this study was to assess the prognostic potential of kinase activity levels, to identify potential mechanisms of resistance and to predict treatment success of HER2-targeted therapy combined with chemotherapy. Indeed, our system-wide kinase activity analysis allowed us to link kinase activity to treatment response. Overall, high kinase activity in the HER2-pathway was associated with good treatment outcome. We found eleven kinases differentially regulated between treatment outcome groups, including well-known players in therapy resistance, such as p38a, ERK, and FAK, and an unreported one, namely MARK1. Lastly, we defined an optimal signature of four kinases in a multiple logistic regression diagnostic test for prediction of treatment outcome (AUC = 0.926). This kinase signature showed high sensitivity and specificity, indicating its potential as predictive biomarker for treatment success of HER2-targeted therapy.
Collapse
Affiliation(s)
- Donna O. Debets
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, 3584 CH Utrecht, the Netherlands
| | - Erik L. de Graaf
- Pepscope B.V, Nieuwe Kanaal 7, 6709 PA Wageningen, the Netherlands
| | - Marte C. Liefaard
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Gabe S. Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, University of Amsterdam, Amsterdam, the Netherlands
| | - Esther H. Lips
- Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anna Ressa
- Pepscope B.V, Nieuwe Kanaal 7, 6709 PA Wageningen, the Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, 3584 CH Utrecht, the Netherlands
| |
Collapse
|
39
|
Sun J, Zhang Y, Zhang Q, Hu L, Zhao L, Wang H, Yuan Y, Niu H, Wang D, Zhang H, Liu J, Feng X, Su X, Qiu J, Sun J, Xu H, Zhang C, Wang K, Bi Y, Engleman EG, Shen L. Metabolic regulator LKB1 controls adipose tissue ILC2 PD-1 expression and mitochondrial homeostasis to prevent insulin resistance. Immunity 2024; 57:1289-1305.e9. [PMID: 38772366 DOI: 10.1016/j.immuni.2024.04.024] [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: 08/18/2023] [Revised: 02/06/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Jiping Sun
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youqin Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qingbing Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Hu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Linfeng Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Yue Yuan
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Hongshen Niu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dongdi Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huasheng Zhang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianyue Liu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xujiao Feng
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaohui Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Catherine Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Kathleen Wang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Lei Shen
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
40
|
Trelford CB, Shepherd TG. LKB1 biology: assessing the therapeutic relevancy of LKB1 inhibitors. Cell Commun Signal 2024; 22:310. [PMID: 38844908 PMCID: PMC11155146 DOI: 10.1186/s12964-024-01689-5] [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: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Liver Kinase B1 (LKB1), encoded by Serine-Threonine Kinase 11 (STK11), is a master kinase that regulates cell migration, polarity, proliferation, and metabolism through downstream adenosine monophosphate-activated protein kinase (AMPK) and AMPK-related kinase signalling. Since genetic screens identified STK11 mutations in Peutz-Jeghers Syndrome, STK11 mutants have been implicated in tumourigenesis labelling it as a tumour suppressor. In support of this, several compounds reduce tumour burden through upregulating LKB1 signalling, and LKB1-AMPK agonists are cytotoxic to tumour cells. However, in certain contexts, its role in cancer is paradoxical as LKB1 promotes tumour cell survival by mediating resistance against metabolic and oxidative stressors. LKB1 deficiency has also enhanced the selectivity and cytotoxicity of several cancer therapies. Taken together, there is a need to develop LKB1-specific pharmacological compounds, but prior to developing LKB1 inhibitors, further work is needed to understand LKB1 activity and regulation. However, investigating LKB1 activity is strenuous as cell/tissue type, mutations to the LKB1 signalling pathway, STE-20-related kinase adaptor protein (STRAD) binding, Mouse protein 25-STRAD binding, splicing variants, nucleocytoplasmic shuttling, post-translational modifications, and kinase conformation impact the functional status of LKB1. For these reasons, guidelines to standardize experimental strategies to study LKB1 activity, associate proteins, spliced isoforms, post-translational modifications, and regulation are of upmost importance to the development of LKB1-specific therapies. Therefore, to assess the therapeutic relevancy of LKB1 inhibitors, this review summarizes the importance of LKB1 in cell physiology, highlights contributors to LKB1 activation, and outlines the benefits and risks associated with targeting LKB1.
Collapse
Affiliation(s)
- Charles B Trelford
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada.
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Trevor G Shepherd
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
41
|
Teuwen JTJ, van der Vorst EPC, Maas SL. Navigating the Maze of Kinases: CaMK-like Family Protein Kinases and Their Role in Atherosclerosis. Int J Mol Sci 2024; 25:6213. [PMID: 38892400 PMCID: PMC11172518 DOI: 10.3390/ijms25116213] [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: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Circulating low-density lipoprotein (LDL) levels are a major risk factor for cardiovascular diseases (CVD), and even though current treatment strategies focusing on lowering lipid levels are effective, CVD remains the primary cause of death worldwide. Atherosclerosis is the major cause of CVD and is a chronic inflammatory condition in which various cell types and protein kinases play a crucial role. However, the underlying mechanisms of atherosclerosis are not entirely understood yet. Notably, protein kinases are highly druggable targets and represent, therefore, a novel way to target atherosclerosis. In this review, the potential role of the calcium/calmodulin-dependent protein kinase-like (CaMKL) family and its role in atherosclerosis will be discussed. This family consists of 12 subfamilies, among which are the well-described and conserved liver kinase B1 (LKB1) and 5' adenosine monophosphate-activated protein kinase (AMPK) subfamilies. Interestingly, LKB1 plays a key role and is considered a master kinase within the CaMKL family. It has been shown that LKB1 signaling leads to atheroprotective effects, while, for example, members of the microtubule affinity-regulating kinase (MARK) subfamily have been described to aggravate atherosclerosis development. These observations highlight the importance of studying kinases and their signaling pathways in atherosclerosis, bringing us a step closer to unraveling the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Jules T. J. Teuwen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80336 München, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
42
|
Wang HC, Li L, Zhang JH, Yao ZH, Pang BP. MicroRNA miR-7-5p targets MARK2 to control metamorphosis in Galeruca daurica. Comp Biochem Physiol B Biochem Mol Biol 2024; 272:110967. [PMID: 38521445 DOI: 10.1016/j.cbpb.2024.110967] [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: 01/16/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
The MARK2 gene, coding microtubule affinity-regulating kinase or serine/threonine protein kinase, is an important modulator in organism microtubule generation and cell polarity. However, its role in the metamorphosis of insects remains unknown. In this study, we found a conserved miRNA, miR-7-5p, which targets MARK2 to participate in the regulation of the larval-pupal metamorphosis in Galeruca daurica. The dual luciferase reporter assay showed that miR-7-5p interacted with the 3' UTR of MARK2 and repressed its expression. The expression profiling of miR-7-5p and MARK2 displayed an opposite trend during the larval-adult development process. In in-vivo experiments, overexpression of miR-7-5p by injecting miR-7-5p agomir in the final instar larvae down-regulated MARK2 and up-regulated main ecdysone signaling pathway genes including E74, E75, ECR, FTZ-F1 and HR3, which was similar to the results from knockdown of MARK2 by RNAi. In contrast, repression of miR-7-5p by injecting miR-7-5p antagomir obtained opposite effects. Notably, both overexpression and repression of miR-7-5p in the final instar larvae caused abnormal molting and high mortality during the larval-pupal transition, and high mortality during the pupal-adult transition. The 20-hydroxyecdysone (20E) injection experiment showed that 20E up-regulated miR-7-5p whereas down-regulated MARK2. This study reveals that the accurate regulation of miRNAs and their target genes is indispensable for insect metamorphosis.
Collapse
Affiliation(s)
- Hai-Chao Wang
- Research Center for Grassland Entomology, Inner Mongolia Agricultural University, Hohhot, China
| | - Ling Li
- Research Center for Grassland Entomology, Inner Mongolia Agricultural University, Hohhot, China
| | - Jing-Hang Zhang
- Inner Mongolia Center for Plant Protection and Quarantine, Hohhot, China
| | - Zhi-Han Yao
- Research Center for Grassland Entomology, Inner Mongolia Agricultural University, Hohhot, China
| | - Bao-Ping Pang
- Research Center for Grassland Entomology, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
43
|
Langer HT, Rohm M, Goncalves MD, Sylow L. AMPK as a mediator of tissue preservation: time for a shift in dogma? Nat Rev Endocrinol 2024:10.1038/s41574-024-00992-y. [PMID: 38760482 DOI: 10.1038/s41574-024-00992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Ground-breaking discoveries have established 5'-AMP-activated protein kinase (AMPK) as a central sensor of metabolic stress in cells and tissues. AMPK is activated through cellular starvation, exercise and drugs by either directly or indirectly affecting the intracellular AMP (or ADP) to ATP ratio. In turn, AMPK regulates multiple processes of cell metabolism, such as the maintenance of cellular ATP levels, via the regulation of fatty acid oxidation, glucose uptake, glycolysis, autophagy, mitochondrial biogenesis and degradation, and insulin sensitivity. Moreover, AMPK inhibits anabolic processes, such as lipogenesis and protein synthesis. These findings support the notion that AMPK is a crucial regulator of cell catabolism. However, studies have revealed that AMPK's role in cell homeostasis might not be as unidirectional as originally thought. This Review explores emerging evidence for AMPK as a promoter of cell survival and an enhancer of anabolic capacity in skeletal muscle and adipose tissue during catabolic crises. We discuss AMPK-activating interventions for tissue preservation during tissue wasting in cancer-associated cachexia and explore the clinical potential of AMPK activation in wasting conditions. Overall, we provide arguments that call for a shift in the current dogma of AMPK as a mere regulator of cell catabolism, concluding that AMPK has an unexpected role in tissue preservation.
Collapse
Affiliation(s)
- Henning Tim Langer
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riβ, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus DaSilva Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lykke Sylow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
Cherfan C, Chebly A, Rezvani HR, Beylot-Barry M, Chevret E. Delving into the Metabolism of Sézary Cells: A Brief Review. Genes (Basel) 2024; 15:635. [PMID: 38790264 PMCID: PMC11121102 DOI: 10.3390/genes15050635] [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: 03/29/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Primary cutaneous lymphomas (PCLs) are a heterogeneous group of lymphoproliferative disorders caused by the accumulation of neoplastic T or B lymphocytes in the skin. Sézary syndrome (SS) is an aggressive and rare form of cutaneous T cell lymphoma (CTCL) characterized by an erythroderma and the presence of atypical cerebriform T cells named Sézary cells in skin and blood. Most of the available treatments for SS are not curative, which means there is an urgent need for the development of novel efficient therapies. Recently, targeting cancer metabolism has emerged as a promising strategy for cancer therapy. This is due to the accumulating evidence that metabolic reprogramming highly contributes to tumor progression. Genes play a pivotal role in regulating metabolic processes, and alterations in these genes can disrupt the delicate balance of metabolic pathways, potentially contributing to cancer development. In this review, we discuss the importance of targeting energy metabolism in tumors and the currently available data on the metabolism of Sézary cells, paving the way for potential new therapeutic approaches aiming to improve clinical outcomes for patients suffering from SS.
Collapse
Affiliation(s)
- Carel Cherfan
- BRIC, BoRdeaux Institute of onCology, UMR 1312, Inserm, Université de Bordeaux, 33000 Bordeaux, France; (C.C.); (H.R.R.); (M.B.-B.)
| | - Alain Chebly
- Center Jacques Loiselet for Medical Genetics and Genomics (CGGM), Faculty of Medicine, Saint Joseph University, Beirut P.O. Box 17-5208, Lebanon;
| | - Hamid Reza Rezvani
- BRIC, BoRdeaux Institute of onCology, UMR 1312, Inserm, Université de Bordeaux, 33000 Bordeaux, France; (C.C.); (H.R.R.); (M.B.-B.)
| | - Marie Beylot-Barry
- BRIC, BoRdeaux Institute of onCology, UMR 1312, Inserm, Université de Bordeaux, 33000 Bordeaux, France; (C.C.); (H.R.R.); (M.B.-B.)
- Dermatology Department, Centre Hospitalier Universitaire de Bordeaux, 33075 Bordeaux, France
| | - Edith Chevret
- BRIC, BoRdeaux Institute of onCology, UMR 1312, Inserm, Université de Bordeaux, 33000 Bordeaux, France; (C.C.); (H.R.R.); (M.B.-B.)
| |
Collapse
|
45
|
Haberman N, Cheung R, Pizza G, Cvetesic N, Nagy D, Maude H, Blazquez L, Lenhard B, Cebola I, Rutter GA, Martinez-Sanchez A. Liver kinase B1 (LKB1) regulates the epigenetic landscape of mouse pancreatic beta cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593867. [PMID: 38798508 PMCID: PMC11118353 DOI: 10.1101/2024.05.13.593867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Liver kinase B1 (LKB1/STK11) is an important regulator of pancreatic β-cell identity and function. Elimination of Lkb1 from the β-cell results in improved glucose-stimulated insulin secretion and is accompanied by profound changes in gene expression, including the upregulation of several neuronal genes. The mechanisms through which LKB1 controls gene expression are, at present, poorly understood. Here, we explore the impact of β cell- selective deletion of Lkb1 on chromatin accessibility in mouse pancreatic islets. To characterize the role of LKB1 in the regulation of gene expression at the transcriptional level, we combine these data with a map of islet active transcription start sites and histone marks. We demonstrate that LKB1 elimination from β-cells results in widespread changes in chromatin accessibility, correlating with changes in transcript levels. Changes occurred in hundreds of promoter and enhancer regions, many of which were close to neuronal genes. We reveal that dysregulated enhancers are enriched in binding motifs for transcription factors important for β-cell identity, such as FOXA, MAFA or RFX6 and we identify microRNAs (miRNAs) that are regulated by LKB1 at the transcriptional level. Overall, our study provides important new insights into the epigenetic mechanisms by which LKB1 regulates β-cell identity and function.
Collapse
|
46
|
Lagoudaki ED, Koutsopoulos AV, Sfakianaki M, Papadaki C, Manikis GC, Voutsina A, Trypaki M, Tsakalaki E, Fiolitaki G, Hatzidaki D, Yiachnakis E, Koumaki D, Mavroudis D, Tzardi M, Stathopoulos EN, Marias K, Georgoulias V, Souglakos J. LKB1 Loss Correlates with STING Loss and, in Cooperation with β-Catenin Membranous Loss, Indicates Poor Prognosis in Patients with Operable Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:1818. [PMID: 38791897 PMCID: PMC11120022 DOI: 10.3390/cancers16101818] [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: 04/07/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
To investigate the incidence and prognostically significant correlations and cooperations of LKB1 loss of expression in non-small cell lung cancer (NSCLC), surgical specimens from 188 metastatic and 60 non-metastatic operable stage I-IIIA NSCLC patients were analyzed to evaluate their expression of LKB1 and pAMPK proteins in relation to various processes. The investigated factors included antitumor immunity response regulators STING and PD-L1; pro-angiogenic, EMT and cell cycle targets, as well as metastasis-related (VEGFC, PDGFRα, PDGFRβ, p53, p16, Cyclin D1, ZEB1, CD24) targets; and cell adhesion (β-catenin) molecules. The protein expression levels were evaluated via immunohistochemistry; the RNA levels of LKB1 and NEDD9 were evaluated via PCR, while KRAS exon 2 and BRAFV600E mutations were evaluated by Sanger sequencing. Overall, loss of LKB1 protein expression was observed in 21% (51/248) patients and correlated significantly with histotype (p < 0.001), KRAS mutations (p < 0.001), KC status (concomitant KRAS mutation and p16 downregulation) (p < 0.001), STING loss (p < 0.001), and high CD24 expression (p < 0.001). STING loss also correlated significantly with loss of LKB1 expression in the metastatic setting both overall (p = 0.014) and in lung adenocarcinomas (LUACs) (p = 0.005). Additionally, LKB1 loss correlated significantly with a lack of or low β-catenin membranous expression exclusively in LUACs, both independently of the metastatic status (p = 0.019) and in the metastatic setting (p = 0.007). Patients with tumors yielding LKB1 loss and concomitant nonexistent or low β-catenin membrane expression experienced significantly inferior median overall survival of 20.50 vs. 52.99 months; p < 0.001 as well as significantly greater risk of death (HR: 3.32, 95% c.i.: 1.71-6.43; p <0.001). Our findings underscore the impact of the synergy of LKB1 with STING and β-catenin in NSCLC, in prognosis.
Collapse
Affiliation(s)
- Eleni D. Lagoudaki
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Anastasios V. Koutsopoulos
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Chara Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Georgios C. Manikis
- Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece; (G.C.M.); (K.M.)
| | - Alexandra Voutsina
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Maria Trypaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Eleftheria Tsakalaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Georgia Fiolitaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Dora Hatzidaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Emmanuel Yiachnakis
- Laboratory of Bio-Medical Data Analysis Digital Applications and Interdisciplinary Approaches, University of Crete, 71003 Heraklion, Greece;
| | - Dimitra Koumaki
- Department of Dermatology, University General Hospital of Heraklion, Voutes, 71500 Heraklion, Greece;
| | - Dimitrios Mavroudis
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - Maria Tzardi
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Efstathios N. Stathopoulos
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Kostas Marias
- Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece; (G.C.M.); (K.M.)
| | - Vassilis Georgoulias
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - John Souglakos
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| |
Collapse
|
47
|
Priyanka P, Gopalakrishnan AP, Nisar M, Shivamurthy PB, George M, John L, Sanjeev D, Yandigeri T, Thomas SD, Rafi A, Dagamajalu S, Velikkakath AKG, Abhinand CS, Kanekar S, Prasad TSK, Balaya RDA, Raju R. A global phosphosite-correlated network map of Thousand And One Kinase 1 (TAOK1). Int J Biochem Cell Biol 2024; 170:106558. [PMID: 38479581 DOI: 10.1016/j.biocel.2024.106558] [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: 08/12/2023] [Revised: 02/19/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Abstract
Thousand and one amino acid kinase 1 (TAOK1) is a sterile 20 family Serine/Threonine kinase linked to microtubule dynamics, checkpoint signaling, DNA damage response, and neurological functions. Molecular-level alterations of TAOK1 have been associated with neurodevelopment disorders and cancers. Despite their known involvement in physiological and pathophysiological processes, and as a core member of the hippo signaling pathway, the phosphoregulatory network of TAOK1 has not been visualized. Aimed to explore this network, we first analyzed the predominantly detected and differentially regulated TAOK1 phosphosites in global phosphoproteome datasets across diverse experimental conditions. Based on 709 qualitative and 210 quantitative differential cellular phosphoproteome datasets that were systematically assembled, we identified that phosphorylation at Ser421, Ser9, Ser965, and Ser445 predominantly represented TAOK1 in almost 75% of these datasets. Surprisingly, the functional role of all these phosphosites in TAOK1 remains unexplored. Hence, we employed a robust strategy to extract the phosphosites in proteins that significantly correlated in expression with predominant TAOK1 phosphosites. This led to the first categorization of the phosphosites including those in the currently known and predicted interactors, kinases, and substrates, that positively/negatively correlated with the expression status of each predominant TAOK1 phosphosites. Subsequently, we also analyzed the phosphosites in core proteins of the hippo signaling pathway. Based on the TAOK1 phosphoregulatory network analysis, we inferred the potential role of the predominant TAOK1 phosphosites. Especially, we propose pSer9 as an autophosphorylation and TAOK1 kinase activity-associated phosphosite and pS421, the most frequently detected phosphosite in TAOK1, as a significant regulatory phosphosite involved in the maintenance of genome integrity. Considering that the impact of all phosphosites that predominantly represent each kinase is essential for the efficient interpretation of global phosphoproteome datasets, we believe that the approach undertaken in this study is suitable to be extended to other kinases for accelerated research.
Collapse
Affiliation(s)
- Pahal Priyanka
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Athira Perunelly Gopalakrishnan
- Center for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Mahammad Nisar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | | | - Mejo George
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Levin John
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Diya Sanjeev
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Tanuja Yandigeri
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Sonet D Thomas
- Center for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Ahmad Rafi
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Shobha Dagamajalu
- Center for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Anoop Kumar G Velikkakath
- Center for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Saptami Kanekar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| | | | | | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore 575018, India.
| |
Collapse
|
48
|
Ma S, Ming Y, Wu J, Cui G. Cellular metabolism regulates the differentiation and function of T-cell subsets. Cell Mol Immunol 2024; 21:419-435. [PMID: 38565887 PMCID: PMC11061161 DOI: 10.1038/s41423-024-01148-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
T cells are an important component of adaptive immunity and protect the host from infectious diseases and cancers. However, uncontrolled T cell immunity may cause autoimmune disorders. In both situations, antigen-specific T cells undergo clonal expansion upon the engagement and activation of antigens. Cellular metabolism is reprogrammed to meet the increase in bioenergetic and biosynthetic demands associated with effector T cell expansion. Metabolites not only serve as building blocks or energy sources to fuel cell growth and expansion but also regulate a broad spectrum of cellular signals that instruct the differentiation of multiple T cell subsets. The realm of immunometabolism research is undergoing swift advancements. Encapsulating all the recent progress within this concise review in not possible. Instead, our objective is to provide a succinct introduction to this swiftly progressing research, concentrating on the metabolic intricacies of three pivotal nutrient classes-lipids, glucose, and amino acids-in T cells. We shed light on recent investigations elucidating the roles of these three groups of metabolites in mediating the metabolic and immune functions of T cells. Moreover, we delve into the prospect of "editing" metabolic pathways within T cells using pharmacological or genetic approaches, with the aim of synergizing this approach with existing immunotherapies and enhancing the efficacy of antitumor and antiinfection immune responses.
Collapse
Affiliation(s)
- Sicong Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Yanan Ming
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Jingxia Wu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| | - Guoliang Cui
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| |
Collapse
|
49
|
Xu Z, Davies ER, Yao L, Zhou Y, Li J, Alzetani A, Marshall BG, Hancock D, Wallis T, Downward J, Ewing RM, Davies DE, Jones MG, Wang Y. LKB1 depletion-mediated epithelial-mesenchymal transition induces fibroblast activation in lung fibrosis. Genes Dis 2024; 11:101065. [PMID: 38222900 PMCID: PMC7615521 DOI: 10.1016/j.gendis.2023.06.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/29/2023] [Accepted: 06/28/2023] [Indexed: 01/16/2024] Open
Abstract
The factors that determine fibrosis progression or normal tissue repair are largely unknown. We previously demonstrated that autophagy inhibition-mediated epithelial-mesenchymal transition (EMT) in human alveolar epithelial type II (ATII) cells augments local myofibroblast differentiation in pulmonary fibrosis by paracrine signalling. Here, we report that liver kinase B1 (LKB1) inactivation in ATII cells inhibits autophagy and induces EMT as a consequence. In IPF lungs, this is caused by downregulation of CAB39L, a key subunit within the LKB1 complex. 3D co-cultures of ATII cells and MRC5 lung fibroblasts coupled with RNA sequencing (RNA-seq) confirmed that paracrine signalling between LKB1-depleted ATII cells and fibroblasts augmented myofibroblast differentiation. Together these data suggest that reduced autophagy caused by LKB1 inhibition can induce EMT in ATII cells and contribute to fibrosis via aberrant epithelial-fibroblast crosstalk.
Collapse
Affiliation(s)
- Zijian Xu
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Elizabeth R. Davies
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Liudi Yao
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Yilu Zhou
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Juanjuan Li
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Aiman Alzetani
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
- University Hospital Southampton, Southampton SO16 6YD, UK
| | - Ben G. Marshall
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
- University Hospital Southampton, Southampton SO16 6YD, UK
| | - David Hancock
- Oncogene Biology, The Francis Crick Institute, London NW1 1AT, UK
| | - Tim Wallis
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
- University Hospital Southampton, Southampton SO16 6YD, UK
| | - Julian Downward
- Oncogene Biology, The Francis Crick Institute, London NW1 1AT, UK
| | - Rob M. Ewing
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Donna E. Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
50
|
Bendzunas GN, Byrne DP, Shrestha S, Daly LA, Oswald SO, Katiyar S, Venkat A, Yeung W, Eyers CE, Eyers PA, Kannan N. Redox Regulation of Brain Selective Kinases BRSK1/2: Implications for Dynamic Control of the Eukaryotic AMPK family through Cys-based mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.05.561145. [PMID: 38586025 PMCID: PMC10996518 DOI: 10.1101/2023.10.05.561145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
In eukaryotes, protein kinase signaling is regulated by a diverse array of post-translational modifications (PTMs), including phosphorylation of Ser/Thr residues and oxidation of cysteine (Cys) residues. While regulation by activation segment phosphorylation of Ser/Thr residues is well understood, relatively little is known about how oxidation of cysteine residues modulate catalysis. In this study, we investigate redox regulation of the AMPK-related Brain-selective kinases (BRSK) 1 and 2, and detail how broad catalytic activity is directly regulated through reversible oxidation and reduction of evolutionarily conserved Cys residues within the catalytic domain. We show that redox-dependent control of BRSKs is a dynamic and multilayered process involving oxidative modifications of several Cys residues, including the formation of intramolecular disulfide bonds involving a pair of Cys residues near the catalytic HRD motif and a highly conserved T-Loop Cys with a BRSK-specific Cys within an unusual CPE motif at the end of the activation segment. Consistently, mutation of the CPE-Cys increases catalytic activity in vitro and drives phosphorylation of the BRSK substrate Tau in cells. Molecular modeling and molecular dynamics simulations indicate that oxidation of the CPE-Cys destabilizes a conserved salt bridge network critical for allosteric activation. The occurrence of spatially proximal Cys amino acids in diverse Ser/Thr protein kinase families suggests that disulfide mediated control of catalytic activity may be a prevalent mechanism for regulation within the broader AMPK family.
Collapse
Affiliation(s)
- George N. Bendzunas
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Dominic P Byrne
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Safal Shrestha
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Leonard A Daly
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Sally O. Oswald
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Samiksha Katiyar
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Aarya Venkat
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Wayland Yeung
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Claire E Eyers
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
- Centre for Proteome Research, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Patrick A Eyers
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA
| | - Natarajan Kannan
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|